ion transport mechanisms during hyposmotic regulatory and

202
Wright State University Wright State University CORE Scholar CORE Scholar Browse all Theses and Dissertations Theses and Dissertations 2009 Ion Transport Mechanisms during Hyposmotic Regulatory and Ion Transport Mechanisms during Hyposmotic Regulatory and Isosmotic Apoptotic Volume Decreases in a Human Lens Isosmotic Apoptotic Volume Decreases in a Human Lens Epithelial Cells Line Epithelial Cells Line Ameet Ajit Chimote Wright State University Follow this and additional works at: https://corescholar.libraries.wright.edu/etd_all Part of the Biomedical Engineering and Bioengineering Commons Repository Citation Repository Citation Chimote, Ameet Ajit, "Ion Transport Mechanisms during Hyposmotic Regulatory and Isosmotic Apoptotic Volume Decreases in a Human Lens Epithelial Cells Line" (2009). Browse all Theses and Dissertations. 303. https://corescholar.libraries.wright.edu/etd_all/303 This Dissertation is brought to you for free and open access by the Theses and Dissertations at CORE Scholar. It has been accepted for inclusion in Browse all Theses and Dissertations by an authorized administrator of CORE Scholar. For more information, please contact [email protected].

Upload: others

Post on 30-Apr-2022

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Ion Transport Mechanisms during Hyposmotic Regulatory and

Wright State University Wright State University

CORE Scholar CORE Scholar

Browse all Theses and Dissertations Theses and Dissertations

2009

Ion Transport Mechanisms during Hyposmotic Regulatory and Ion Transport Mechanisms during Hyposmotic Regulatory and

Isosmotic Apoptotic Volume Decreases in a Human Lens Isosmotic Apoptotic Volume Decreases in a Human Lens

Epithelial Cells Line Epithelial Cells Line

Ameet Ajit Chimote Wright State University

Follow this and additional works at: https://corescholar.libraries.wright.edu/etd_all

Part of the Biomedical Engineering and Bioengineering Commons

Repository Citation Repository Citation Chimote, Ameet Ajit, "Ion Transport Mechanisms during Hyposmotic Regulatory and Isosmotic Apoptotic Volume Decreases in a Human Lens Epithelial Cells Line" (2009). Browse all Theses and Dissertations. 303. https://corescholar.libraries.wright.edu/etd_all/303

This Dissertation is brought to you for free and open access by the Theses and Dissertations at CORE Scholar. It has been accepted for inclusion in Browse all Theses and Dissertations by an authorized administrator of CORE Scholar. For more information, please contact [email protected].

Page 2: Ion Transport Mechanisms during Hyposmotic Regulatory and

ION TRANSPORT MECHANISMS DURING HYPOSMOTIC REGULATORY AND

ISOSMOTIC APOPTOTIC VOLUME DECREASES IN A HUMAN LENS

EPITHELIAL CELL LINE.

A dissertation submitted in partial fulfillment

of the requirements for the degree of

Doctor of Philosophy

By

AMEET AJIT CHIMOTE

M.B.B.S. Nagpur University (India), 1999

M.S. Wright State University, 2003.

____________________________ 2009

Wright State University

Page 3: Ion Transport Mechanisms during Hyposmotic Regulatory and

WRIGHT STATE UNIVERSITY

SCHOOL OF GRADUATE STUDIES

June 30, 2009.

I HEREBY RECOMMEND THAT THE DISSERTATION PREPARED UNDER MY SUPERVISION BY Ameet Ajit Chimote ENTITLED “Ion transport mechanisms during hyposmotic regulatory and isosmotic apoptotic volume decreases in a human lens epithelial cells line” BE ACCEPTED IN PARTIAL FULFILLMENT OF THE DEGREE OF Doctor of Philosophy ____________________________________

Peter K. Lauf, M.D. Dissertation Director ____________________________________ Gerald Alter, Ph.D. Director, Biomedical Sciences,Ph.D. Program

____________________________________ Joseph F. Thomas, Jr., Ph.D.

Dean, School of Graduate Studies Committee on Final Examination _______________________________ Peter K. Lauf, M.D. _______________________________ Norma C. Adragna, Ph.D.

_______________________________ Robert W. Putnam, Ph.D. _______________________________ Daniel T. Organisciak, Ph.D. _______________________________ Thomas L. Brown, Ph.D. _______________________________ Panagiotis A. Tsonis, Ph.D.

Page 4: Ion Transport Mechanisms during Hyposmotic Regulatory and

iii

ABSTRACT: Chimote, Ameet Ajit, PhD., Biomedical Sciences Ph.D. Program, Cell Biophysics Group, Wright State University, 2009. ION TRANSPORT MECHANISMS DURING HYPOSMOTIC REGULATORY AND ISOSMOTIC APOPTOTIC VOLUME DECREASES IN A HUMAN LENS EPITHELIAL CELL LINE.

Living cells maintain constant volume in response to physiological

stresses by altering trans-membrane ion, solute and water flow. In the present

study, early and late membrane transport changes in human lens epithelial (HLE-

B3) cells under hyposmotic and apoptotic stress were compared. Cell potassium

(Ki), rubidium (Rbi)-uptake and water content were measured by atomic

absorption spectrophotometry and gravimetry, respectively. Intracellular chloride

concentration [Cl]i was determined with the fluorescence dye N-

(ethoxycarbonylmethyl)-6-methoxyquinolinium bromide (MQAE), during

regulatory volume decrease (RVD) after hyposmotic stress and apoptotic volume

decrease (AVD) induced by staurosporine (STP), a protein-kinase inhibitor. Cell

water increased in hyposmotic balanced salt solution (BSS) as compared to

isosmotic BSS by > 2-fold at 5 min and decreased within 30 min to baseline

values, accompanied by K efflux commensurate with cell swelling and RVD.

Clotrimazole (CTZ) reduced Ki and water loss in hyposmotic BSS, and Rbi

uptake, suggesting a role for intermediate conductance K (IK) channels during

RVD. This was verified by reverse transcriptase polymerase chain reaction (RT-

PCR), Western blot and immunochemistry of HLE-B3 cells.

Apoptosis measured 2 h after 2 µM STP treatment was significant and

accompanied by 20% and 41% water loss after 30 min and 2 h, respectively.

Page 5: Ion Transport Mechanisms during Hyposmotic Regulatory and

iv

During apoptosis, Ki fell at a rate of 0.007/min until 30 min and decreased to 42%

by 2 h. [Cl]i decreased by 25% and 19% below baseline values at 30 min and 60

min, respectively. High extracellular K, and 2 mM 4-aminopyridine (4-AP)

significantly reduced apoptosis whereas the presence of CTZ did not. Annexin

labeling appeared at 15 min suggesting lipid changes preceded the water and K

loss initiating AVD. In contrast, loss of K, water, and most likely Cl differed by an

order of magnitude between RVD and AVD at early time points. Quantitative PCR

revealed decreased expression of IK channels during prolonged hyposmotic and

apoptotic stress. These studies suggest that in HLE-B3 cells, IK channels

partake in RVD, but cannot yet be discounted in AVD, which in part commences

through 4-AP-sensitive voltage-gated K channels.

Page 6: Ion Transport Mechanisms during Hyposmotic Regulatory and

v

TABLE OF CONTENTS

I. Background and literature review 1 1.1. Structure and function of the human lens 1 1.2. Ion transport in the lens 2 1.3. Volume regulation in cells 5 1.4. Ion channels involved during RVD in lens and other cells 10 1.5. Apoptosis in mammalian cells 11 1.6. Membrane changes in apoptosis 14 1.7 Abnormal lens pathology : Cataract 14 1.8. Apoptosis in the lens 15 1.9. Apoptotic volume decrease 17 1.10. RVI response during AVD 22 1.11. Role of ion channels during apoptosis 23 1.12. Ca-activated K channels 27 II. Rationale and Hypothesis 29

2.1.Aim 1 29 2.2. Aim 2 29 2.3. Aim 3 30 2.4. Significance 30 III. Materials and Methods 32 3.1.Reagents 32 3.1.1. Chemicals 32 3.1.2. Inhibitors 32 3.1.3. Molecular and immunological tools 33 3.2. Solutions and media 34 3.3. Cell culture 34 3.4. Measurement of ion fluxes 35 3.5. Measurement of cell water 36

Page 7: Ion Transport Mechanisms during Hyposmotic Regulatory and

vi

3.6. Measurement of intracellular chloride concentration [Cl]i 36

3.6.1. Treatment of HLE-B3 cells with MQAE 36 3.6.2. Measurement of MQAE fluorescence 37 3.6.3. Calibration of MQAE fluorescence 37 3.6.4. Calculation of [Cl]i 37 3.7. DNA fragmentation assay 40 3.8. Cell death detection ELISA 40 3.9. Reverse transcriptase polymerase chain reaction (RT-PCR) 41 3.10. siRNA transfection 43 3.11. RT-qPCR for hyposmotic and apoptotic stress 43 3.12. Western Blot 44 3.13. Immunofluorescence staining and fluorescence microscopy 45 3.14. Annexin-V-FLUOS staining 46 3.15. Statistical Analysis 47 IV. Results 49 4.1. Characterization of HLE-B3 cells 49 4.2. Measurement of intracellular potassium concentration [K]i 49 4.3. Measurement of intracellular chloride concentration [Cl]i 55 4.3.1. Characterization of MQAE fluorescence 55 4.3.2. Measurement of [Cl]i 63 4.4. RVD response in HLE-B3 cells 70 4.4.1. Effect of hyposmotic stress on cell water content 70 4.4.2. Effect of RVD on cellular K and [Cl]i 70 4.4.3. Effect of RVD on cell survival 71 4.5. AVD response in HLE-B3 cells 80 4.5.1. Time course of cell death 80 4.5.2. Effect of apoptotic stress on cell volume and cell water 88 4.5.3. Effect of AVD on cellular K and [Cl]i 88 4.6. Membrane transport changes accompanying RVD 98 4.7. Molecular and immunochemical studies 109

Page 8: Ion Transport Mechanisms during Hyposmotic Regulatory and

vii

4.7.1. KCC isoforms in HLE-B3 cells 109 4.7.2. Ca-dependent K channel expression in HLE-B3 cells 109 4.8. Effect of IK channel gene knockdown on Rb uptake in RVD 110 4.9. Membrane transport changes accompanying AVD 119 4.10. Effect of prolonged stress on IK channel 138 4.11. Early and late membrane changes during AVD 144 4.11.1. Annexin binding during AVD 144 4.11.2. Annexin binding during AVD with caspase inhibitor 144 V. Discussion 152 5.1. HLE-B3 cells as an in-vitro model system 152 5.2. Measurement of Ki and water 152 5.3. Measurement of [Cl]i 153 5.4. Regression analysis vs. Stern-Volmer analysis for [Cl]i 154 5.5. Events during RVD 158 5.6. Cellular changes during AVD 157 5.7. Ion channels involved during RVD 160 5.8. RNAi studies 162 5.9. Ion channels involved during AVD 162 5.10. Role of 4-AP sensitive channel in RVD and AVD 164 5.11. Effect of prolonged stress on IK channel gene expression 165 5.12. Membrane changes during AVD 166 5.13. Caspase independence of AVD 168 5.14. Apoptosis vs. necrosis in STP-treated cells 168 VI. Summary 171 VII. References 174 VIII. Appendix : List of Abbreviations 187

Page 9: Ion Transport Mechanisms during Hyposmotic Regulatory and

viii

LIST OF FIGURES

Figure Page 1. Anatomy of the human lens 4 2. Volume regulation in a normal cell to osmotic stress 7 3. Transporters and channels involved in RVD and RVI 9 4. Overview of extrinsic and intrinsic apoptotic pathways 13 5. Apoptotic volume decrease 20 6. Immunofluorescence labeling of HLE-B3 cells 52 7. MQAE fluorescence as a function of chloride concentration 57 8. MQAE fluorescence as a function of dose and time of incubation 59 9. MQAE fluorescence as a function of tributyltin and nigericin 62 10. Measurement of [Cl]i by regression analysis 66 11. Stern-Volmer plot for MQAE fluorescence 68 12. Effect of hyposmotic stress on cell water 73 13. Ki as a function of time during hyposmotic stress 75 14. Effect of Hyposmotic stress on MQAE fluorescence 77 15. Cell death detection ELISA after incubation with hyposmotic media 79 16. DNA fragmentation assay after STP treatment 83 17. Cell Death Detection ELISA after STP treatment 85 18. Effect of STP treatment on total protein content 87 19. Immunofluorescent labeling of HLE-B3 cells after STP incubation 91 20.Effects of STP treatment on cell water 93 21. Effects of STP treatment on Ki 95 22. Effect of STP treatment on MQAE fluorescence 97 23.Effect of IK channel inhibitor on Rb uptake during RVD 101

Page 10: Ion Transport Mechanisms during Hyposmotic Regulatory and

ix

LIST OF FIGURES

Figure Page 24.Effect of Ca-dependent K channel inhibitors on Rb influx during RVD 103 25.Effect of IK channel inhibition by CTZ on cell water during RVD 106 26.Effect of IK channel inhibition on MQAE fluorescence during RVD 108 27.Immunofluorescence of KCC isoforms in HLE-B3 cells 113 28.Molecular evidence of Ca dependent K channels in HLE-B3 cells 115 29.Effect IK channel gene knock down by RNAi on Rb influx 117 30.Cell death detection ELISA after STP treatment in the high [K]o 122 31.Effect of CTZ on Rb uptake as a function of time during AVD 124 32.Effect of CTZ on Ki as a function of time during AVD 126 33.Effect of CTZ on cell water during AVD 128 34.Effect on CTZ on MQAE fluorescence during AVD 130 35.Effect of CTZ on STP induced cell death 132 36.Effect of K channel inhibitors on Rb influx during RVD 135 37.Cell Death Detection ELISA after STP treatment in the presence of 4-AP 137 38.IK channel gene expression on prolonged hyposmotic stress 141 39.IK channel gene expression as a function of STP incubation time 143 40.Late effects on Annexin V binding after incubation with 2 μM STP 147 41.Early effects on Annexin V binding after incubation with 2 μM STP 149 42.Effect of caspase inhibitor on annexin binding during AVD 151 43.Scheme of events during AVD 173

Page 11: Ion Transport Mechanisms during Hyposmotic Regulatory and

x

LIST OF TABLES

Table Page 1. Potassium channels involved in apoptosis 25 2. Chloride channels involved in apoptosis 26 3. Physiological role of IK channels 28 4. Composition of equilibration solutions for [Cl]i measurements 39 5. Primer Sequences for RT-PCR 42 6. Antibody concentrations for immunofluorescence experiments 48 7. Measured Ki in HLE-B3 cells 53 8. Measured cell water content in HLE-B3 cells 54 9. Stern -Volmer Constant (KCl) and [Cl]i in HLE-B3 cells 69 10. Effect of Ca dependent K channel inhibitors on Rb influx during RVD 104 11.Summary of siRNA experiments 118

Page 12: Ion Transport Mechanisms during Hyposmotic Regulatory and

1

I. Background and Literature Review

1.1. Structure and function of the human lens:

The lens, a transparent biconvex structure surrounded by an elastic

capsule that is made of Type IV collagen, is situated behind the iris and the pupil.

Aqueous humor is present anteriorly whereas the vitreous body is posterior to the

lens. The lens is responsible for refraction and accommodation during normal

vision. Lens epithelial cells (LEC), present only on the anterior surface, are

cuboidal and form a monolayer underneath the lens capsule (Figure 1). At the

equator, the LECs differentiate into lens fiber cells (LFC) constituting the main

mass of the lens. LFCs do not have nuclei and the earliest formed fibers are

situated in the nucleus (Figure 1) whereas newly formed LFCs are present in the

cortex, proximal to the LEC (Bhat 2001;Andley 2007; 2009). The lens is devoid of

blood supply and therefore derives its nutrition by solute transfer from the

surrounding aqueous humor. The human lens grows throughout life, yet lens cell

division occurs on a very limited scale in a narrow band of epithelial cells, and the

rate of growth decreases with age. LECs are the main progenitor cells for the

development of new LFCs. Because of itʼs location along the optic axis of the

eye, the human lens, particularly the LECs are chronically exposed to

environmental stressors, especially solar and ultraviolet UVA (315-400 m), UVB

Page 13: Ion Transport Mechanisms during Hyposmotic Regulatory and

2

(280-315 nm) and UVC (100-280nm) radiations (Andley 2009; Hightower et al.,

1994).

1.2. Ion transport in the lens

Though the lens is immersed in a fluid environment, its own water content

is only 60%. The lens loses its transparency at a water content > 60% (Mathias

et al. 2004). The normal lens has a high K and low Na content, and maintains

water and ions with the Na/K pump (Cui et al., 2002), ion channels like K, Na and

Cl channels and cotransporters for Na,K and 2Cl (NKCC) and K,Cl (KCC) (Lauf

et al., 2006; Misri et al., 2006). These are present mainly in the LECs (Mathias et

al., 2004; Candia et al., 2004; Bhat et al., 2001). Several models have described

fluid and ion transport through the lens and have been reviewed in Mathias et al.

(2007). One model of homeostasis states that water enters the lens from the

anterior surface through water channels and leaves the lens from the posterior

surface (Fischbarg et al., 2003). Another model proposes that Na and water enter

from both the anterior and posterior faces of the lens with Na being pumped out

by the Na/K pump, densely packed at the lens equator (Mathias et al., 2004).

Thus, ion transport is important in LECs for volume regulation and maintenance

of a viable epithelial monolayer.

Page 14: Ion Transport Mechanisms during Hyposmotic Regulatory and

3

Figure 1: Anatomy of the human lens

Schematic drawing of a lens sectioned along the optic axis to illustrate the

location of lens epithelial cells. Epithelial cell proliferation occurs in the equatorial

region of the lens, called the germinative zone. Cells in the transitional zone

differentiate to secondary lens fiber cells by loss of nuclei and cell organelles.

Throughout life, layers of newly formed fiber cells cover the older cells. As a

result, older secondary fiber cells are located closer to the center of the lens and

form the nucleus of the lens. The primary fiber cells are formed during

embryogenesis. Zonular fibers support the lens from the ciliary body (not shown).

Adapted from Andley, 2008.

Page 15: Ion Transport Mechanisms during Hyposmotic Regulatory and

4

Figure 1:

Page 16: Ion Transport Mechanisms during Hyposmotic Regulatory and

5

1.3. Volume regulation in cells

Most living cells maintain constant volume to ensure their proper

functioning under physiological conditions. However, physiological processes like

cell division, differentiation, maturation and apoptosis, all involve significant

changes in cell volume, which involve ion transporters, ion channels and water

channels. In response to hyper- or hypo-osmotic environments, cell volume is

also regulated by various specifically designed pathways for ion and water

movements localized in the plasma membrane. Two principal mechanisms are

outlined in Figure 2. Regulatory volume decrease (RVD) is a compensatory

response to hypotonic or hyposmolar swelling in which water enters the cell

causing a volume increase. This triggers a net K , Cl, and water efflux due to the

K-Cl cotransporter and K channels, Cl channels, K/H and Cl/HCO3 exchangers.

The result is cells lose water and return to their original size. In addition, by virtue

of its 3Na/2K exchange, the Na/K pump is poised to aid the cell in solute and

hence water loss, i.e. dehydration. Regulatory volume increase (RVI) is a

response to hypertonic or hyperosmolar conditions to offset osmotically driven

cell water loss and volume reduction, triggering activation of NKCC, and the

Na/H-and Cl/HCO3-exchangers, which cause an influx of Na and Cl, and water

and hence volume recovery. The transporters and channels involved during RVI

and RVD are summarized schematically in Figure 3.

Page 17: Ion Transport Mechanisms during Hyposmotic Regulatory and

6

Figure 2: Volume regulation in a normal cell exposed to osmotic stress

Schematic representation of volume regulatory pathways in mammalian cells.

When exposed to hyposmotic stress, cells swell due to water influx and as a

response to swelling, undergo regulatory volume decrease (RVD) by efflux of K,

Cl and H2O, restoring normal cell volume. On exposure to hyperosmotic solution,

cells undergo shrinkage and as a response, regulatory volume increase (RVI) by

influx of Na, K , Cl and H2O, restoring cell volume back to baseline.

Page 18: Ion Transport Mechanisms during Hyposmotic Regulatory and

7

Figure 2:

Page 19: Ion Transport Mechanisms during Hyposmotic Regulatory and

8

Figure 3: Transporters and channels involved in RVD and RVI in a typical

mammalian cell.

The effectors of RVD are K-Cl cotransporters (KCC), various voltage gated K,

including Ca-dependent K channels, and Cl channels, such as volume-sensitive

outwardly rectifying chloride channels (VSOR). RVI is brought about by the Na-K-

2Cl cotransporter (NKCC), and by the Na/H (NHE) and Cl/HCO3 exchangers.

Page 20: Ion Transport Mechanisms during Hyposmotic Regulatory and

9

Figure 3:

Page 21: Ion Transport Mechanisms during Hyposmotic Regulatory and

10

1.4. Ion channels involved during RVD in lens and other epithelial cells.

Cells maintain constant volume in response to a variety of physiological

and pathological stresses by changing trans-membrane ion, solute and water

flow (Hoffmann et al., 2009). This paradigm also applies to lens epithelial cells,

which critically depend on volume regulatory mechanisms such as ion channels

(Rhodes et al., 2003), ion exchangers (Candia O, 2004; Mathias et al., 2007;

Mathias and Rae, 2004) and cotransporters (Lauf et al., 2006) to safeguard their

transition into lens fiber cells while maintaining transparency (Beebe et al., 1990).

Recent studies suggest that during hyposmotically-induced RVD of primary

human lens epithelial (FHL124) cells (Lauf et al., 2008), Ca-activated

intermediate conductance K channels (IK, KCa3.1) operate in concert with

volume-sensitive outwardly rectifying anion channels (VSOR), also called

volume-regulated anion channels (VRAC) (Okada and Maeno, 2001; Okada et

al., 2001), to permit isotonic solution loss. RVD may also be initiated by other Ca-

activated K channels present in these cells such as the big and small

conductance K channels (BK and SK, respectively), and K-Cl cotransporters

(Lauf et al., 2006; Misri et al., 2006). BK and IK channels mediate RVD in

osteoblasts (Weskamp et al., 2000) and volume-sensitive IKvol channels in

Ehrlich ascites tumor cells (Hoffmann et al., 2007, Hoffmann et al., 2009). IK

channels also play a role in RVD of T-cells, human tracheal epithelial cells, and

mouse erythroid cells (Wang et al., 2003; Wehner, 2006).

Page 22: Ion Transport Mechanisms during Hyposmotic Regulatory and

11

1.5. Apoptosis in mammalian cells.

During apoptosis or programmed cell death, a cell is removed in response

to an apoptotic signal. Apoptosis is involved in a variety of physiological and

pathological processes and is necessary for development and embryogenesis.

Loss of apoptosis is a manifestation of uncontrolled cell division, a cancerous

condition. Excessive apoptosis is seen in several diseases like autoimmune

diseases, like in acquired immune deficiency syndrome (AIDS), and

neurodegenerative disorders (Bortner et al 1998). Apoptosis is characterized by

cell shrinkage, nuclear condensation and internucleosomal DNA fragmentation

and is mediated by intrinsic and extrinsic pathways, which are summarized in

Figure 4.

The intrinsic or the mitochondrial pathway is initiated within the cell by

DNA damage, oxidative stress, mitochondrial damage, etc. and involves the

release of cytochrome c from the mitochondria. Also shown in Figure 4 is the

putative mechanism of action of staurosporine (STP), a general kinase inhibitor,

which has been used to induce apoptosis in this study. The effector caspase 9

activates caspase 3, which in turn activates endonucleases responsible for DNA

fragmentation and cell death. The extrinsic or death receptor pathway is initiated

by signaling from outside the cell, usually mediated by receptors such as TNF-α,

tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and the

Fas receptor. Here, caspase 8 becomes activated first, which in turn activates

caspases 3, 6 and 7.

Page 23: Ion Transport Mechanisms during Hyposmotic Regulatory and

12

Figure 4: Overview of extrinsic (receptor mediated) and intrinsic

(mitochondrial) apoptotic pathways.

Scheme illustrating the extrinsic (receptor-mediated) and intrinsic (mitochondrial)

apoptotic pathways in response to stress and highlighting the downstream

effectors of apoptosis leading to caspase activation and DNA fragmentation.

Also shown are the modulators of apoptosis: Pro-apoptotic proteins such as

apoptotic peptidase activating factor 1 (Apaf-1), responsible for activation of

caspase 9, BH3 interacting domain (BID) and anti-apoptotic protein Bcl-2.

Staurosporine, a kinase inhibitor used in the present study, is thought to induce

apoptosis through the intrinsic mitochondrial pathway. Adapted from: Hengartner

M.O, Nature. 407:770 (2000).

Page 24: Ion Transport Mechanisms during Hyposmotic Regulatory and

13

Figure 4:

Page 25: Ion Transport Mechanisms during Hyposmotic Regulatory and

14

1.6. Membrane changes in apoptosis

Apoptosis is characterized by cell shrinkage, translocation of

phosphatidylserine (PS) to the cell surface, caspase activation and ultimately

DNA fragmentation and cell breakdown (Elliott and Higgins, 2002). The exposure

of PS on the outer leaflet of the plasma membrane is a known crucial marker for

cells undergoing apoptosis to facilitate recognition, engulfment and degradation

by phagocytes (Savil and Fadok, 2000). In normal cells, PS is present on the

inner leaflet of the cell membrane. During cell injury or cell death, the PS

translocation to the outer leaflet leading to a loss of membrane symmetry, is

mediated by phospholipid scramblases in the presence of cytosolic Ca (Lang et

al., 2004, Sims P and Wiedmer T, 2000). It is known that externalization of PS

during apoptosis precedes caspase activation and DNA fragmentation by several

hours during the apoptotic cascade and can be detected by annexin binding in

the presence of cytosolic Ca (Martin et al., 1995). Annexin V is a Ca-dependent

phospholipid-binding protein which has a high affinity for PS. Hence, this protein

is used as a probe for PS exposure on the outer cell leaflet during the early

stages of apoptosis (Vermes et al., 1995).

1.7. Abnormal lens pathology: Cataract

Cataract, a condition resulting from clouding of the crystalline lens of the

eye, is the leading cause of blindness worldwide with a staggering 20 million

cases globally (World Health Organization bulletin 2001). In the absence of a

strategy for preventing cataract, the only treatment available is surgical removal

Page 26: Ion Transport Mechanisms during Hyposmotic Regulatory and

15

of the cataractous lens. About 1.6 million patients are operated for cataract

annually in the United States alone. About 85% of cataracts are age-related and

the incidence of cataract increases with age. In the next 15 years, it is predicted

that the population above the age of 60 years old will double, subsequently

doubling the incidence of cataract.

1.8. Apoptosis in the lens

1.8.1. Stressors

Chemical or environmental stressors induce apoptosis. Ultraviolet (UV)

rays from solar radiations are the most important environmental stressors

targeting LECs, the primary targets for UVB radiation (Hightower et al., 1994). In

several experimental models, UV light is used to trigger apoptosis in tissues like

skin, bone marrow, and the cornea. UV-mediated apoptosis occurs through the

p53 and c-Jun N-terminal kinase (JNK) pathways (Wang et al., 2005). UVC

irradiation initiates complex cellular events at the membrane level in corneal

epithelial cells which include activation of G-proteins, membrane receptors such

as platelet derived growth factors (PDGF), transcription factors and tyrosine

kinases (Wang et al., 2007). The important signaling pathways involved in UVC-

induced apoptosis are the JNK pathway (Wang et al., 2007) and p38 (Bodero et

al., 2003). Studies on corneal epithelial cells reveal that the UVC-induced K

channel activation initiates the apoptotic cascade and is an earlier event than

DNA damage (Wang et al 2005). Hydrogen peroxide (H2O2) is another commonly

used stressor, inducing apoptosis in cells by the production of reactive oxygen

Page 27: Ion Transport Mechanisms during Hyposmotic Regulatory and

16

species (ROS), i.e. via oxidative stress. H2O2 depletes cellular glutathione (GSH),

inducing oxidative stress. This causes structural changes in proteins (protein-S-

S-glutathione formation) and leads to cell death (Long et al., 2005; Lou 2003).

1.8.2. Staurosporine as inducer of apoptosis

A compound used to induce apoptosis in cell culture models is the

chemical stressor staurosporine (STP), a kinase inhibitor obtained from

bacterium Streptomyces staurosporeus (Maeno et al., 2000; Rüegg and Burgess,

1989). The structure of STP consists of a sugar molecule along with a

heterocyclic indole carbazole group. STP inhibits kinases by binding and

competing with ATP at its binding site thus preventing substrate phosphorylation

of proteins (Rüegg and Burgess, 1989; Karaman et al., 2008). In cardiac

myocytes, in the absence of ATP, STP induces necrotic cell death (Shiraishi et

al., 2001). In the presence of ATP, STP induces apoptosis by the intrinsic

(mitochondrial) pathway by activation of caspase 3 (Yue et al., 1998; Roucou et

al., 2001; Shiraishi et al., 2001). STP has been used to induce apoptosis in

several studies on cell death in various ocular cell culture models such as HLE-

B3 cells (Andley et al., 1998), bovine lens epithelial cells (Andersson et al.,

2000), cultured rabbit lenses (Langford et al., 2006) and in human corneal

epithelial cells (HCE) (Thuret et al., 2003).

1.8.3. Apoptosis in lens epithelial cells

Several studies have provided evidence of apoptosis in the human lens.

UVB irradiation, as well as oxidative stress induced by H2O2, lead to apoptosis in

Page 28: Ion Transport Mechanisms during Hyposmotic Regulatory and

17

the SV-40 immortalized HLE–B3 human lens epithelial cell line (Long et al.,

2004). In mouse lens epithelial cell models, tumor necrosis factor alpha (TNF-α)

also induces apoptosis (Awasthi et al., 2004). Analysis of cataractous lenses

procured from post-surgical patients demonstrated evidence of apoptosis as

detected by terminal deoxynucleotidyl transferase (TdT) labeling and DNA

fragmentation assays (Li et al., 1995). In lens epithelial cell cultures of UVB-

irradiated rats, rapid apoptosis was observed and proposed to contribute to

cataract formation (Li et al., 1996). In murine models, apoptosis was found to be

an important feature of TGF-β- induced cataract (Maruno et al., 2002).

1.9. Apoptotic volume decrease

Cell shrinkage in isosmotic solutions is one of the earliest morphological

events during programmed cell death and, has been coined apoptotic volume

decrease (AVD). It facilitates macrophages to engulf the dying cells (Bortner et

al., 2004). Although AVD is a ubiquitous and distinctive feature common to all

cells (Bortner et al., 1998), the time frame for AVD to occur depends on the cell

type and varies with the apoptotic stimulus (Heimlich et al., 2004). In cells treated

with a pro-apoptotic agent, AVD induction precedes cytochrome c release from

the mitochondria, caspase-3 activation and DNA laddering (Maeno et al., 2000;

Bortner CD and Cidlowski JA, 2007). AVD is mainly a result of the loss of

intracellular ions (Figure 5). Several studies have shown that during AVD,

changes in ion fluxes alter cellular ions, thus causing an extensive loss of

intracellular water responsible for the volume decrease (Heimlich et al., 2004;

Page 29: Ion Transport Mechanisms during Hyposmotic Regulatory and

18

Nobel et al., 2000; Bortner et al., 2000; Bortner et al., 2004). AVD in response to

a pro-apoptotic stimulus, causes efflux of K (Bortner et al., 2002), Cl (Okada et

al., 2004), with electroneutrality preserved due to anion loss (Bortner et al.,

2004). There is an activation of ion channels that promote K and Cl efflux from

the cells (Okada 2004; Bortner et al., 2000) accompanied by an inhibition of the

Na/K pump (Nobel et al., 2000) resulting in a decreased K influx into the cell and

increased intracellular Na levels. STP-induced apoptosis was characterized by

an increase in intracellular Na, a decrease in intracellular Cl and K

concentrations, and a decrease in the K/Na ratio due to inhibition of the Na/K

pump ( Arrebola et al., 2005). During AVD, as a result of the apoptotic stimulus

and the subsequent ion efflux, aquaporin expression is upregulated affecting

water transport and enhancing volume decrease (Jablonski et al., 2004). It is

believed that this ion and water efflux during AVD eventually results in apoptosis

by activation of caspases and endonucleases (Bortner et al., 2004). However,

there have been published reports that AVD can occur independently of caspase

activation in U937 cells (Maeno et al., 2000) and in cerebrocortical neurons

treated with nitric oxide (NO) (Bossy-Wetzel et al., 2004). Ion and electrolyte

extrusion during AVD and RVD may involve the same ion channels and

transporters (Bortner 2003), however, the activation and signaling mechanism for

the two are different (Heimlich et al., 2004).

Page 30: Ion Transport Mechanisms during Hyposmotic Regulatory and

19

Figure 5: Apoptotic volume decrease

AVD occurs under isosmotic conditions when cells are exposed to a pro-

apoptotic stimulus (see text for details). AVD is brought about by efflux of K, Cl

and H2O, causing a reduction in cell volume, triggering caspase activation and

cell death by DNA fragmentation.

Page 31: Ion Transport Mechanisms during Hyposmotic Regulatory and

20

Figure 5:

Page 32: Ion Transport Mechanisms during Hyposmotic Regulatory and

21

1.9.1 Potassium ions in apoptosis

With the exception of red blood cells of ruminants (Lauf et al., 2001) and

dogs (Fujise et al., 2001), most mammalian cells have a high concentration of

intracellular potassium [K]i as compared to a low extracellular concentration of

potassium [K]o: this gives rise to a concentration gradient that facilitates loss of

Ki. Almost all cells have an active Na/K pump (Na/K -ATPase), which pumps 3

sodium ions out of the cell for every 2 potassium ions into the cell, thereby

maintaining a concentration gradient of ions. Due to the constant pumping activity

of the Na/K pump, normal cells maintain a high intracellular K concentration [K]i

,which protects cells against apoptosis. High [K]i inhibits both intrinsic and

extrinsic apoptotic pathways (Thompson et al., 2001). In addition, Jurkat, cells

cultured in high [K]o media do not exhibit an apoptotic cascade and volume

decrease (Hughes et al., 1997). In Jurkat cells, when [K]i, is decreased during

RVD, there is an 85% potentiation of apoptosis, i.e., the cells die while losing K

instead of regaining their original volume by eliciting a RVI response (Bortner et

al., 1996). Loss of Ki promotes the conversion of inactive procaspases to active

caspases, which in turn leads to apoptosis (Hughes et al., 1997). In scenarios,

where the compensatory RVI response is inhibited or overridden, high [K]o

inhibits apoptosis (Heimlich et al., 2004). When, by some unknown mechanism,

shrunken cells cannot revert back to their original size by compensatory RVI, the

apoptotic pathway is activated with subsequent “cell death” (Heimlich et al.,

2004).

Page 33: Ion Transport Mechanisms during Hyposmotic Regulatory and

22

1.9.2. Chloride ions in apoptosis

During cell volume decrease by RVD or AVD, the loss of Ki is

accompanied by a concurrent loss of Cli to maintain the electroneutrality of the

cell (Bortner CD and Cidlowski JA, 2007). Okada (2004) reported that the loss of

Ki during AVD is accompanied by an anionic current due to Cl efflux by Cl

channels. Loss of Cli is important for the apoptotic pathways for it activates

caspases, affects membrane potential and caspase-activated DNAse (CAD)

activation (Okada 2004). Inhibition of Cl efflux by chloride channel blockers

prevents or diminishes apoptosis in various cell model systems, highlighting the

fact that Cl efflux is also involved in apoptosis (Heimlich G and Cidlowski JA,

2006; Porcelli et al., 2004; Okada Y and Maeno E, 2001).

1.10. RVI and AVD response

While undergoing AVD, cells fail to counteract volume loss by regulatory

volume increase (RVI) (Hoffmann et al., 2009; Bortner CD and Cidlowski JA,

2007; Maeno et al., 2006) which otherwise commences during hyperosmotic cell

shrinkage (Heimlich et al., 2004). Upon prolonged incubation in hypertonic

medium, T-cells and U937 cells lacking a normal RVI response undergo

apoptosis, while COS-7 cells, HeLa and L-cells exhibiting a functional RVI

response fail to demonstrate cell death (Shimizu et al., 2007; Bortner and

Cidlowski, 2007). Studies on the inhibition of hypertonicity-induced cation

channels in HeLa cells reported a dose-dependent sensitization of the cells to

Page 34: Ion Transport Mechanisms during Hyposmotic Regulatory and

23

apoptosis due to an impaired RVI response (Shimizu et al., 2006). During a RVI

response due to hypertonic stress in HeLa cells, hypertonicity induced cation

channels (HICC) are activated by mediation of intracellular kinases (Numata et

al., 2008). In the same HeLa cells, when kinases were inactivated by treatment

with STP, the activity of HICC was significantly reduced, suggesting that RVI was

not induced during AVD. On the other hand, when the cells were incubated in

hypertonic (600 mOsm) medium, the incidence of AVD and apoptosis was

significantly reduced on treatment with STP. These results demonstrate that

HICC are activated during AVD and rescue cells from STP- induced apoptosis

(Numata et al., 2008).

1.11. Role of ion channels during apoptosis

The exact mechanism of K loss during AVD is not fully understood and

has been ascribed to stimulation of K channels, causing a K efflux (Heimlich et

al., 2004). These K channels are cell type- and stimulus-specific (Bortner et al.,

2004). In UVC-treated leukemia cells, an inwardly rectifying K channel is involved

(Wang et al., 1997) as compared to outwardly rectifying K channels in TGF-β-

induced apoptosis in a rat liver cell line (Heimlich et al., 2004). The various K

channels involved in induction of apoptosis in various cell types are summarized

in Table 1. Several studies have confirmed a loss of Na/K pump activity during

apoptosis (Nobel et al., 2000), which decreases Ki and increases Nai (Heimlich et

al., 2004). On the other hand, it has also been shown in studies on vascular

smooth muscle cells that inversion of intracellular Na/K ratio attenuated the

Page 35: Ion Transport Mechanisms during Hyposmotic Regulatory and

24

apoptotic response (Orlov et al, 1999, Orlov et al., 2004). The loss of Cl from

Jurkat cells occurs through activation of an outwardly rectifying chloride current

(ORCC) as demonstrated by Fas ligation (extrinsic pathway) (Szabo et al., 1998).

The Cl channels that participate in apoptosis are summarized in Table 2.

Page 36: Ion Transport Mechanisms during Hyposmotic Regulatory and

25

Table 1: Potassium channels in apoptosis

Cell Model Type of K channel

Apoptotic inducer

Apoptotic inhibitor

Reference

Erythrocytes IK channels Ionomycin Charybdotoxin, clotrimazole, high

[K]o

Lang et al., 2003

Cardiomyocytes Voltage-gated K channels

Staurosporine 4-AP, TEA Ekhterae et al., 2003

COS-7 cells Voltage-gated K channels

Staurosporine 4-AP Brevnova et al., 2004

Corneal epithelial cells

Voltage-gated K channels

UV irradiation 4-AP Wang et al., 2003

U937 cells Volume sensitive K

channel

Staurosporine Ba Maeno et al., 2000

Myeloblastic leukemia cells

Voltage-gated K channels

UV irradiation 4-AP Wang et al., 1999

Pulmonary artery smooth muscle

cells

Voltage-gated K channels

Staurosporine 4-AP, High [K]o Krick et al., 2001

CD4+ lymphocytes

Ca activated K channel

Calcimycin Charybdotoxin, clotrimazole,

quinine

Elliott and Higgins, 2003

Cortical neurons Delayed rectifier K channels

Amyloid β-peptide

TEA Yu et al., 2006

Rat liver cells Inward rectifier K channels

TNF-α Barium, quinine Nietsch et al., 2000

For abbreviations, please see Appendix.

Page 37: Ion Transport Mechanisms during Hyposmotic Regulatory and

26

Table 2: Chloride channels in apoptosis

Cell Model Type of Cl-channel

Apoptotic inducer

Apoptotic inhibitor

Reference

Jurkat cells ORCC CD 95 Glibenclamide Szabo et al., 1998

Non-pigmented ciliary epithelium

ORCC Staurosporine NPPB Coca-Prados et al., 1995

Rat Liver cells Not defined TNF-α NPPB Nietsch et al., 2000

HeLa, PC12, U937 VSOR Staurosporine NPPB, DIDS, SITS

Maeno et al., 2000

Cultured cortical neurons

CLC-2 CLC-3

Staurosporine, serum deprivation

DIDS, SITS, NPPB

Wei et al., 2004

Mouse cortical neurons

VSOR NMDA NPPB Inoue and Okada, 2007

Rat Cortical neurons

Not defined Staurosporine SITS Small et al., 2002

Jurkat T-cells VSOR UV-C SITS Heimlich G and Cidlowski J,

2006

For abbreviations, please see Appendix.

Page 38: Ion Transport Mechanisms during Hyposmotic Regulatory and

27

1.12. Ca-activated K channels

Ca-activated K channels are ubiquitous in mammalian cells and display a

wide repertoire of functions in different cell types. On the basis of their

electrophysiological characteristics, they are divided into three subtypes: voltage-

dependent, large conductance K channels (BK), voltage-independent, small

conductance K channels (SK) and inwardly rectifying, intermediate conductance

K channels (IK channels) (Begenisich et al., 2004). IK channels are prominently

expressed in cells involved in salt and fluid transport, including the colon, salivary

glands, lung, liver and vascular endothelium (Begenisich et al., 2004; Wei et al.,

2005). IK channels have been shown to play an important role in RVD in T-cells,

human tracheal epithelial cells, mouse erythroid cells (Wang et al., 2003) and

primary human LEC (Lauf et al., 2008). In primary cultures of T-lymphocytes, IK

channel activity is required for cell shrinkage, phosphatidyl serine (PS)

translocation and cell death (Elliot et al. 2003). In cataractous lenses of patients

suffering from myotonic dystrophy, there is an increased expression of SK3

channels (Rhodes et al., 2006). The physiological role of IK channels in various

cell types is summarized in Table 3.

Page 39: Ion Transport Mechanisms during Hyposmotic Regulatory and

28

Table 3: Physiological role of IK channels

Cell Type Function Reference

Erythrocytes Volume regulation Apoptosis (Eryptosis)

Begenisich et al., 2004 Lang et al., 2003

Lymphocytes Activation, proliferation Grissmer et al., 1993

Vascular smooth muscle cells

Proliferation Köhler et al., 2003

Endothelium Vasodilatation Eichler et al., 2003

Mesenteric arteries Angiogenesis Grgic et al., 2005

CD4+ lymphocytes Apoptosis Elliott and Higgins, 2003

Intestinal epithelial cells RVD Wang et al., 2003

Human tracheal cell line RVD Vasquez et al., 2001

T lymphocytes Volume regulation Begenisich et al., 2004

FHL 124 human lens epithelial cells

RVD Lauf et al., 2008

Page 40: Ion Transport Mechanisms during Hyposmotic Regulatory and

29

II. Rationale and Hypothesis

The goal of this dissertation is to characterize, in an immortalized human

lens epithelial cell line (HLE-B3), some of the transport mechanisms responding

to hyposmotic stress with RVD and to a pro-apoptotic stress with AVD. To

accomplish this, the timelines of cell water, K and Cl loss, and induction of cell

death are compared. The nature of potential K channels involved by

pharmacological and molecular means is investigated. The RVD and AVD

responses are characterized by proposing the following hypothesis:

Stressor induced changes in membrane permeability are determinants of

regulatory and apoptotic volume decrease (RVD and AVD) in HLE-B3 cells.

The following specific aims will test this hypothesis:

2.1. Measurement of the baseline K content and Cl concentrations and cell

volume in HLE-B3 cells. Volume decrease accompanies efflux of K, Cl and

water from the cell. Currently, there are no published data on the baseline

intracellular K and Cl levels in HLE-B3 cells. Baseline values of K content are

measured by atomic absorption spectrophotometry while baseline Cl

concentration by measuring the fluorescence of the Cl sensitive dye, MQAE.

2.2. Demonstration of stress-induced RVD and AVD in HLE-B3 cells by time

course and dose response of the baseline parameters (see 2.1.). RVD and

Page 41: Ion Transport Mechanisms during Hyposmotic Regulatory and

30

AVD are characterized by loss of K, Cl and water from the cell. The timeline of

these changes will be measured and compared in HLE-B3 cells.

2.3. Define major volume regulatory pathways in RVD and AVD,

functionally and at the molecular level. The participation of IK channels during

RVD has been revealed in human intestinal epithelial cells (Wang et al., 2003)

and FHL 124 cells (Lauf et al., 2008). The presence of IK channels will be

confirmed in HLE-B3 cells by molecular biology and functional studies. The effect

of IK channel inhibition will be studied during the RVD response. As shown in

Tables 1 and 3, numerous ion channels are involved during AVD and RVD. The

involvement of K channels during AVD will be determined by functional studies in

the presence of inhibitors. The results described in sections 1.13.2 and 1.13.3

will offer an insight into the physiological changes occurring during RVD and AVD

responses in HLE-B3 cells.

2.4. Significance:

Recent studies demonstrated the molecular and functional presence of IK

channels in FHL 124 primary human lens epithelial cells during hyposmotic RVD

(Lauf et al., 2008). However, it is not known if they participate during AVD by

exhibiting kinetics similar to RVD. The results presented in this dissertation will

offer insights into the following areas: 1) Provide a side by side comparison of the

cellular mechanisms of hyposmotic RVD and apoptotic AVD in HLE-B3 cells. 2)

As LEC are the primary targets of environmental stressors, such as UV

radiations (Hightower et al., 1994), understanding AVD will provide information

Page 42: Ion Transport Mechanisms during Hyposmotic Regulatory and

31

on the early events occurring at the membrane level in HLE-B3 cells during

stressor-induced apoptosis potentially leading to cataract formation. Thus, this

study will provide new information on the possible molecular basis of cataract

formation.

Page 43: Ion Transport Mechanisms during Hyposmotic Regulatory and

32

III. Materials and Methods

3.1. Reagents

3.1.1. Chemicals.

Analytic grade chemicals such as MgCl2, perchloric acid (PCA), dimethyl

sulfoxide (DMSO), tris(hydroxymethyl)aminomethane (Tris), 4-(2-hydroxyethyl)-1-

piperazineethanesulfonic acid (HEPES), potassium chloride (KCl), sodium

hydroxide (NaOH), and fetal bovine serum (FBS) were procured from Fisher

Scientific (Fair Lawn, NJ); ultrapure RbCl was obtained from Alfa (Danvers, MA);

NaCl was from Calbiochem (La Jolla, CA); bovine serum albumin (BSA), CaCl2,

and 3-[N-morpholino] propane sulfonic acid (MOPS) were from Sigma Chemical

(St. Louis, MO); and CsCl, minimum essential medium (MEM), glucose, penicillin,

streptomycin, and amphotericin from Invitrogen-Life Technologies (Carlsbad,

CA).The fluorescent dye N-(ethoxycarbonylmethyl)-6-methoxyquinolinium

bromide (MQAE) was obtained from Invitrogen (Carlsbad, CA).

3.1.2. Inhibitors.

Ouabain, bumetanide, triethylammonium (TEA), staurosporine (STP),

barium, 1-[(2-chlorophenyl) diphenylmethyl]-1H-pyrazole (TRAM-34), and

clotrimazole (CTZ) were procured from Sigma Chemicals and Ba from J. T. Baker

Page 44: Ion Transport Mechanisms during Hyposmotic Regulatory and

33

(Phillipsburg, NJ). Caspase inhibitor Q-VD-OPh NM was a kind gift from Apoptrol

LLC (Beavercreek, OH).

3.1.3. Molecular and immunological tools.

E.Z.N.A. Total RNA Isolation System was purchased from Omega Bio-tek

(Norcross, GA), TaqMan® Reverse Transcription Reagents were obtained from

Applied Biosystems (Branchburg, NJ) and human primers from Integrated DNA

Technologies (Coralville, IA). FastStart PCR Master Mix was purchased from

Roche Diagnostics (Indianapolis IN). Assay-on-DemandTM primers and 2X

TaqMan® Fast Universal PCR Master Mix for qPCR expression were obtained

from Applied Biosystems. Mem-PER protein extraction kit, Halt protease

inhibitors cocktail, and PAGEprep protein clean up kit were from Pierce

Biotechnology (Rockford, IL). Primary antibody against IK channels was obtained

from Alomone laboratories (Jerusalem, Israel). Horseradish peroxidase (HRP)-

coupled donkey anti-rb IgG (H + L chains) for Western blot analysis and a Cy3-

labeled donkey anti-rb IgG for immunofluorescence were procured as secondary

antibodies from Jackson Immunoresearch Laboratories (West Grove, PA), and

fluorescein-labeled donkey anti-rb IgG secondary antibody was from Vector

Laboratories (Burlingame, CA). Lumi-Light Western Blotting substrate was

obtained from Roche Diagnostics (Indianapolis, IN), and Fujifilm Super RX

autoradiography film from Fisher Scientific (Fair Lawn, NJ). Staurosporine-

induced apoptosis was analyzed with a Cell Death Detection Elisa (Roche

Diagnostics Indianapolis IN) and annexin binding with Annexin-V-FLUOS staining

Page 45: Ion Transport Mechanisms during Hyposmotic Regulatory and

34

kit (Roche Diagnostics Indianapolis IN). Cell culture plates and dishes were

obtained from Fisher Scientific (Fairlawn, NJ).

3.2. Solutions and Media

Balanced salt solution (BSS-NaCl) consisted of 20 mM HEPES-Tris buffer

(pH 7.4) containing (in mM): 132 NaCl, 5 KCl, 2 CaCl2, 1 MgCl2, and 10 glucose

and the osmolality was adjusted using NaCl and measured with an Advanced

Micro-Osmometer, model 330 (Advanced Instruments, Norwood, MA). The 300

mOsm washing solution contained 112 mM MgCl2 and 10 mM Tris-MOPS (pH

7.4) at 4°C. As listed in section 2.6, buffer solutions for calibration and

measurement of the intracellular Cl concentration, [Cl]i, were prepared as

described elsewhere (Koncz et al., 1994) and are listed in Table 4.

3.3. Cell Culture

HLE-B3 cells, originally developed by Andley et al., 1994, were purchased

from ATCC (American Type Culture Collection, Rockville, MD) and grown in

MEM supplemented with 20% FBS, penicillin (100 units/ml), streptomycin (100

mg/ml) and amphotericin B (25 mg/ml). HLE-B3 cells were grown to confluence

in an incubator in the presence of 5% CO2 at 37°C. Cells were split after rinsing

with Ca-Mg-free phosphate-buffered saline, PBS (Invitrogen, Carlsbad CA),

addition of 3-5 ml 0.25 % trypsin/EDTA solution (Gibco, Invitrogen,CA) to a 100

mm plate, incubation at 37°C for 10 min, and subsequent neutralization with 3-5

ml complete growth medium containing serum. The suspended cells were

centrifuged at 1000 rpm in a swinging bucket type centrifuge for 3 min, and the

Page 46: Ion Transport Mechanisms during Hyposmotic Regulatory and

35

pellet resuspended in 10 ml of fresh medium. The cells were counted using a

hemocytometer and plated to the desired density described for each experiment

in the subsequent sections and cells no later than passage 60 were used.

3.4. Measurement of Ion Fluxes

Cellular Rb uptake (Rbi) and K content (Ki) in nmoles/mg protein were

measured adapting a protocol for ion fluxes in primary FHL124 lens epithelial

cells (Lauf et al., 2008). HLE-B3 cells were seeded into 12-well plates, grown to

confluence and washed 3 times with 300 mOsm BSS-NaCl solution at 37°C.

Cells were equilibrated in BSS-NaCl-BSA solution for the required time as per

protocol in the presence of inhibitors, or staurosporine treatment or pre-exposing

the cells to BSS-NaCl-BSA solution of different osmolalities. Subsequently, cells

were exposed to flux media containing BSS-Rb/NaCl-BSA as previously

described (Adragna et al., 1991, Lauf et al., 2008) for 10 min. A final wash was

performed with a cold isosmotic washing solution containing 112 mM MgCl2 and

10 mM MOPS/Tris, pH 7.4 at 4oC. Ions were extracted with ice-cold 5%

perchloric acid with 4 mM CsCl for 15 min. 1 N NaOH was added to the plate and

incubated at 37°C for 30 min and the proteins were determined in the lysates by

the BCA method (Pierce Rockford IL). Ki was measured by atomic absorption

spectrophotometry with a Na-K lamp and 85Rb uptake by flame emission

spectrophotometry in a Perkin-Elmer 5000 Atomic Absorption spectrophotometer

(Perkin-Elmer, Boston MA). Based on the absorbance, Rbi and Ki, in nanomoles

per milligram protein, were calculated for each well as described (Adragna et al.,

Page 47: Ion Transport Mechanisms during Hyposmotic Regulatory and

36

1991).

3.5.Measurement of Cell Water

Cell water was measured in HLE-B3 cells using the method described

earlier for FHL 124 cells (Lauf et al., 2008). HLE-B3 cells were plated on 35 mm

dishes and grown to confluence. The medium was aspirated and cells were

washed in BSS-NaCl solutions of various osmolarities and incubated in the

presence or absence of drugs in BSS-NaCl-BSA solution with the appropriate

osmolality. The incubation media were aspirated, residual moisture blotted

thoroughly and dishes weighed for wet weight (WWT) and subsequently dried in

a drying oven at 80°C for 48 h to obtain the dry weight (DWT). Dishes were

incubated for 15 min at 4°C with ice-cold 5% perchloric acid solution with 4 mM

CsCl to extract ions as described (Lauf et. al., 2008) and supernatants collected.

1 N NaOH (3 ml/dish) was added and the total protein content in the dish

determined by BCA assay as described earlier. Cell water content was calculated

from the difference between WWT and DWT in mg/ mg protein per 35 mm dish.

3.6. Measurement of the Intracellular Chloride Concentration [Cl]i

The fluorescent dye N-(ethoxycarbonylmethyl)-6-methoxyquinolinium

bromide (MQAE) was used by modifying methods described by Koncz et al.

(1994) and West et al. (1996).

3.6.1. Treatment of HLE-B3 cells with MQAE

To HLE-B3 cells, cultured in 24-well plates, MQAE was added at a final

concentration of 5 mM in the presence of serum-free medium and cells were

Page 48: Ion Transport Mechanisms during Hyposmotic Regulatory and

37

incubated in an incubator at 37°C in presence of 5% CO2 for 2 h.

3.6.2. Measurement of MQAE fluorescence:

After MQAE loading, HLE-B3 cells were incubated with the appropriate Cl

containing buffer (listed in Table 1) for 45 min either in the presence or absence

of ionophores. The fluorescence of the plate was measured in Fusion™

Universal Microplate Analyzer (Perkin-Elmer, Boston MA) under excitation of 360

nm and emission of 460 nm as described (West et al 1996). The fluorescence

measured was used to calculate [Cl]i as described in the subsequent section.

3.6.3. Calibration of MQAE fluorescence

MQAE fluorescence was calibrated against [Cl]i using the double ionophore

technique with 5mM nigericin and 10 mM tributyltin in high K media. Tributyltin is

an ionophore that exchanges an intracellular Cl for OH, while nigericin

exchanges a K for H. The ionophores were mixed with a mixture of various ratios

of high K and high nitrate buffers to produce [Cl]i between 0 and 60 mM as

described in Table 4. The fluorescence was measured for each equilibration

solution as mentioned in section 3.6.2.

3.6.4.Calculation of [Cl]i

Using the equilibration solutions, the Stern-Volmer quenching constant for

chloride (KCl) was derived from the Stern-Volmer equation for fluorescence

quenching:

F0/F = 1+ KCl[Cl]i

Where F0 and F are MQAE fluorescence in the absence and presence of [Cl]i,

Page 49: Ion Transport Mechanisms during Hyposmotic Regulatory and

38

respectively. The Stern-Volmer constant KCl at equilibration was used in the

equation to determine the unknown [Cl]i from the measured fluorescence F.

Page 50: Ion Transport Mechanisms during Hyposmotic Regulatory and

39

Table 4: Composition of equilibration solutions for [Cl]i measurements

Solutions Concentration (mM) Chloride 0 10 20 30 60

HEPES 5 5 5 5 5 MgSO4 0.8 0.8 0.8 0.8 0.8

NaH2PO4 1 1 1 1 1 Calcium acetate 0.5 0.5 0.5 0.5 0.5

Sodium gluconate 10 10 10 10 10 KCl 0 10 20 30 60

KNO3 60 50 40 30 0 Potassium gluconate 40 40 40 40 40

Potassium bicarbonate 18 18 18 18 18 Tributyltin (μM) 10 10 10 10 10 Nigericin (μM) 5 5 5 5 5

Page 51: Ion Transport Mechanisms during Hyposmotic Regulatory and

40

3.7. DNA fragmentation assay

DNA fragmentation assay was performed as described by Caserta et al.

(2003). HLE-B3 cells were plated at a density of 1 x 106 cells/100 mm dish and

treated with STP or vehicle as per experimental protocol. Cells were collected

and lysed in HL buffer (10 mM Tris, pH 8.0, 1 mM EDTA, and 0.1% Triton X-100)

for 15 min. The DNA from cell lysate was extracted with an equal volume of

phenol and then phenol:chloroform:isoamyl alcohol (25:24:1) and precipitated

with an equal volume of isopropanol and 5 M NaCl. The precipitated cytosolic

DNA was re-suspended in Tris/EDTA, pH 8.0, containing DNase-free RNase A

and incubated at 37◦ C for 30 min. The DNA was separated on a 1.2% agarose

gel in 1X TBE containing ethidium bromide and visualized with Fujifilm LAS-1000

Plus Gel Documentation System (Fuji, Tokyo Japan).

3.8. Cell Death Detection ELISA:

Staurosporine-induced apoptosis was analyzed using Cell Death

Detection Elisa (Roche Diagnostics Indianapolis IN). HLE-B3 cells were grown at

a density of 105 cells/ml in 6-well tissue culture plates and subjected to the

appropriate treatment. Following treatment, cells were trypsinized, and the cell

pellets harvested and lysed as per manufacturerʼs instructions. After lysis, ELISA

was performed on the supernatants using an anti-histone antibody and anti DNA-

peroxidase (POD) antibody and quantified by photometric analysis. Plates were

read in a plate reader at 405 nm and 490 nm for reference. Development of an

intense color in the STP-treated as compared to control cells indicated DNA

Page 52: Ion Transport Mechanisms during Hyposmotic Regulatory and

41

fragmentation, a characteristic of apoptosis.

3.9. Reverse Transcriptase Polymerase Chain Reaction (RT-PCR):

Primers for Ca-dependent K channels were designed as per Table 5 and

purchased from Integrated DNA Technologies (Coralville, IA). HLE-B3 cells were

seeded at density of 2 x 105 cells/well in 6 well plates and the appropriate

treatment was carried out with BSS-NaCl-BSA of varying osmolalities or by

incubation with staurosporine. Total RNA was isolated from HLE-B3 cells using

E.Z.N.A. total RNA isolation kit (Omega Biotech, GA), as recommended by the

manufacturer. 0.5 µg RNA was used to synthesize cDNA using TaqMan®

Reverse Transcription Reagents (Applied Biosystems) as per manufacturerʼs

instructions. PCR was performed using 1X FastStart PCR Master Mix (Roche

Diagnostics, Indianapolis IN), 100 ng cDNA and 300 nM each of forward and

reverse primers. The mixture was transferred to an Applied Biosystems

GeneAmp 2700 thermocycler and cycled as follows: denaturation by heating to

95°C for 4 min, followed by 40 cycles each at 95°C for 30 sec, annealing at 55°C

for 30 sec and elongation at 72°C for 30 sec. A final extension was performed by

heating to 72°C for 7 min. RNA and primers were denatured by incubating at

65°C for 5 min, and placed on ice. The PCR products were verified by

electrophoresis in 1% agarose with ethidium bromide and visualized with Fujifilm

LAS-1000 Plus Gel Documentation System (Fuji, Tokyo Japan).

Page 53: Ion Transport Mechanisms during Hyposmotic Regulatory and

42

Table 5: Primer Sequences for RT-PCR

Primer Sequence Expected PCR Product Size

BK S AACCCGCCCTATGAGTTTG BK AS GGATGGGATGGAGTGAACAG 159 bp

Actin S CTCCTGAGCGCAAGTACTCC

Actin AS CACCTTCACCGTTCCAGTTT 297 bp

SK1 S TCTCGTTGTCCTCTACCATGC SK1 AS AGTGTCTTCATGACGAAGCG 366 bp SK2 S TTACCCTGGAAACAAAACTAGAGACTT

SK2 AS TGCCTGATGGTCTGGCTTATG 79 bp SK3 S TCCAAGATGCAGAATGTCATGTATG

SK3 AS GGTGAGATGCTCCAGCTTCG 108 bp

IK S TCTCAATCAAGTCCGCTTCC IK AS AGCATGAGACTCCTTCCTGC 457 bp

Page 54: Ion Transport Mechanisms during Hyposmotic Regulatory and

43

3.10. siRNA transfection

Pre-designed siRNA against IK channel (kcnn4 gene) along with negative

control scrambled siRNA were obtained from Ambion (Applied Biosystems). HLE-

B3 cells were seeded in 6-well plates at a density of 2 x 105 cells/well. Cells were

transfected with 100 nM siRNA using oligofectamine, in serum and antibiotic free

medium. The medium was changed after 6 h to serum containing medium, and

the cells were grown for 24 h. On day 2, siRNA transfection was repeated as

described earlier. The cells were grown overnight in serum containing medium,

after which total RNA was extracted from the cells using E.Z.N.A. total RNA

isolation kit (Omega Bio-tek, Norcross GA) as described by the manufacturer.

500 ng RNA was used to synthesize cDNA using TaqMan® Reverse

Transcription Reagents (Applied Biosystems) as per manufacturerʼs instructions.

Predesigned Assay-on-DemandTM Primers for IK channel and GAPDH as internal

control were obtained from Applied Biosystems. qPCR reaction was set up in a

96-well plate by adding 40 ng cDNA, 1X TaqMan® Fast Universal PCR Master

Mix (Applied Biosystems) and 1 µl of Assay-on-DemandTM Primers. All samples

were run in triplicate with GAPDH as an internal control. qPCR reaction was

cycled in ABI 7900 HT thermocycler (Applied Biosystems) and relative quantity

(RQ) and 95% confidence intervals were analysed for IK channel gene

expression using SDS2.2 (Applied Biosystems).

3.11. RT-qPCR for hyposmotic and apoptotic stress

Page 55: Ion Transport Mechanisms during Hyposmotic Regulatory and

44

HLE-B3 cells were seeded in 6 well plates at a density of 2 x 105 cells/well

and the appropriate treatment was carried out with BSS-NaCl-BSA of varying

osmolalities or by incubation with STP. Total RNA was isolated from HLE-B3

cells using E.Z.N.A. total RNA isolation kit as described earlier. Five hundred ng

RNA was used to synthesize cDNA using TaqMan® Reverse Transcription

Reagents (Applied Biosystems) as per manufacturerʼs instructions. Predesigned

Assay-on-DemandTM Primers for qPCR expression of IK channel were obtained

from Applied Biosystems. qPCR reaction was set up in a 96-well plate by adding

40 ng cDNA, 1x TaqMan® Fast Universal PCR Master Mix (Applied Biosystems)

and 1 µl of Assay-on-DemandTM Primers. All samples were run in triplicate and

GAPDH was used as an internal control. qPCR reaction was cycled in an ABI

7900 HT thermocycler (Applied Biosystems) and relative quantity (RQ) and 95%

confidence intervals were analyzed for IK channel using statistical software

SDS2.2 (Applied Biosystems)

3.12 Western Blot:

Intermediate K conductance (IK) channel proteins were demonstrated by

western blot analysis as previously described (Misri et al., 2006, Lauf et al.,

2008) involving the following steps. HLE-B3 cells were cultured to sub-confluency

in 100 mm tissue culture plates. Membrane proteins were extracted using Mem-

Per® eukaryotic protein extraction reagent kit in the presence of Halt™protease

inhibitor cocktail (Pierce, Rockford IL) according to the manufacturerʼs

instructions. Cell lysates were mixed with 2X Laemmli SDS electrophoresis

Page 56: Ion Transport Mechanisms during Hyposmotic Regulatory and

45

sample buffer (Sigma Aldrich, St. Louis MO) in a 1:1 ratio and 20 mg of protein

was loaded on to each lane of a 0.75 mm thick 7.5% polyacrylamide gel in a

Mini-V 8.0 Vertical Gel System (GIBCO®BRL, Invitrogen, Carlsbad CA) for 2 h at

120 V. The protein bands were transferred from the gel to a polyvinylidene

difluoride (PVDF) membrane (Bio-rad, Hercules CA) in a blotting system (Mini-V

8.10Blot Module, GIBCO®BRL, Invitrogen, Carlsbad CA) overnight at 90V. The

membrane was blocked with TBS-T (20 mM Tris-Cl, 500 mM NaCl, pH 7.5 plus

0.05% Tween-20) with 10% milk at room temperature for 1 h and subsequently

incubated overnight at 4 ºC with primary antibody against IK channels (Alomone

Laboratories, Jerusalem, Israel) in a 1:200 fold dilution. The blot was washed

thrice with TBS-T and incubated with horse-radish peroxidase (HRP) coupled

donkey anti-rabbit IgG (Jackson Laboratories) in a 1:5000 dilution for 90 min at

room temperature and developed on X-ray film (Fisher Scientific, Fair Lawn NJ)

after exposure to Lumi-Light Western Blotting substrate (Roche Diagnostics,

Indianapolis IN) for 5 min.

3.13. Immunofluoresence Staining and Fluorescence Microscopy

Immunofluorescence was performed as described by Misri et al., (2006)

and Lauf et al (2008). HLE-B3 cells were plated on an 8 well Lab-Tek Chamber

Slide ™(NUNC) at a density of 6 x 104 cells/well as described in section A. Cells

at 40% confluence were fixed and permeabilized with a freshly prepared 4%

paraformaldehyde solution containing 0.1% saponin solution at 4ºC for 30 min,

washed thrice with 1X solution of PBS for 5 min each and subsequently blocked

Page 57: Ion Transport Mechanisms during Hyposmotic Regulatory and

46

with 3% normal goat serum in PBS for 1 h at 4ºC. Cells were incubated overnight

with primary antibody diluted as shown in Table 6, at 4ºC and then incubated with

Cy3-conjugated or fluorescein-conjugated donkey anti-rabbit IgG (Jackson

Laboratories) at 1:250 fold dilution for 90 min at room temperature under light

protection. The slide was mounted using Vectashield™ with DAPI (4',6-

diamidino-2-phenylindole) as mounting medium and examined using appropriate

filters on a Nikon Labophot epifluorescence microscope (Nikon Corporation,

Japan). Images were obtained under 20X, 40X and oil immersion 60X objectives

using SPOT digital color camera (Diagnostic Instruments, Sterling Heights MI)

and analyzed using SPOT Advanced image analysis software (Diagnostic

Instruments, Sterling Heights MI).

3.14. Annexin-V-FLUOS staining

HLE-B3 cells were plated on an 8-well Lab-Tek Chamber Slide ™ (NUNC)

at 1 x 104 cells/well (Misri et al., 2006, Gagnon et al., 2007) and subjected to

appropriate treatment. Annexin-FLUOS staining kit was obtained from Roche

Diagnostics and the labeling solution was prepared according to the

manufacturerʼs protocol. After treatment, the removable chambers on the slides

were removed (according to manufacturerʼs instructions), the medium was

aspirated and cells were incubated in the dark with the labeling solution for 10

min and analyzed using appropriate filters on a Nikon Labophot epifluorescence

microscope (Nikon Corporation, Japan). Images were obtained under 20X, 40X

and oil immersion 60X objectives using SPOT digital color camera (Diagnostic

Page 58: Ion Transport Mechanisms during Hyposmotic Regulatory and

47

Instruments, Sterling Heights MI) and analyzed using SPOT Advanced image

analysis software (Diagnostic Instruments, Sterling Heights MI).

3.15. Statistical Analysis:

Unpaired or paired Student t-tests and one-way ANOVA between the

groups were calculated using STATISTIX 7 software (Analytical Software,

Tallahassee FL). p values < 0.05 were considered significant. Charts were

plotted using Origin 7 software (Origin labs, Northampton MA).

Page 59: Ion Transport Mechanisms during Hyposmotic Regulatory and

48

Table 6: Antibody concentrations for immunofluorescence experiments

Antibody Role Dilution Source

Anti-IK Primary 1:100

Anti-BK Primary 1:100

Anti-SK2 Primary 1:100

Anti-SK3 Primary 1:100

Alomone Labs, Jerusalem, Israel

Anti-β-crystallin Primary 1:200

Anti-KCC1 ECL Primary 1:200

Anti-KCC1CTD Primary 1:200

Anti-KCC3 ECL Primary 1:200

Anti-KCC3 CTD Primary 1:200

As described in Misri et al., 2006

Anti-KCC4 Primary 1:250 Alpha Diagnostics Inc, TX

Cy3 conjugated donkey anti-rabbit

Secondary 1:250

Fluorescein conjugated donkey anti-rabbit

Secondary 1:500

Jackson Immunoresearch Laboratories, West Grove, PA

Page 60: Ion Transport Mechanisms during Hyposmotic Regulatory and

49

IV. Results:

4.1 Characterization of HLE-B3 cells

HLE-B3 is an immortalized cell line obtained by explant culture of isolated

epithelium fragments from human lenses from infants undergoing treatment for

retinopathy of prematurity. The cells were immortalized by infection with

adenovirus Ad12-SV40 (Andley et al., 1994). Positive immunofluorescence

staining of cultured HLE-B 3 cells using an antibody against β-crystallin, a marker

protein for the lens epithelial cells, confirmed that HLE-B3 cells were lens

epithelial in origin (Figure 6).

4.2 Measurement of intracellular potassium concentration [K]i

Intracellular potassium content (Ki) was measured in HLE-B3 cells by

atomic absorption spectrophotometry (see Materials and Methods). The

methodology for measurement of Ki was based on previous studies in vascular

endothelial cells (Adragna et al., 1991), vascular smooth muscle cells (Adragna

et al., 2000), transfected HEK 293 cells (Zhang et al., 2001) and primary FHL

124 LEC (Lauf et al,. 2008). Briefly, Ki in HLE-B3 cells was measured after

washing in 300 mOsm BSS-NaCl at 37℃ and was expressed in nmoles/mg

protein. The values of measured Ki in individual experiments are summarized in

Table 7 and from these values, a mean Ki was calculated to be 16.59mM.

Page 61: Ion Transport Mechanisms during Hyposmotic Regulatory and

50

Baseline values of cell water were measured (see Materials and Methods)

by gravimetric analysis, as described in previous studies using FHL 124 primary

LEC (Lauf et al., 2008) and are summarized in Table 8. The mean baseline cell

water content was determined to be 100 mg/mg protein.

[K]i in the HLE-B3 cells was calculated as the total Ki in nmol/mg protein

measured per total cell water content in mg/mg protein. The data from Tables 7

and 8 were used to estimate [K]i from the following relationships:

Measured Ki= 1,659 nmol/mg protein

Measured cell water = 100 mg/mg protein

[K]i= Ki/cell water

[K]i = (1,659 nmol/mg protein)/(100 mg/mg protein)

[K]i = 16.6 nmoles /mg H2O= 16.6 mM.

The values of [K]i obtained above were lower than anticipated for high K cell and

suggests that the total K content measured was too low or the measured cell

water content was too high in this experiment. A detailed explanation for these

results is provided in section V (Discussion).

Page 62: Ion Transport Mechanisms during Hyposmotic Regulatory and

51

Figure 6: Immunofluorescence labeling of HLE-B3 cells.

HLE-B3 cells were grown on chamber slides, fixed in 4% paraformaldehyde,

permeabilized with 0.01% saponin and incubated with a 1:200 fold dilution of β-

crystallin antibody overnight (see Materials and Methods). Cells were treated with

a 1:250 fold diluted Cy-3 conjugated donkey anti-rabbit antibody, whereas nuclei

were stained with DAPI and observed with a fluorescence microscope under 20X

magnification.

Page 63: Ion Transport Mechanisms during Hyposmotic Regulatory and

52

Figure 6:

Page 64: Ion Transport Mechanisms during Hyposmotic Regulatory and

53

Table 7: Measured Ki in HLE-B3 cells

Cell Line Passage Ki

nmol/mg protein

Mean nmol/mg protein

SD Mean Ki nmol/mg protein

SEM

1 B3 71 1,684.17 1,539.63 125.47 B3 71 1,458.71

B3 71 1,476.01 2 B3 71 1,923.65 1,934.28 61.38 1,659.025 87.37 B3 71 1,878.91

B3 71 2,000.29 3 B3 44 1,657.62 1,589.77 83.90 B3 44 1,615.73

B3 44 1,495.95 4 B3 44 1,623.82 1,572.42 69.04 B3 44 1,493.94

B3 44 1,599.51

Page 65: Ion Transport Mechanisms during Hyposmotic Regulatory and

54

Table 8: Measured cell water content in HLE-B3 cells

# Mean Cell Water mg H2O/ mg protein

(From individual experiments)

Mean Cell Water mg H2O/ mg protein

SEM

1 100

2 96 100.25 3.86

3 98

4 103

Page 66: Ion Transport Mechanisms during Hyposmotic Regulatory and

55

4.3 Measurement of intracellular chloride concentration [Cl]i

4.3.1 Characterization of MQAE fluorescence

Chloride measurements were carried out using the chloride-sensitive

fluorescent dye MQAE, a water-soluble quinolinium derivative. It detects [Cl]i by

diffusion-limited collisional quenching (Jayaraman et al., 2000) where [Cl]i is the

quencher. Once in solution, MQAE diffuses into the cell, reaches an excited state

due to hydrolyzation of the ester bond and forms a complex with the quencher

(Cl) called “encounter complex”. Subsequently, intermolecular charge transfer

occurs within the complex, followed by quenching and then dissociation of the

complex. HLE-B3 cells were loaded with medium containing 5 mM MQAE and

subsequently equilibrated in solutions containing variable [Cl]o and ionophores

(see Materials and Methods). Consistent with fluorescence quenching of MQAE

by Cl, fluorescence was maximum at 0 mM [Cl]o and exponentially decreased

with increasing [Cl]o in the equilibration solution (Figure 7). At a constant dye

loading time of 3 h, measured MQAE fluorescence increased as a function of dye

concentration in the absence of [Cl]o (Figure 8A). Loading HLE-B3 cells with

medium containing 5 mM MQAE for 0-6 h in the absence of [Cl]o, yielded

maximum fluorescence between 1-3 h (Figure 8B). Based on these observations,

a MQAE loading dose of 5 mM for 2 h was selected for all subsequent

measurements of [Cl]i.

Page 67: Ion Transport Mechanisms during Hyposmotic Regulatory and

56

Figure 7: MQAE fluorescence as a function of chloride concentration.

HLE-B3 cells were grown on a 6-well tissue culture plate and loaded with

medium containing 5 mM MQAE for 2h and subsequently incubated in

equilibration buffers containing 0, 10, 20, 30 and 60 mM [Cl-]i, respectively, in the

presence of 10 μM tributyltin and 5 μM nigericin for 45 min. MQAE fluorescence

was measured at 360 nm excitation and 460 nm emission (see Materials and

Methods) and plotted on Y-axis in terms of relative fluorescence units (RFU).

Data represent mean ± SEM for three independent experiments, each

experiment done in quadruplicate.

Page 68: Ion Transport Mechanisms during Hyposmotic Regulatory and

57

Figure 7:

Page 69: Ion Transport Mechanisms during Hyposmotic Regulatory and

58

Figure 8: MQAE fluorescence as a function of dose and time of incubation

with MQAE.

Quantification of MQAE fluorescence at 360 nm excitation and 460 nm emission

was done (see Materials and Methods) in HLE-B3 cells incubated in 0 mM Cl-

containing buffer for 45 min in the presence of 10 μM tributyltin and 5 μM

nigericin for 45 min as a function of MQAE concentration (A) and time of

incubation (B). MQAE fluorescence was plotted on y-axis in terms of relative

fluorescence units (RFU). Results are mean ± SEM for three separate

experiments each done in triplicate.

Page 70: Ion Transport Mechanisms during Hyposmotic Regulatory and

59

Figure 8:

Page 71: Ion Transport Mechanisms during Hyposmotic Regulatory and

60

Previous studies describing the methodology for measurement of [Cl]i

used the “double ionophore technique” by equilibrating MQAE loaded cells with

equilibration solutions containing ionophores tributyltin (Cl/OH) and nigericin

(K/H) prior to fluorescence measurements (Koncz et al., 1994, West et al., 1996,

Miyazaki et al., 2007). When HLE-B3 cells were equilibrated in 0 mM [Cl]o for 45

min in the presence of varying tributyltin and constant nigericin concentrations,

MQAE fluorescence decreased at tributyltin concentrations greater than 30 μM

(Figure 9A). In a similar experiment wherein cells were equilibrated in 0 mM [Cl]o

for 45 min containing 10 μM tributyltin and varying nigericin concentrations,

maximum MQAE fluorescence occurred at a nigericin concentration of 5 μM

(Figure 9B). Therefore, final optimum concentrations of 10 μM tributyltin and 5

μM nigericin was added to the equilibration solutions and used to measure [Cl]i in

HLE-B3 cells.

Page 72: Ion Transport Mechanisms during Hyposmotic Regulatory and

61

Figure 9: MQAE fluorescence as a function of tributyltin and nigericin.

MQAE fluorescence in relative fluorescence units (RFU) was measured at 360

nm excitation and 460 nm emission in HLE-B3 cells incubated for 45 min in 0 mM

[Cl]o solution with varying doses of (A) tributyltin in the presence of 5 μM nigericin

and (B) nigericin in presence of 10 μM tributyltin. Data represent mean ± SEM for

three independent experiments done in triplicate.

Page 73: Ion Transport Mechanisms during Hyposmotic Regulatory and

62

Figure 9:

Page 74: Ion Transport Mechanisms during Hyposmotic Regulatory and

63

4.3.2 Measurement of [Cl]i

[Cl]i was initially estimated in HLE-B3 cells using the following two

methods:

4.3.2.1. Using regression analysis

To estimate [Cl]i, an equilibration (calibration) curve was plotted displaying

the MQAE fluorescence measured at 460 nm as a function of chloride

concentration (see Materials and Methods) after incubation for 45 min (Figure 10

A). MQAE fluorescence for HLE-B3 cells cultured in isosmotic media was

measured as 777 units. A linear relationship characterized the logarithm of

fluorescence versus chloride concentration (Figure 10 B). Based on the linear

equation of y= A+B.x, an intercept A= 8.98 and slope B= -0.027, a y value of 6.65

was calculated for the antilog of the fluorescence of 777 units, which upon

substitution in the above regression equation generated an x value of 86.1 mM

[Cl]i. Three similar experiments were performed and the mean [Cl]i of 87.3 ± 1.22

mM was calculated by regression analysis. This value unfortunately was outside

the range of Cl-standards used and hence the value needs to be interpreted with

caution.

4.3.2.2. Using Stern-Volmer quenching equation

As described earlier, MQAE, once within the cell, is excited after

hydrolysis of itʼs ester group and itʼs fluorescence is quenched by [Cl]i, the

kinetics of which is quantified by the Stern-Volmer equation (Koncz et al.,1994)

(see Materials and Methods). The Stern-Volmer plot showing fluorescence

Page 75: Ion Transport Mechanisms during Hyposmotic Regulatory and

64

intensity (F0/F, Y-axis) as a function of [Cl]i (X-axis value) is displayed in Figure

11. The slope of the linear regression line is the Stern-Volmer constant (KCl) for

[Cl]i, which was calculated as 50 M-1. MQAE fluorescence in HLE-B3 cells with

unknown [Cl]i was measured as 777 units. These values were used to calculate

the baseline [Cl]i of 93 mM in the HLE-B3 cells. Baseline [Cl]i values along with

their Stern-Volmer constants measured in different experiments are summarized

in Table 9. As seen in table 9, a baseline [Cl]i value of 111.44 mM was calculated

for HLE-B3 cells and these values are compared to reported [Cl]i values in other

mammalian cell types in Section V .

Page 76: Ion Transport Mechanisms during Hyposmotic Regulatory and

65

Figure 10: Measurement of [Cl]i by regression analysis.

(A) MQAE fluorescence was measured in terms of relative fluorescence units

(RFU), after equilibration in buffers containing 0 10, 20, 30 and 60 mM [Cl]o and

in the presence of 10 μM tributyltin and 5 μM nigericin for 45 min. (B) The

logarithm of measured MQAE fluorescence was plotted as a linear function of Cl

concentration (insert). The unknown Cl concentration was calculated by

substituting the value of logarithm of MQAE fluorescence for the unknown

sample in the equation of a straight line. Results are mean ± SD of a

representative experiment done in multiples of 3 out of a total of three

experiments. The mean value of [Cl]i calculated from all three experiments was

87.3 ± 1.22 mM

Page 77: Ion Transport Mechanisms during Hyposmotic Regulatory and

66

Figure 10:

Page 78: Ion Transport Mechanisms during Hyposmotic Regulatory and

67

Figure 11: Stern-Volmer plot for MQAE fluorescence.

MQAE fluorescence was measured after incubating cells in buffers containing 0,

10, 20, 30 and 60 mM [Cl]o in the presence of 10 μM tributyltin and 5 μM nigericin

for 45 min (see Materials and Methods). The ratio of F0/F (ordinate) was plotted

as a function of [Cl-]i (abscissa), where F0 and F are MQAE fluorescence in the

absence and presence of [Cl-]i, respectively. The calculated slope from the linear

fit of the data points yielded the Stern-Volmer constant (KCl) for fluorescence

quenching, which was used to determine the unknown [Cl-]i from the measured

fluorescence F by the Stern-Volmer equation (see Materials and Methods). A

typical Stern-Volmer plot for a representative experiment done in triplicate is

shown in the figure and the regression line gives the Stern-Volmer constant (KCl,

which is the slope of the line) of 60 M-1 (r2= 0.989)

Page 79: Ion Transport Mechanisms during Hyposmotic Regulatory and

68

Figure 11:

Page 80: Ion Transport Mechanisms during Hyposmotic Regulatory and

69

Table 9: Stern -Volmer Constant (KCl) and [Cl]i in HLE-B3 cells

Expt # KCl M-1

KCl(M-1) Mean ± SEM

[Cl]i mM

[Cl]i (mM) Mean ± SEM

1 50 52.2 ± 1.5 117 111.4 ± 3.4

2 60 128

3 50 99

4 50 108

5 50 116

6 50 121

7 60 96

8 50 106

9 50 112

Page 81: Ion Transport Mechanisms during Hyposmotic Regulatory and

70

4.4 RVD response in HLE-B3 cells

4.4.1 Effect of hyposmotic stress on cell water content

For hyposmotic stress, HLE-B3 cells were exposed to 300 mOsm

(isosmotic) and 150 mOsm (hyposmotic) BSS and cell water was measured

gravimetrically as a function of time in the presence of 1 mM ouabain and 10 μM

bumetanide as described in previous studies (Lauf et al., 2008). As compared to

isosmotic BSS, cell water increased in hyposmotic BSS about 2-fold in the first 5

min, consistent with hyposmotic swelling of HLE-B3 cells, and fell within 30 min

to below baseline values commensurate with RVD (Figure 12A). Figure 12B

reveals that the ratio of cell water in hyposmotic/isosmotic media, i.e. the

fractional change, was 2 fold at 5 and 10 min and then decreased to base line

values with a calculated efflux rate of ~0.04/min (Figure 12B, insert). As seen in

Figure 12B, the water efflux during RVD was maximal between 10 and 15 min,

and the efflux rate between these two time points was calculated as 0.11/min

3.4.2. Effect of RVD on cellular K and [Cl]i

Ki was measured by atomic absorption spectrophotometry as a function of

time, in the presence of 1mM ouabain and 10 μM bumetanide, during hyposmotic

incubation. Figure 13A shows that after hyposmotic stress, Ki levels decreased

in the first 30 min by 42% with, on the conservative side, a rate constant of

0.03/min as compared to 0.004/min for isosmotic controls (Figure 13B). Thus,

Figures 12 and 13 demonstrate that cell water is equal to or greater than K loss.

Page 82: Ion Transport Mechanisms during Hyposmotic Regulatory and

71

To determine the change in [Cl]i, MQAE fluorescence measurements were

carried out in the presence and absence of 1mM ouabain and 10 μM

bumetanide, after incubation for 30 min in 300 and 150 mOsm media. Figure 14

reveals a 60% increase in MQAE fluorescence upon incubation with 150 mOsm

BSS as compared to incubation with 300 mOsm BSS, meaning that a dilution of

[Cl]i due to water influx and cell swelling had occured.

4.4.3. Effect of RVD on cell survival

To confirm that incubation in hyposmotic media did not alter cell survival in

HLE-B3 cells, DNA fragmentation was quantified in HLE-B3 cells incubated with

150 mOsm hyposmotic solution for 1 h and 2 h using cell death ELISA, (see

Materials and Methods). As shown in Figure 15, no significant change occurred

in the absorbance and hence apptosis in cells incubated in 150 mOsm solution,

as compared to cells incubated in 300 mOsm isosmotic solution for 2 h (Figure

15).

Page 83: Ion Transport Mechanisms during Hyposmotic Regulatory and

72

Figure 12: Effect of hyposmotic stress on cell water.

Cell water was measured gravimetrically,(see Materials and Methods) after

incubating HLE-B3 cells in isosmotic (300 mOsm) or hyposmotic (150 mOsm)

balanced salt solution (BSS). (A) Cell water (ordinate) as function of time

(abscissa) in isosmotic and hyposmotic BSS. (B) Fractional (ratio of

hyposmotic/isosmotic) change in cell water (ordinate) as function of time

(abscissa) were plotted from means in A. Insert in B: natural logarithm of

fractional change of cell water (Δ) vs. time, providing a water efflux coefficient of

0.04/min for the time periods between 5 and 30 min. As described in the

accompanying text, water efflux rate for RVD between time points of 10 and 15

min was calculated as 0.11/min. Data represent mean ± SEM for three

independent experiments done in triplicate. ❋ indicates statistical significance

(p<0.05).

Page 84: Ion Transport Mechanisms during Hyposmotic Regulatory and

73

Figure 12:

Page 85: Ion Transport Mechanisms during Hyposmotic Regulatory and

74

Figure 13: Ki as a function of time during hyposmotic stress.

(A) Ki (in nmol/mg protein, ordinate) measured as a function of time (abscissa) in

isosmotic (300 mOsm) and hyposmotic (150 mOsm) media (see Materials and

Methods). (B) Relative Ki change during 30 min in 150 mOsM hyposmotic media

as a function of time compared to baseline values of Ki (at t=0). The slopes of the

lines yield efflux rates of 0.004/min for isosmotic and 0.03/min for hyposmotic

incubations. Data represent mean ± SEM for for three similar experiments done

in quadruplicate.

Page 86: Ion Transport Mechanisms during Hyposmotic Regulatory and

75

Figure 13:

Page 87: Ion Transport Mechanisms during Hyposmotic Regulatory and

76

Figure 14: Effect of Hyposmotic stress on MQAE fluorescence.

MQAE fluorescence was measured in HLE-B3 cells loaded with 5 mM MQAE

after incubation with isosmotic (300 mOsm) and hyposmotic (150 mOsm) media

for 30 min, as described in the presence or absence of pharmacological inhibitors

of the Na/K pump, (ouabain) and NKCC (bumetanide). Results summarized in

Panel A are mean ± SD of a representative experiment done in triplicate. Total of

two identical experiments done in triplicate are summarized in Panel B are mean

± range.

Page 88: Ion Transport Mechanisms during Hyposmotic Regulatory and

77

Figure 14:

B

Osm Treatment Mean MQAE fluorescence (RFU) ± range

300 mOsm 1,220 ± 133

300 mOsm Ouabain + Bumetanide 1,346 ± 115

150 mOsm 1,937 ± 164

150 mOsm Ouabain + Bumetanide 1,801 ± 302

Page 89: Ion Transport Mechanisms during Hyposmotic Regulatory and

78

Figure 15: Cell death detection ELISA after incubation with hyposmotic

media.

Quantification of apoptosis by absorbance at 405 nm and 495 nm using Cell

Death Detection ELISATM was done (see Materials and Methods) in HLE-B3 cells

treated in isosmotic (300 mOsm) medium for 2 h and hyposmotic (150 mOsm)

medium for 1 and 2 h. In Panel A, data are mean ± SD of a representative

experiment done in triplicate. Data from two separate experiments, each done in

triplicate are summarized as mean ± range in Panel B.

Page 90: Ion Transport Mechanisms during Hyposmotic Regulatory and

79

Figure 15:

A

B

Osm Incubation time Mean Absorbance (RFU) ± range

300 mOsm 2 h 0.61 ± 0.02

150 mOsm 1 h 0.58 ± 0.05

150 mOsm 2 h 0.57 ± 0.02

Page 91: Ion Transport Mechanisms during Hyposmotic Regulatory and

80

4.5. AVD response in HLE-B3 cells.

4.5.1. Time course of cell death

AVD was studied in HLE-B3 cells after inducing apoptosis with STP, a well

known pro-apoptotic fungus-derived peptide that inhibits a broad spectrum of

protein kinases. STP displaces ATP from its binding site and thus compromises

dephosphorylation of a variety of vital cellular metabolic functions (Rüegg

and Burgess 1989). Prior to studying cell water, [Cl]i and K/Rb transport during

AVD, the STP-induced onset and endpoint of cell death was studied in HLE-B3

cells using DNA fragmentation analysis. However, cytosolic DNA isolated from

HLE-B3 cells treated with 2 μM STP for 3 h did not induce the characteristic

“DNA ladder” pattern associated with internucleosomal cleavage, the end stage

of apoptosis, whereas a ladder was observed in control HRP-1 cells treated with

TGF-β (Figure 16, lane 3). As an alternative to detect this end point in the

apoptotic cascade, Cell Death Detection ELISATM, which quantifies DNA

fragmentation by histone-complexed DNA fragments (see Materials and

Methods), was performed. Significant changes in the absorbance differential

between 405 and 490 nm on the ordinate indicating apoptotic cell death occurred

on treatment with 1 and 2 μM STP (Figure 17A) and on incubation for 2, 3 and 4

h with 2 μM STP, as compared to DMSO-treated (control) cells (Figure 17B).

Measurement of total intracellular proteins is important for calculating K

and Rb uptake as well as water content in the cells. However, there was a loss of

cell adhesion on treatment with STP for prolonged periods. Accordingly, total

Page 92: Ion Transport Mechanisms during Hyposmotic Regulatory and

81

protein levels were studied after treating cells with 2 μM STP as a function of

time. Significant loss of protein (75 and 90% at 3 and 5 h, respectively) occurred

in STP-treated cells for time intervals > 2h, but not with vehicle for 5 h (Figure

18). Based on these findings, pre-incubation was done with 2 μM STP for 2 h,

and no further measurements of cell water and ions were performed beyond this

time.

Page 93: Ion Transport Mechanisms during Hyposmotic Regulatory and

82

Figure 16: DNA fragmentation assay after STP treatment

HLE-B3 cells were treated with 2 μM STP for 3 h. Cytosolic DNA was isolated

and electrophoresis was performed on a 2% agarose gel stained with ethidium

bromide and photographed under UV light. HLE-B3 cells treated with DMSO

(vehicle) were the negative control, whereas HRP-1 cells (kindly donated by Dr.

Thomas L. Brown) treated with 1μg/μl TGF-β were used as positive control. Lane

1 and 2: HLE-B3 cells treated with vehicle (DMSO) and with STP, respectively;

Lane 3: HRP-1 cells treated with TGF-β (positive control). Notice that HRP-1 cells

in Lane 3 demonstrated a laddering with fragments of 150 bp in size as

previously described (Caserta et al., 2003). Shown here is a representative

experiment from a total of three identical experiments.

Page 94: Ion Transport Mechanisms during Hyposmotic Regulatory and

83

Figure 16:

180 bp

360 bp

540 bp

720 bp

900 bp

1080 bp

Page 95: Ion Transport Mechanisms during Hyposmotic Regulatory and

84

Figure 17: Cell Death Detection ELISA after STP treatment

Quantification of apoptosis by absorbance at 405 nm and 495 nm using Cell

Death Detection ELISATM was done (see Materials and Methods) in control

(DMSO)- and STP-treated cells as a function of STP dose (A) and time of

incubation (B). Data represent mean ± SEM for for three independent

experiments done in triplicate. ″❋″ indicates statistical significance for p value

<0.05.

Page 96: Ion Transport Mechanisms during Hyposmotic Regulatory and

85

Figure 17:

Page 97: Ion Transport Mechanisms during Hyposmotic Regulatory and

86

Figure 18: Effect of staurosporine treatment on total protein content.

HLE-B3 cells were treated with 2μM STP or DMSO (control) from 0-5 h and total

protein content in mg was measured by the bicinchoninic acid (BCA) assay (see

Materials and Methods). Data in Panel A are mean ± SD of a representative

experiment done in quadruplicate. A total of three independent experiments were

performed and the data are summarized in Panel B as mean ± SEM. The p

values reported in table for STP-treated cells indicated in panel B are in

comparison to the controls.p value < 0.05 was considered statistically significant.

Page 98: Ion Transport Mechanisms during Hyposmotic Regulatory and

87

Figure 18:

A

B

Treatment Incubation time (h)

Mean Total Protein (mg)

SEM p value

0 0.2473 0.048 - Control

5 0.1575 0.066 0 0.2161 0.075 1 0.2261 0.048 > 0.05

2 0.2060 0.046 > 0.05

3 0.0505 0.022 < 0.01

4 0.0387 0.013 <0.01

STP

5 0.0103 0.008 <0.01

Page 99: Ion Transport Mechanisms during Hyposmotic Regulatory and

88

4.5.2. Effect of apoptotic stress on cell volume and cell water content

As demonstrated by Bortner et al., 2004, a pro-apoptotic stimulus in Jurkat

cells changes their membrane potential, opening various ion channels with

subsequent apoptotic volume decrease due to K, Cl and water loss (Cidlowski et

al., 2000, Okada et al., 2001). Cell shrinkage was visually verified by brightfield

microscopy in STP-treated cells after 45 min (Compare Figures 19A and 19 B)

and by immunofluorescence of HLE-B3 cells treated with 2μM STP for 3 h, using

1:100 fold diluted anti–rabbit lens β-crystallin antibody, but not in vehicle-treated

cells as controls (Compare Figures 19C and 19D). Relative cell volume was

measured as cell water content by gravimetric analysis between 5-120 min after

STP treatment (Figure 20): 25% water loss was detected at 30 min and 41% at

45 min, suggesting AVD.The water loss was only 10% at 120 min and lower

compared to 45 minute time point, suggesting transient water loss during AVD.

4.5.3. Effect of AVD on cellular K and [Cl]i

Ki (in nmol/mg protein) was measured after incubating HLE-B3 cells in 2

μM STP or vehicle (controls) at early time points of 5-45 min (Figure 21A) and at

later time points of 1 and 2 h after STP treatment (Figure 21C). Figure 21A and B

show that the change in Ki after STP treatment, relative to vehicle-treated cells,

was insignificant for the first 30 min, with an efflux rate of < 0.007/min, suggesting

that, in contrast to hyposmotic stress, K efflux was not significant early after STP

treatment. However, significant Ki loss with 45% of baseline values occurred

later, 2 h after STP treatment (Figure 21C). Thus the kinetics of K loss and hence

Page 100: Ion Transport Mechanisms during Hyposmotic Regulatory and

89

AVD in STP-treated HLE-B cells in isosomotic BSS is much slower than that of

RVD induced by hyposmotic swelling.

To quantify change in [Cl]i during AVD, MQAE fluorescence was

measured after treating HLE-B3 cells with STP for a course of 2 h Fluorescence

increased by 25% of baseline values in the first 30 min and 19% between 30-60

min suggesting transient [Cl]i changes (Figure 22).

Page 101: Ion Transport Mechanisms during Hyposmotic Regulatory and

90

Figure 19: Immunofluorescent labeling of HLE-B3 cells after STP

incubation.

HLE-B3 cells were incubated with 2 μM STP for 45 min and observed under

brightfield microscope at 40X magnification (B), while DMSO-treated cells were

used as controls (A). Immunofluorescence was performed on cells incubated with

STP (D) and control treated cells (C) for 3 h after they were fixed and

permeabilized with 4% paraformaldehyde and 0.01% saponin, and labeled with

anti-β crystallin antibody (1:200) for immunofluorescence. DAPI was used as

nuclear stain (blue) while fluorescein-conjugated donkey anti-rabbit antibody

(1:250) served as secondary antibody (green). Fluorescence microscopy was

performed at 20X magnification

Page 102: Ion Transport Mechanisms during Hyposmotic Regulatory and

91

Figure 19:

Page 103: Ion Transport Mechanisms during Hyposmotic Regulatory and

92

Figure 20: Effects of staurosporine treatment on cell water.

Cell water was measured gravimetrically as a function of time, after incubating

HLE-B3 cells in 2 µM STP or in DMSO (control) alone from 5-120 min. Data

represent mean ± SEM for three separate experiments (n=3) each done in

multiples of 4. �❋� indicates statistical significance (p value <0.05).

Page 104: Ion Transport Mechanisms during Hyposmotic Regulatory and

93

Figure 20:

Page 105: Ion Transport Mechanisms during Hyposmotic Regulatory and

94

Figure 21: Effects of staurosporine treatment on Ki.

Ki (in nmol/mg protein) measured as a function of time, after incubating HLE-B3

cells in 2 μM STP or DMSO (control) from 5-45 min (A), and for 0, 1 and 2 h (C).

(B) Fold change in Ki after STP treatment, compared to values of Ki in vehicle-

treated (ordinate) as ∆ Ki plotted as function of time (abscissa). Data represent

mean ± range for two separate experiments done in quadruplicate. �❋� indicates

statistical significance (p value <0.05) in STP treated cells as compared to

controls.

Page 106: Ion Transport Mechanisms during Hyposmotic Regulatory and

95

Figure 21:

Page 107: Ion Transport Mechanisms during Hyposmotic Regulatory and

96

Figure 22: Effect of STP treatment on MQAE fluorescence.

MQAE fluorescence (in Relative Fluorescence Units, RFU) was measured in

HLE-B3 cells loaded with 5 mM MQAE after incubation with 2μM STP or DMSO

alone (control) from 0-2 h. Data represent mean ± SEM for three identical

experiments each performed in quadruplicate.�❋� indicates statistical

significance (p value <0.05).

Page 108: Ion Transport Mechanisms during Hyposmotic Regulatory and

97

Figure 22:

Page 109: Ion Transport Mechanisms during Hyposmotic Regulatory and

98

4.6. Membrane transport changes accompanying RVD

Previous studies have shown evidence for IK channels during hyposmotic

swelling of primary LECʼs (Lauf et al., 2008). To test for the presence of a similar

K channel in HLE-B3 cells, the effect of the IK channel inhibitors on RVD-

mediated uptake of 85Rb, a K congener, and Ki loss were studied. Figure 23

shows Rb uptake increased hyperbolically with time in isosmotic BSS controls

and was several fold higher in hyposmotic BSS. This hyposmotic effect was

prevented by addition of 50 μM of the IK channel inhibitor CTZ suggesting IK

channels are involved. As shown in Figure 24, the increased Rb influx in

hyposmotic BSS was also sharply reduced by 50 μM TRAM-34, presumed to be

a more selective IK channel inhibitor, while there was no effect with general

inhibitors of BK channels (2 mM TEA) and voltage-gated K channels (1 mM Ba)

(Figure 24, Table 10). Figure 25A shows that cell water after incubation in

hyposmotic medium containing CTZ remained elevated at levels not significantly

different from the 5 min value in the absence of CTZ, and that the fractional

change in cell water due to RVD was completely prevented by CTZ (Figure 25B).

Thus, similar to FHL124 cells (Lauf et al., 2008), these data suggest that also in

HLE-B3 cells IK channels participate in RVD.

To test whether the increase in MQAE fluorescence during hyposmotic

stress demonstrated in Figure 14 was dependent on the inhibition of IK channels

by CTZ, HLE B3 cells loaded with MQAE were incubated in hyposmotic medium

in the presence and absence of 50 μM CTZ, while controls were incubated in

Page 110: Ion Transport Mechanisms during Hyposmotic Regulatory and

99

isosmotic medium. As shown in Figure 26, the significant increase in MQAE

fluorescence in hyposmotic medium as compared to isosmotic controls remained

unchanged upon addition of CTZ to the hyposmotic medium, suggesting that the

change in [Cl]i is independent of IK channel activation.

Page 111: Ion Transport Mechanisms during Hyposmotic Regulatory and

100

Figure 23: Effect of IK channel inhibitor on Rb uptake during RVD.

Panel A: Rb uptake (in nmol/mg protein, ordinate) was measured after incubation

in hyposmotic (150 mOsm) BSS and hyposmotic BSS containing 50 µM of the IK

channel inhibitor clotrimazole (CTZ), as compared to isosmotic (300 mOsm)

control at 15 and 30 min after incubation and plotted as a second degree

polynomial fit. Shown here is a representative experiment where data represent

mean ± SD for samples done in quadruplicate. Three independent experiments

were performed and the results are summarized in Panel B as mean ± SEM. P

values of statistical significance (p<0.05) are indicated in Panel B and shown by

�❋� in Panel A.

Page 112: Ion Transport Mechanisms during Hyposmotic Regulatory and

101

Figure 23:

A

B

# Time (min) mOsm Rb uptake (nmol/mg protein)

SEM p value Compared to #

A 15 300 38.11 0.25

B 150 87.23 0.93 <0.05 A

C 150 + CTZ 39.54 0.62 <0.05 B

D 30 300 49.207 0.49

E 150 108.77 0.89 <0.05 D

F 150 + CTZ 45.053 1.38 <0.05 F

Page 113: Ion Transport Mechanisms during Hyposmotic Regulatory and

102

Figure 24: Effect of Ca-dependent K channel inhibitors on Rb influx during

RVD

Rb influx at 15 min (in nmol/mg protein, ordinate) was measured in HLE-B3 cells

after incubation with hyposmotic (150 mOsm) and isosmotic (300 mOsm) BSS in

the presence of of IK channel inhibitors: 50 µM CTZ and 50 µM TRAM-34, K

channel inhibitors, Ba (1 mM), and TEA (2 mM). Shown here is a representative

experiment out of three identical experiments, where data represent mean ⑥ SD

for samples done in quadruplicate. The data along with statistical analysis are

summarized in Table 10 as mean ± SEM. Statistical significance at p<0.05 in

hypotonic cells is compared to isosmotic controls and are represented by �❋�,

and in 150 mOsm solutions containing CTZ and TRAM-34 are compared to 150

mOsm control and are represented as �#�.

Page 114: Ion Transport Mechanisms during Hyposmotic Regulatory and

103

Figure 24:

Page 115: Ion Transport Mechanisms during Hyposmotic Regulatory and

104

Table 10 Effect of Ca dependent K channel inhibitors on Rb influx during

RVD

300 mOsm 150 mOsm

Rb Influx (nmol/mg protein) at

15 min (Mean ± SEM)

p value Compared to

Control 17.65 ± 4.5 112.74 ± 4.9 <0.05 300 mOsm control

CTZ 17.83 ± 4.8 29.96 ± 3.7 <0.05

Tram 29.02 ± 0.8 63.64 ± 1.7 <0.05

Ba 28.32 ± 0.64 128.62 ± 5.6 Not Significant

TEA 26.52 ± 0.2 102.11 ± 1.3 Not Significant

150 mOsm control

Page 116: Ion Transport Mechanisms during Hyposmotic Regulatory and

105

Figure 25: Effect of IK channel inhibition by CTZ on cell water during RVD.

(A) Cell water measured gravimetrically as a function of time (ordinate) after

incubating HLE-B3 cells in isosmotic (300 mOsm), hyposmotic (150 mOsm) or

hyposmotic solution in presence of 50 µM CTZ for 5, 15 and 30 min. (B)

Fractional change of cell water as function of time. Data represent mean and

error bars indicate SEM for three independent experiments (n=3), each done in

triplicate. �❋� indicate statistical significance at p<0.05 in values of cell water in

150 v/s. 300 mOsm medium.

Page 117: Ion Transport Mechanisms during Hyposmotic Regulatory and

106

Figure 25:

Page 118: Ion Transport Mechanisms during Hyposmotic Regulatory and

107

Figure 26: Effect of inhibition of IK channels on MQAE fluorescence during

RVD.

MQAE fluorescence was measured after 15 min incubation in hyposmotic (150

mOsm) BSS as compared to isosmotic (300 mOsm) control in the presence or

absence of 50 μM CTZ (see Materials and Methods). Data represent mean ±

range for two identical experiments (n=2) each done in quadruplicate and the

values are indicated in B.

Page 119: Ion Transport Mechanisms during Hyposmotic Regulatory and

108

Figure 26:

A

B

# Treatment Absorbance (RFU)

Mean ± range

1 Vehicle (Control) 14,447 ± 816

2 Vehicle (Control) + CTZ 14,361 ± 789

3 STP 16,373 ± 821

4 STP +CTZ 16,921 ± 850

Page 120: Ion Transport Mechanisms during Hyposmotic Regulatory and

109

4.7. Molecular and immunochemical studies

4.7.1 KCC isoforms in HLE-B3 cells

Previous published studies from this laboratory demonstrate the functional

presence of the K-Cl cotransporter in HLE-B3 cells, which plays an important role

in the regulation of K transport (Lauf et al., 2006) and cell volume regulation in

LEC. Immunofluorescence labeling of HLE-B3 cells for KCC isoforms (Figure

27A-F) (see Materials and Methods) revealed positive labeling including

membrane staining for KCC1 ECL (A), KCC1 CTD (B), KCC3 ECL (D), KCC3

CTD (E) and KCC4 (F) (ECL- Extracellular loop, CTD-C-terminus domain of

KCC), while none was observed for control treated cells, which were incubated in

secondary antibody only (C). DAPI was used as the nuclear stain whereas Cy3-

conjugated donkey anti-rabbit antibody served as secondary antibody.

4.7.2. Ca -dependent K channel expression in HLE-B3 cells.

Functional studies suggested the presence of IK channels in HLE-B3 cells.

However, it is not known whether IK channels and other Ca-dependent K

channels are expressed at the mRNA and protein level in these cells. RT-PCR

analysis of HLE-B3 cells revealed the presence of BK channel (159 bp), IK

channel (457 bp), SK2 (79 bp), and SK3 (108 bp) mRNA with β-Actin (195 bp)

and -RT used as controls (Figure 28A). Western Blot analysis of protein extracts

from HLE-B3 cells revealed a single ~ 75 kDa protein band signifying the

presence of IK channels. Rat brain extracts were used as positive control with

antibody dilutions given in the legend to Figure 28B. Immunofluorescence

Page 121: Ion Transport Mechanisms during Hyposmotic Regulatory and

110

labeling of HLE-B3 cells fixed in 4% paraformaldehyde and 0.01% saponin

showed cytoplasmic and membrane presence of BK, IK, SK2 and SK3 channel

proteins staining. DAPI was used as nuclear stain whereas Cy3-conjugated

donkey anti-rabbit antibody served as secondary antibody (Figure 28C). Staining

was more intense with SK3 antibody and hence DAPI staining cannot be

observed in the superimposed images.

4.8. Effect of IK channel gene knock down by RNAi on Rb uptake during

RVD:

Results described in the previous sections indicate possible involvement

of IK channels based on physiological studies with inhibitors. Since IK channels

were present at the RNA and protein levels in B3 cells, their role during RVD

could be confirmed by performing Rb uptake in HLE-B3 cells transfected with

siRNA against IK channels to knock down the expression of the IK channel gene.

qPCR analysis showed a 24% decrease in IK channel gene expression in siRNA-

transfected cells as compared to untransfected cells (Figure 29A). Scrambled

siRNA was used as control. There was no change in Rb influx (Figure 29B) in

siRNA treated cells after hyposmotic stress at 15 min, as compared to the Rb

influx in controls. Rb influx at 15 min under isosmotic conditions in transfected

and untransfected cells was measured to determine the baseline values, while

HLE-B3 cells incubated in hyposmotic medium in the presence of 50 μM CTZ

were used as positive controls (Figure 29B). The results of siRNA studies are

summarized in Table 11.

Page 122: Ion Transport Mechanisms during Hyposmotic Regulatory and

111

siRNA transfection efficiency was checked by transfecting HLE-B3 cells

with siGLO transfection kit as described in Materials and Methods and 72% of

HLE-B3 cells were transfected (data not shown). These results suggest that,

though significant number of HLE-B3 cells were being transfected by siRNA, the

transfection was not adequate to produce a significant decrease in IK channel

gene expression and subsequently produce a detectable functional effect.

Page 123: Ion Transport Mechanisms during Hyposmotic Regulatory and

112

Figure 27: Immunofluorescence of KCC isoforms in HLE-B3 cells

HLE-B3 cells were fixed in 4% paraformaldehyde and 0.01% saponin, treated

with appropriate antibodies against KCC as outlined in Table 6, and subsequently

observed using fluorescence microscopy (A-C, E, F) and laser confocal

microscopy (D) under a 60X oil immersion objective. DAPI was used as nuclear

stain while Cy3-conjugated donkey anti-rabbit antibody was used as secondary

antibody at a 1:250 fold dilution. Positive labeling was observed for KCC1 ECL

(A), KCC1 CTD (B), KCC3 ECL (D), KCC3 CTD (E) and KCC4 (F), while no

labeling was observed for control treated cells that were incubated in the

secondary antibody only (C). For antibody dilutions, see Materials and Methods.

Images correspond to a representative experiment from a total of three identical

experiments.

Page 124: Ion Transport Mechanisms during Hyposmotic Regulatory and

113

Figure 27:

Page 125: Ion Transport Mechanisms during Hyposmotic Regulatory and

114

Figure 28: Molecular evidence of Ca-dependent K channels in HLE-B3

cells.

(A) RT-PCR analysis of HLE-B3 cells reveals presence of BK channel (159 bp),

IK channel (457 bp), SK2 (79 bp), and SK3 (108 bp) mRNA while β-Actin (195

bp) and -RT were used as controls. (B) Western blot analysis of protein extracts

from HLE-B3 cells for presence of IK channel protein. Rat brain extracts were

used as positive control for this experiment (labeled as “control”). Primary

antibody dilution was 1:500, while secondary HRP conjugated donkey anti-rabbit

antibody was 1:2000 fold diluted (C) Immunofluorescence labeling of HLE-B3

cells fixed in 4% paraformaldehyde and 0.01% saponin revealed presence of IK,

BK, SK2 and SK3 channel proteins by fluorescence microscopy, observed under

10X magnification. DAPI was used as nuclear stain (blue) while Cy3-conjugated

donkey anti-rabbit antibody (red) as secondary antibody at a 1:250 fold dilution.

Antibody dilutions are summarized in Table 6.

Page 126: Ion Transport Mechanisms during Hyposmotic Regulatory and

115

Figure 28:

Page 127: Ion Transport Mechanisms during Hyposmotic Regulatory and

116

Figure 29: Effect of IK channel gene knock down by RNAi on Rb influx

(A) IK channel gene expression in siRNA-transfected HLE-B3 cells: HLE-B3 cells

were transfected using oligofectamine (see Materials and Methods). qPCR

analysis for gene expression of IK channel in cells transfected with two separate

pre-designed siRNAs (siRNA1 and siRNA2) as well as untransfected controls

and scrambled siRNA negative control. Y-axis is the Relative Quantity (RQ)

value in log10. as compared to endogenous GAPDH expression and the fold

change is indicated above the bars. Statistical significance was determined by

calculating the 95% confidence interval for the RQ value and is indicated in the

graph by error bars. (B) Rb influx at 30 min in IK channel siRNA-transfected

HLE-B3 cells after incubation in isosmotic (300 mOsm) and hyposmotic (150

mOsm) media. siRNA transfection, as well as Rb influx measurements were

performed as described in Materials and Methods. HLE-B3 cells treated with

CTZ and incubated in hyposmotic media were used as a positive control. Shown

in panels A and B is a representative experiment where data represent mean ±

SD for samples done in triplicate. All the data from siRNA studies are

summarized in Table 11.

Page 128: Ion Transport Mechanisms during Hyposmotic Regulatory and

117

Figure 29:

Page 129: Ion Transport Mechanisms during Hyposmotic Regulatory and

118

Table 11: Summary of siRNA experiments:

Expt Transfection IK channel qPCR Rb influx x 15 min in 150 mOsm BSS

siRNA 2: 100 ng 25% inhibition No Effect 7

siRNA 1: 100 ng 10 % activation No Effect

siRNA 2: 100 ng 24% inhibition No Effect 9

siRNA 1: 100 ng 17% activation No Effect

siRNA 2: 100 ng 32% inhibition No Effect 12

siRNA 1: 100 ng 14% inhibition No Effect

Page 130: Ion Transport Mechanisms during Hyposmotic Regulatory and

119

4.9. Membrane transport changes accompanying AVD:

Physiological [K]i prevents apoptosis, which is also inhibited in the

presence of elevated [K]o, (Thompson et al., 2001, Bortner et al., 1997). To

further characterize the K loss inducing AVD, HLE-B3 cells were treated with

STP or vehicle in the presence of normal (5mM) and elevated (135 mM) [K]o for 2

h and the Cell Death ELISA was performed. In the presence of 135 mM [K]o, the

degree of DNA fragmentation, and hence apoptosis, was reduced by 21% in

STP-treated cells, whereas DNA fragmentation remained unchanged in vehicle-

treated cells (Figure 30).

IK channels are stress-activated channels, and along with hyposmotic

stress, they mediate apoptotic cell death in granulocytes (Fay et al., 2006), but

their role during AVD of LECs is generally not defined. Since both FHL124 cells

(Lauf et al., 2008) and HLE-B3 cells (this study) exhibit CTZ- and TRAM34-

sensitive hyposmotically activated IK channels, we tested whether Rb uptake

stimulated by STP would be inhibited by CTZ. Figure 31 shows a significant

increase of Rb uptake above vehicle-treated controls at 45 and 60 min, which

was not inhibited by 50 μM CTZ. Likewise, the loss of Ki in STP-treated cells after

30 and 60 min was not abrogated by addition of CTZ to the STP-containing

isosmotic medium (Figure 32). As expected, loss of cell water at 30 and 60 min

after STP treatment was not prevented by addition of CTZ to the incubation

medium (Figure 33). At an early time point of 15 min, MQAE fluorescence was

significantly increased in STP-treated cells in the presence or absence of CTZ,

signifying that the change in [Cl]i is independent of IK channel inhibition (Figure

Page 131: Ion Transport Mechanisms during Hyposmotic Regulatory and

120

34). Thus, the pharmacological data do not support a role of IK channels in AVD

of HLE-B3 cells. Furthermore, there was no significant decrease in the amount of

DNA fragmentation in cells incubated with 2 μM STP for 3 h in the presence or

absence of CTZ, and the values were significantly higher as compared to DMSO-

treated controls (Figure 35).

Page 132: Ion Transport Mechanisms during Hyposmotic Regulatory and

121

Figure 30: Cell death detection ELISA after STP treatment in the presence

of low and high [K]o.

Panel A shows quantitation of apoptosis by measurement of absorbance at 405

nm and 495 nm using Cell Death Detection ELISATM(see Materials and Methods)

for 2 h in vehicle (DMSO) - and STP-treated cells in the presence of low [K]o (5

mM) and high [K]o (135 mM). A representative experiment is shown in Panel A

where bars represent mean ± SD for samples done in triplicate. Summary of data

as mean ± range are shown in panel B for two identical experiments each

performed in triplicate. Comparative statistical analysis is also summarized in

panel B as indicated by p values and significant statistical differences (p<0.05)

are indicated in panel A.

Page 133: Ion Transport Mechanisms during Hyposmotic Regulatory and

122

Figure 30:

A

B

# KO Treatment Absorbance (RFU)

Mean ± range

p value Compared to #

A 5 mM Vehicle (Control) 0.97 ± 0.017 -

B STP 1.55 ± 0.029 < 0.05 A

C 135 mM Vehicle (Control) 1.16 ± 0.034 > 0.05 A

D STP 1.28 ± 0.066 < 0.05 B

Page 134: Ion Transport Mechanisms during Hyposmotic Regulatory and

123

Figure 31: Effect of CTZ on Rb uptake as a function of time during AVD.

Rb uptake (nmol/mg protein) was measured as a function of time after incubating

HLE-B3 cells in 2 µM STP, vehicle (control) or 2 µM STP in the presence of 50

µM CTZ from 5-60 min in the presence of 1 mM ouabain and 10 µM bumetanide

(see Materials and Methods). Data represent mean ± SEM for three identical

experiments done in quadruplicate. �❋� represents statistically significant p-value

<0.05.

Page 135: Ion Transport Mechanisms during Hyposmotic Regulatory and

124

Figure 31:

Page 136: Ion Transport Mechanisms during Hyposmotic Regulatory and

125

Figure 32: Effect of CTZ on Ki as a function of time during AVD.

Ki (nmol/mg protein, ordinate) was measured as a function of time (abscissa)

after incubating HLE-B3 cells in 2 μM STP alone, control (DMSO) or 2 μM STP

along with 50 μM CTZ for 10 , 30 and 60 min in the presence of 1 mM ouabain

and 10 μM bumetanide (see Materials and Methods). Data represent mean ±

SEM for three identical experiments (n=3), each done in quadruplicate.

Page 137: Ion Transport Mechanisms during Hyposmotic Regulatory and

126

Figure 32:

Page 138: Ion Transport Mechanisms during Hyposmotic Regulatory and

127

Figure 33: Effect of CTZ on cell water during AVD.

Cell water measured gravimetrically after 5, 30 and 60 min incubation of HLE-B3

cells in isosmotic medium containing 2 μM STP, DMSO (control) or 2 μM STP in

the presence of 50 μM CTZ. Data represent mean ± SEM for three separate

experiments (n=3), each done in quadruplicate.

Page 139: Ion Transport Mechanisms during Hyposmotic Regulatory and

128

Figure 33:

Page 140: Ion Transport Mechanisms during Hyposmotic Regulatory and

129

Figure 34: Effect on CTZ on MQAE fluorescence during AVD.

MQAE fluorescence was measured after 15-min incubation in isosmotic medium

containing 2 μM STP and vehicle (DMSO) in the presence or absence of 50 μM

CTZ (see Materials and Methods). Data represent mean ± SEM for three

independent experiments (n = 3), each performed in quadruplicate.

Page 141: Ion Transport Mechanisms during Hyposmotic Regulatory and

130

Figure 34:

Page 142: Ion Transport Mechanisms during Hyposmotic Regulatory and

131

Figure 35: Effect of CTZ on STP-induced apoptosis.

Quantitation of apoptosis by measuring absorbance at 405 nm and 495 nm using

Cell Death Detection ELISATM was done (see Materials and Methods) in vehicle

(DMSO)- and STP-treated cells in the presence of CTZ for 1 h and 3 h. Data

represent mean ± SEM for three independent experiments, each performed in

triplicate.

Page 143: Ion Transport Mechanisms during Hyposmotic Regulatory and

132

Figure 35:

Page 144: Ion Transport Mechanisms during Hyposmotic Regulatory and

133

The prevention of apoptotic cell death by increasing [K]o provided

sufficient evidence of involvement of a K channel in STP-induced AVD and cell

death in HLE-B3 cells. To further determine the type of K channels involved

during STP-induced AVD, we measured Rb influx in cells incubated with STP-

containing medium for 45 min in the presence of inhibitors for delayed rectifying

K channels (1mM Ba), large conductance BK channels (2 mM TEA) and voltage-

gated A type K channels (2 mM 4-AP) (Figure 36 A, B, C). The increase in Rb

uptake after incubation with 2 μM STP (Figure 36 A, B and C) was prevented by

adding 2 mM 4-AP to the medium (Figure 36A), while incubation with TEA and

Ba had no effect on Rb uptake (Figure 36B, C). Incubation with STP in the

presence of 2 mM 4-AP for 3 h significantly decreased DNA fragmentation

(Figure 37 A and B) as compared to STP-treated cells, suggesting involvement of

A-type voltage-gated K channels during AVD. However, as seen in Figure 37,

there was a significant increase in the measured DNA fragmentation in control

cells incubated with 4-AP as compared to cells incubated only in control (DMSO),

but these values were lower than DNA fragmentation in STP-treated cells.

Page 145: Ion Transport Mechanisms during Hyposmotic Regulatory and

134

Figure 36: Effect of K channel inhibitors on Rb influx during AVD

Rb influx (in nmol/mg protein, ordinate) was measured in HLE-B3 cells (see

Materials and Methods) after incubation with isosmotic BSS for 45 min in the

presence or absence of 2 µM STP, also containing the following K channel

inhibitors: (A) 2mM 4-AP, (B) 2mM TEA and (C) 1 mM Ba. Data represent mean

± SEM for three separate experiments, each performed in quadruplicate. �❋�

represents statistical significance for p < 0.05

Page 146: Ion Transport Mechanisms during Hyposmotic Regulatory and

135

Figure 36:

Page 147: Ion Transport Mechanisms during Hyposmotic Regulatory and

136

Figure 37: Apoptosis after STP treatment in the presence of 4-amino

pyridine.

Apoptosis was quantified by absorbance measurement at 405 nm and 495 nm

(ordinate) in vehicle- and STP-treated cells in the presence of 2 mM 4-AP for 3 h,

while cells treated with DMSO (control) and DMSO along with 2mM 4-AP were

used as control. Data represent mean ± range for two identical experiments,

each performed in triplicate. Comparative statistics for the results are

summarized in Panel B. p-value<0.05 is statistically significant.

Page 148: Ion Transport Mechanisms during Hyposmotic Regulatory and

137

Figure 37:

A

B

Mean Absorbance (RFU) ± range

p value Compared to

A Control 0.2425 ± 0.02

B Control + 4-AP 0.5018 ± 0.03 < 0.05 C

C STP 1.3455 ± 0.03 <0.05 A

D STP+ 4-AP 0.9898 ± 0.0002 < 0.05 C

Page 149: Ion Transport Mechanisms during Hyposmotic Regulatory and

138

4.10. Effect of prolonged hyposmotic and apoptotic stress on IK channel

gene expression.

If indeed IK channels rather than other Ca-activated K channels play a

major role in RVD and AVD, then, prolonged incubation in either hyposmotic BSS

or in the presence of STP should reveal a down-regulation of IK channels as

opposed to the remainder of the K channels shown in the previous section. An

osmolarity-dependent change of transport protein expression has been shown in

eukaryocytes (Hoffmann et al., 2009).

Towards this goal, HLE-B3 cells were incubated in 250, 200 and 150

mOsm hyposmotic media for 24 h and IK channel gene expression was

quantified by qPCR analysis (see Materials and Methods). Cells incubated in 250

mOsm BSS did not show any significant change in gene expression as compared

to controls incubated in 300 mOsm BSS, while IK gene expression was

decreased by 60% in 200 mOsm and 80% in 150 mOsm BSS (Figure 38A). The

time dependence of prolonged hyposmotic stress on IK channel expression is

shown in HLE-B3 cells incubated in 150 mOsm hyposmotic BSS for 1, 3, 6, 12

and 24 h, as quantified by qPCR. There was no significant change in IK gene

expression up to 6 h in hyposmotic media as compared to controls incubated in

isosmotic 300 mOsm media for 24 h, while IK gene expression decreased by

70% after 12 h and 80% after 24 h incubation in 150 mOsm BSS (Figure 38B).

To study the effect of prolonged apoptotic stress, HLE-B3 cells were

incubated in 2 μM STP for 1 and 3 h and IK channel gene expression was

Page 150: Ion Transport Mechanisms during Hyposmotic Regulatory and

139

quantified by qPCR analysis. Cells incubated in STP did not show any significant

change in gene expression up to 1 h as compared to untreated or vehicle-treated

controls at 24 h, while gene expression was decreased by 88% after 3 h

incubation with STP, while the gene expression in control-treated cells was not

downregulated (Figure 39). Although IK gene expression appeared to become nil

even at later time points up to 24 h as shown in Figure 39, we like to attribute

less significance to these data in light of protein changes and DNA fragmentation

in the samples occurring after 3 h (Figure 18). Nevertheless, the molecular data

presented here suggest that IK channel expression is down-regulated or

negatively affected both after prolonged incubation in hyposmotic media and after

STP treatment. This supports the hypothesis that IK channels may indeed play a

major role in RVD, however, their role during AVD cannot be stated definitively on

account of the significant protein loss and DNA fragmentation at 3 h and is

unlikely based on functional and inhibitor studies.

Page 151: Ion Transport Mechanisms during Hyposmotic Regulatory and

140

Figure 38: IK channel gene expression on prolonged hyposmotic stress.

HLE-B3 cells were incubated in (A) 250, 200 and 150 mOsm hyposmotic media

for 24 h and (B) in 150 mOsm hyposmotic media for 1, 3, 6, 12 and 24 h and IK

channel gene expression was quantified by qPCR (see Materials and Methods).

The ordinate shows the Relative Quantity (RQ) value of IK channel gene

expression as compared to endogenous GAPDH expression and represents the

fold-change in gene expression. Statistical significance was determined by

calculating the 95% confidence interval for the RQ value and is indicated in the

graph by error bars. Data correspond to mean ± SD of three identical

experiments done in multiples of 3. �❋� denotes statistical significance at p>0.05.

Page 152: Ion Transport Mechanisms during Hyposmotic Regulatory and

141

Figure 38:

Page 153: Ion Transport Mechanisms during Hyposmotic Regulatory and

142

Figure 39: IK channel gene expression in HLE-B3 cells during apoptotic

stress, as a function of incubation with STP

HLE-B3 cells were incubated in 2 µM STP for 1, 3, 6, 12 and 24 h, and IK

channel gene expression was quantified by qPCR (see Materials and Methods).

The ordinate shows the relative quantity (RQ) value of IK channel gene

expression as compared to endogenous GAPDH expression and represents the

fold-change in gene expression. Statistical significance was determined by

calculating the 95% confidence interval for the RQ value and is indicated in the

graph by error bars. Data correspond to mean ± SD of of three identical

experiments done in multiples of 3 �❋� denotes statistical significance at p<0.05.

Page 154: Ion Transport Mechanisms during Hyposmotic Regulatory and

143

Figure 39:

Page 155: Ion Transport Mechanisms during Hyposmotic Regulatory and

144

4.11. Early membrane changes during AVD:

4.11.1. Annexin Binding during AVD.

Translocation of phosphatidylserine (PS) from the inner to the outer

plasma membrane leaflet is an early event during apoptosis (Vermis et al 1995).

Fluorescein-tagged Annexin-V binds to the exposed PS, which can be detected

by fluorescence microscopy, while propidium iodide (PI) will stain the nuclear

DNA. Annexin-V staining was performed (see Materials and Methods) after

treatment with 2μM STP and DMSO (control) treated for 15, 35, 60, 90 and 120

min (Figures 40 and 41) to analyze the early (Figure 41) and late events (Figure

40) at the membrane level during AVD. Membrane staining was present at 60, 90

and 120 min (Figure 40A, 40B, 40C and 40D, arrows respectively) in STP-

exposed but not in control-treated cells (120 min, Figure 40E) consistent with

detection of apoptosis at these time points. Annexin-V staining and hence PS

translocation appeared after 15 min of STP exposure (Figure 41B) as compared

to vehicle-treated cells (Figure 41A) and became clearly visible after 35 min

(Figure 41D, arrows) in STP- but not in control-treated HLE-B3 cells (Figure

41C). These data imply that PS-translocation effectively happened within 15 min

after STP exposure thus heralding this membrane event as one of the earliest

changes in the apoptotic process.

4.11.2. Annexin binding during AVD in the presence of caspase inhibitor

HLE-B3 cells were preincubated with medium containing 20 μM broad

spectrum caspase inhibitor Q-VD-OPh NM for 30 min and subsequently treated

Page 156: Ion Transport Mechanisms during Hyposmotic Regulatory and

145

with 2 μM STP (see Materials and Methods). Cells preincubated in DMSO

(vehicle) were used as controls. Cells were stained with Annexin-V 30 min after

STP incubation and the number of annexin-labeled cells was counted (Figure

42). Positive membrane staining forannexin was observed for STP treated cells

pre-treated without (Figure 42A) or with Q-VD-OPh NM (Figure 42B) as

compared to untreated controls (Figure 42C). There was no statistical difference

in the number of counted cells that were positive for annexin labeling after STP

incubation in the presence or absence of pretreatment with the caspase inhibitor

(Figure 41D). Together, these results suggest that early positive annexin labeling

is independent of caspase activation and most likely precedes and is necessary

for the subsequent changes in membrane permeability.

Page 157: Ion Transport Mechanisms during Hyposmotic Regulatory and

146

Figure 40: Late effects on Annexin V binding after incubation with 2 μM

STP.

HLE-B3 cells were treated with 2 μM STP or DMSO (control) for 60, 90 and 120

min and subsequently labeled with Annexin V (see Materials and Methods).

Panels A-E represent fluorescence microscopy after labeling with Annexin (green

staining) and propidium iodide (PI, red staining) for STP-treated cells at 20 X

magnification for 60 (A), 90 (B) and 120 min (C), at 60X magnification for 120 min

(D), and control (DMSO) treated cells at 120 min (E), also at 60X magnification.

Yellow arrows indicate membrane labeling by Annexin V.

Page 158: Ion Transport Mechanisms during Hyposmotic Regulatory and

147

Figure 40:

Page 159: Ion Transport Mechanisms during Hyposmotic Regulatory and

148

Figure 41: Early effects on Annexin V binding after incubation with 2 μM

STP.

HLE-B3 cells were treated with 2 μM STP or DMSO (control) for 15 and 35 min

and then incubated with Annexin V (see Materials and Methods). Panels A-D

represent fluorescence microscopy after labeling with Annexin ( green) and PI

(red) for control (DMSO) treated cells at 15 (A) and 35 min (C) at 40 X

magnification and cells treated with 2 μM STP for 15 min (B), and 35 min (D), at

40X magnification. Yellow arrows indicate membrane labeling by Annexin V.

Page 160: Ion Transport Mechanisms during Hyposmotic Regulatory and

149

Figure 41:

Page 161: Ion Transport Mechanisms during Hyposmotic Regulatory and

150

Figure 42: Effect of broad spectrum caspase inhibitor on annexin binding

during AVD.

HLE B3 cells were cultured in 8 well chamber slides to 90% confluency and were

pre-incubated for 30 min with 20 μM broad-spectrum caspase inhibitor Q-VD-

OPh NM (kindly donated by Dr. Thomas L. Brown) or DMSO (control), and

treated with 2 μM STP or DMSO (control) for 30 min. The slide was subsequently

stained with Annexin V-PI solution (see Materials and Methods). Fluorescence

microscopy was performed for Annexin-V (green) and PI (red) labeling at 40 X

magnification in STP treated cells (A), cells pre-incubated with Q-VD-OPh prior to

STP treatment (B) and untreated controls (C). The total number of cells with

positive membrane staining for annexin in each well were counted and expressed

in Panel D as a relative percentage normalized to the number of annexin-positive

cells treated with STP, in the absence of caspase inhibitor (from Panel A).

Page 162: Ion Transport Mechanisms during Hyposmotic Regulatory and

151

Figure 42:

V

Page 163: Ion Transport Mechanisms during Hyposmotic Regulatory and

152

Discussion

5.1. HLE-B3 cells as an in-vitro model system

The lens specific origin of HLE-B3 cells line was confirmed by positive

immunofluorescence staining for β-crystallin (Figure 6), which is a structural

protein in the lens of the vertebrate eye (Wang et al., 2004, Chambers and

Russell, 1993). The HLE-B3 cell line is an established model for studying cell

death in LEC as highlighted by several studies (Liyanage et al., 2007, Andley et

al., 2000, Lee et al., 2002, Futter et al., 2005, Flynn et al., 2006). Recently,

studies have been published which demonstrate ion transport properties in HLE-

B3 cells (Cui et al., 2002, Lauf et al., 2006, Misri et al., 2006). HLE-B3 cells can

be cultured up to passage 76 and do not require any special surface for growth,

making them an attractive model to study ion transport as well as apoptosis.

5.2. Measurement of baseline Ki and water

Previous studies in HLE-B3 cells have demonstrated pathways of

electroneutral K transport mainly KCC and NKCC (Lauf et al., 2006). [K]i was

calculated as 16.6 mM as shown in Results from intracellular K and cell water

content (see Tables 7, 8), a value significantly smaller compared to a reported

[K]i in mammalian cells (Yu and Choi, 2000) and [K]i values of 130-140 mM

measured in explant cultures of LEC and whole human lenses (Stewart et al.,

Page 164: Ion Transport Mechanisms during Hyposmotic Regulatory and

153

1988). These low [K]i can be explained by too high values of cell water content in

the particular experiments reported in Table 8, which are 3-4 times higher than

the cell water content measured in FHL 124 primary LEC (Lauf et al., 2008). In

later experiments, the technique of water determinations was refined and lower

water contents were found supporting this conclusion (see Figures 20,25,33).

More sensitive methods with dilution-sensitive fluorescent dyes such as calcein-

AM are needed to confirm the gravimetric analyses.

5.3. Measurement of [Cl]i

Efflux of intracellular chloride is one of the main effectors during cell

volume decrease (reviewed in Hoffman et al., 2009, Miyazaki et al. 2007).

Verkman et. al (1989) described the use of chloride-sensitive fluorescent dyes

MQAE and SPQ for measurement of [Cl]i in porcine kidney cells. The calculations

were performed by Stern-Volmer transformation of measured dye fluorescence in

the presence of halide quencher ([Cl]i). For the current study, MQAE was used

for [Cl]i measurements as it is water soluble, has a high Cl sensitivity and readily

diffuses across the cell membrane, whereas SPQ is water insoluble and

membrane impermeable (Verkman et al., 1989, Koncz et al., 1994). Due to its

membrane permeability, a loss of MQAE from the cell into the incubation medium

could be expected. However, as seen in Figure 8B, the values of measured

MQAE fluorescence remained constant for 3 h after incubation, and there was no

leakage of dye for MQAE loading times < 3 h. A decrease in MQAE fluorescence

3 h after incubation, which agrees with the published reports of dye leakage to

Page 165: Ion Transport Mechanisms during Hyposmotic Regulatory and

154

the extracellular space of the membrane-permeable MQAE (Koncz et al., 1994),

necessitates shorter incubation times with Cl containing equilibration buffers (see

Materials and Methods), which was achieved by addition of ionophores. As

described in previous studies (Koncz et al., 1994, West et al., 1996, Miyazaki et

al., 2007), MQAE-loaded cells were equilibrated in the presence of the Cl/OH

ionophore tributyltin and K/H ionophore nigericin in concentrations plotted in

figure 9A and 9B, prior to fluorescence measurement, which expedited the

process of equilibration ([Cl]i = [Cl]o) while maintaining the electroneutrality of the

cell (the “double ionophore technique”).

5.4. Regression Analysis vs. Stern-Volmer Analysis for determining [Cl]i

[Cl]i was calculated from regression analysis of measured MQAE

fluorescence as described in earlier sections and calculated as 86.1 mM (Figure

10). This value is greater than published reports of [Cl]i of 56.2 mM in porcine

lymphocytes (Pilas et al., 1997), 40 mM in Jurkat T cells (Lai et al., 2003), 52.4

mM in colonic epithelial cells (West et al., 1996) and 41.5 mM in renal epithelial

cells (Miyazaki et al., 2007).

However, while measuring [Cl]i by MQAE fluorescence, the MQAE

fluorescence is quenched by intracellular chloride by forming a complex with the

fluorophore molecule (described in Results). The kinetics are described by the

Stern-Volmer equation (see Materials and Methods). For this reason and as

supported by the literature, Stern-Volmer transformation of the measured

fluorescence and not regression analysis is the method of choice to determine

Page 166: Ion Transport Mechanisms during Hyposmotic Regulatory and

155

[Cl]i from measured MQAE fluorescence (Verkman et al., 1989, Koncz et al.,

1994, West et al., 1996, Miyazaki et al., 2007). From the measured fluorescence

in unknown MQAE loaded cells, the calculated Stern-Volmer constant of 52 M-1

was used to calculate a [Cl]i of 111.44 mM in HLE-B3 cells (Figure 11, Table 9).

The chloride concentration in human lens epithelial cells is not known to the best

of our knowledge, but by comparison, the calculated values are 4-5 fold higher

than [Cl]i value of 18.3 mmol/kg water measured in intact human lenses

(Reviewed in Zhang and Jacob, 1997) . Preliminary measurement of membrane

potential in HLE-B3 cells by whole-cell patch clamp recordings were performed in

collaboration with Dr. Dan Halm at Wright State University and the resting

membrane potential (Em) was measured to be -27 mV, which was comparable to

the reported membrane potential values of -30 mV in intact small rodent and frog

lenses (Mathias et al., 2007). Based on the measured values of Em, calculated

[Cl]i of 111 mM, and a [Cl]o of 150 mM in the incubation media, the Nernst

potential (ECl) was calculated as -11 mV, a value indicating depolarization. The

higher values of [Cl]i measured in HLE-B3 cells along with the depolarized ECl

values could be due to the activity of the Na-K-2Cl cotransporter (NKCC) present

even under isosmotic conditions as described in earlier studies (Lauf et al., 2006)

or a Cl/HCO3 exchanger rapidly equilibrating Cl as in erythrocytes (Gasbjerg and

Brahm, 1991).

Page 167: Ion Transport Mechanisms during Hyposmotic Regulatory and

156

5.5. Events during RVD

Cell volume regulation is an important property of nucleated mammalian

cells and occurs by RVD in response to hyposmosmotic and by RVI to

hyperosmotic stress (Hoffmann et al., 2009, Hoffman EK and Simonsen LO,

1989, Mongin and Orlov, 2001). In several cell culture models, RVD induced by

hyposmotic cell swelling involves net K (Lauf et al., 2008), Cl (Wang et al., 2000;

Wang et al., 2002, Miyazaki et al., 2007) and water efflux (Wang et al., 2003;

Capo-Aponte et al., 2003). Previously, the role of electroneutral K transport

mechanisms such as KCC and NKCC (K-Cl and Na-K-2Cl cotransport,

respectively) in SV-40 transformed HLE-B3 cells was studied under isosmotic

conditions (Lauf et al., 2006), and also the involvement of IK channels in primary

FHL124 LEC under hyposmotic conditions (Lauf et al., 2008). These studies

demonstrated strong evidence for RVD mechanisms, pointing less to KCC

stimulation but rather to hyposmotic activation of an ouabain- and bumetanide-

insensitive and a CTZ- inhibited K efflux and Rb influx, apparently mediated by IK

channels.

One of the principal aims of the present study was to study the membrane

transport events occurring during RVD in HLE-B3 cells with reference to cell

water, Ki, and Cli and to investigate whether RVD-mediating IK channels, if they

are verifiable, are also involved in AVD occurring in isosmotic media following

pro-apoptotic interventions. We detected within 5 min after hyposmotic stress a

2-fold increase in cell water and hence swelling in HLE-B3 cells. It is well known

Page 168: Ion Transport Mechanisms during Hyposmotic Regulatory and

157

from work on a variety of model cells (Hoffmann et al., 2009) that hyposmotic

stress produces an increase in cell volume within seconds which is then followed

by a RVD with kinetics that differ between cell types. Therefore, our water

analysis gives only ball park magnitudes of water gain and hence cell swelling in

particular, since at 5 min RVD must have been well underway, a fact well

supported by the loss of K content discussed below and reported in FHL124 cells

(Lauf et al., 2008). The water efflux rate of 0.04/min is an underestimate,

especially as it incorporates the 30 min time point. When the first 3 time points

were used to estimate the rate of water loss, the value increased to 0.11/min,

consistent with our previous findings of water loss during hyposmotic stress in

FHL 124 cells (Lauf et al., 2008) as well as studies on hyposmotic RVD in other

epithelial cell culture models such as corneal epithelial cells (Capo-Aponte et al.,

2003) and intestinal epithelial cells (Miyazaki et al., 2007).

Prolonged exposure to hyposmotic stress did not affect cell survival as

indicated by absence of DNA fragmentation after 2 h incubation in 150 mOsm

medium (Figure 15). Substantial K efflux with or without documented membrane

hyperpolarization occurs during RVD (Mongin and Orlov, 2001) and also in

FHL124 primary cells within 15 min of incubation in hyposmotic medium (Lauf et

al., 2008). Consistent with these observations, there was a 42% loss of Ki in

HLE-B3 cells 15 min after incubation in 150 mOsm solution with an efflux rate 7.5

times greater than in cells incubated in isosmotic 300 mOsm solution (Figure 13).

Page 169: Ion Transport Mechanisms during Hyposmotic Regulatory and

158

As indicated in Figure 14, there was a 1.6 fold increase in measured

MQAE fluorescence at 30 min in response to hyposmotic media, which is

commensurate with the expected [Cl]i efflux from HLE-B3 cells. However, one of

the main assumptions while measuring [Cl]i using any fluorescent dye is that the

cell volume has to remain constant (Koncz et al., 1994). As described previously,

RVD is associated with osmotic cell swelling followed by volume decrease.

These cell volume changes cause dilution in [Cl]i and intracellular MQAE

concentrations and hence the fluorescence measurements, if not corrected, are

not reflecting the actual loss of Cli during RVD. Therefore, in this situation, a

change in the dye concentration during hyposmotic stress only provides the [Cl]ii

dilution at a particular chosen time point of the experiment that is due to cell

swelling and does not permit calculation of Cl efflux. To ensure accurate [Cl]i

measurements during RVD, the change in MQAE concentration due to cell

volume change has to be normalized by calculating the intracellular water space

as described in studies by Miyazaki et al. (2007) by gravimetric analysis of

intestinal epithelial cells after measurement of MQAE fluorescence. This value of

intracellular water space was used to adjust for the dilution of MQAE, and to

obtain accurate measurements of [Cl]i.

5.6 Cellular events during AVD

Previous studies on apoptosis described cell shrinkage as an early

change observed during the apoptotic cascade, and precedes DNA

fragmentation (Wyllie et al., 1980). This cell shrinkage was later characterized

Page 170: Ion Transport Mechanisms during Hyposmotic Regulatory and

159

under normotonic conditions as a precursor to apoptosis and defined by Okadaʼs

group as apoptotic volume decrease (AVD) (Maeno et al 2000). Studies show

that AVD is accompanied by a decrease in cell water accompanied by an efflux

of K and Cl from the cell (Arrebola et al., 2005). As a first step in characterizing

the isosmotic AVD in HLE-B3 cells, cytoplasmic DNA extracts were tested for

DNA fragmentation (Figure 16). However, unlike previous published studies on

the presence of DNA laddering in HLE-B3 cells on treatment with TGF-β (Lee et

al., 2002), treatment with 2 μM STP for 3 h did not show the presence of DNA

fragmentation, which was observed in control TGF-β-treated HRP-1 cells. As an

alternative method to detect DNA fragmentation, cell death ELISA was performed

and significant apoptosis was observed on treatment with 2 μM STP for 3 h

(Figure 17A,B) accompanied by a significant loss of total proteins at 3 h (Figure

18), consistent with loss of attached HLE-B3 cells to the culture plate. Since Rb

uptake, Ki, as well as intracellular water were measured in terms of total protein

content, these measurements were conducted for time periods < 3 h. Time

periods of < 3h were of interest while measuring water and ions, since it is

hypothesized that these changes of AVD take place much earlier than DNA

fragmentation, which is the end point of apoptosis and takes place between 2 h

and 3 h as seen in Figure 17B

This study characterized AVD in LEC through changes in cell water, Ki

and [Cl]i. The morphologically observed cell shrinkage after STP incubation

(Figure 21A-D) was due to a 20 and 41% decrease in cell water content at 30

Page 171: Ion Transport Mechanisms during Hyposmotic Regulatory and

160

min and 2 h, respectively (Figure 20), accompanied a 15% and 55% loss of Ki,

by 2 h (Figure 21), not consistent with reports of large K losses “early” during

apoptosis (Bortner et al., 1998). As seen in Figure 22, a 25% increase in MQAE

fluorescence occurred within 30 min of incubation with STP under isosmotic

conditions and therefore was not associated with dilutional effects of the dye, as

in the case with hyposmotic cell swelling. Therefore the measured 25% increase

in MQAE fluorescence at 30 min can be interpreted as a 25% decrease in [Cl]i in

accordance with published reports that Cl efflux accompanies K efflux during

AVD (Maeno et al., 2000). However, at the early time point of AVD, [Cl]i and not

Ki loss is significant and accompanies the significant loss of cell water suggesting

that loss of Cli is the driving force for the process of AVD and this loss of anions

is compensated by a loss of Ki at later time points. We also found that even

though loss of cell water, Ki and [Cl]i started as early as 30 min in HLE-B3 cells,

significant cell death in form of DNA fragmentation was not observed until 2 h

after STP treatment, whereas the events occurring in the intermediate period

remain unknown.

5.7. Ion channels involved during RVD

As in FHL124 cells, the K channel activated by swelling and commencing

RVD is likely the IK channel since both CTZ and TRAM-34 inhibited Rb influx

(Figure 23, 24) while TEA and Ba, inhibitors of BK and other voltage-gated K-

channels, respectively, were ineffective. Furthermore, after hyposmotic

incubation, RVD as detected by water content changes was abrogated by

Page 172: Ion Transport Mechanisms during Hyposmotic Regulatory and

161

addition of CTZ to the hyposmotic medium pointing towards the involvement of IK

channels in RVD (Figure 25 A, B). MQAE fluorescence was increased as

mentioned earlier due to [Cl]i dilution during hyposmotic stress (see section 5.7).

Presence of CTZ did not significantly change MQAE fluorescence during

hyposmotic stress (Figure 26). Also, addition of CTZ did not change MQAE

fluorescence during hyposmotic incubation, indicating that CTZ did not have an

inhibitory effect on Cl efflux. However, this cannot be definitively stated due to the

dilution of MQAE during RVD. Therefore to summarize Figures 23-26, CTZ

inhibited IK channels during RVD by affecting K and water efflux from the cell.

Thus despite the presence of a variety of Ca-activated K channels as detected by

RT-PCR and immunochemistry (Figure 28), it appears that both the FHL124

primary cell line as well as the SV40-transformed B3 lens epithelial cell utilize the

same RVD mechanism, i.e. primarily IK channels in conjunction with yet to be

further defined Cl channels that are inhibited by the “selective” KCC inhibitor

DIOA in FHL124 cells (Lauf et al., 2008). The presence of KCC, an important

efflux pathway for K and Cl, is demonstrated by immunofluorescence staining of

HLE-B3 cells (Figure 27). While there are several reports showing beyond doubt

the participation of IK channels in RVD in human intestinal cells (Wang et al.,

2003) and in T-lymphocytes (Khanna et al., 1999), other more classic cell models

such as Ehrlich-ascites tumor cells respond to hyposmotic challenge with

activation of inward rectifying K channels (IKvol), most likely TASK-2 channels,

Page 173: Ion Transport Mechanisms during Hyposmotic Regulatory and

162

and their function is governed by metabolic products deriving from membrane

lipids (reviewed in Hoffmann et al., 2009).

5.8. RNAi studies

As additional evidence for the involvement of IK channels during RVD, Rb

influx was measured in HLE-B3 cells transfected with siRNA designed against IK

channels. There are physiological studies in which down regulation of the IK

channel gene by RNAi inhibited cell proliferation in rat mesenchymal cells (Tao

et al., 2008; Deng et al., 2007). However, there are no known published studies

that investigate the role of IK channels during RVD. As seen in Figure 29 and

Table 10, transfection of IK channel siRNA in HLE-B3 cells failed to produce a

significant decrease in IK channel gene expression, as quantified by qPCR

studies. Since the gene was not down regulated at the mRNA level, at the

functional level Rb influx in siRNA-transfected cells remained unchanged during

hyposmotic stress (Figure 29B).

5.9. Ion channels involved during AVD

The present study was not designed to further detail the regulation of IK

channels in HLE-B3 cells but rather to determine whether the IK channel,

obviously functioning during RVD could be implicated in AVD after programmed

cell death was initiated by a pro-apoptotic agent like STP. Several K channels,

including Ca-dependent K channels, have been shown to play an important role

in cell shrinkage during apoptosis (Elliott and Higgins, 2003; Lang P. et al., 2003);

however, the specific channel involvement depends on the cell type and

Page 174: Ion Transport Mechanisms during Hyposmotic Regulatory and

163

apoptotic stimulus (Bortner and Cidlowski, 2007). The role of K efflux was

confirmed by prevention of apoptosis by high extracellular [K]o (Figure 30)

indicating involvement of K channels or K transporters. In FHL124 cells, Ko

blocks K loss, but not external Rb or Cs, or Na, suggesting that the CTZ-inhibited

K channel has a high PK/Pcation ratio, and hence is most probably an IK channel

(Lauf et al., 2008). In HLE-B3 cells, however, the functional and pharmacological

attempts to block Ki (Figure 31, 32) and water loss (Figure 33) by inhibition of IK

channels, and thus reduce apoptosis (Figure 35), failed. There are at least two

explanations for these findings that need be explored in the future. First, HLE-B3

cells are SV40-transformed cells (Andley et al., 1994) and hence the levels of

phosphorylated p53 are reduced (Levine A. 2009). Studies have revealed that

transporters like the taurine transporter (TauT) are transcriptional targets of p53

(Han et al., 2006), but it is not known what absence or reduction of p53, a tumor

suppressor protein, would do to ion transport through K channels. Second, the

action of CTZ could have been altered by the application of STP, a well-known

non-selective protein kinase inhibitor (Ruegg and Burgess 1989). CTZ exerts its

effect on ion channels in a complex manner. The CTZ binding site on the IK

channels is unknown, and it is known that CTZ binds to cytosolic p450, i.e. it

works from the inside of the cell (Wulff et al, 2001). There are also cytotoxic

effects of CTZ that can induce apoptosis by itself (Ito et al., 2002).

Page 175: Ion Transport Mechanisms during Hyposmotic Regulatory and

164

5.10. Role of 4-AP- sensitive channel in RVD and AVD

While voltage-gated K channels sensitive to 4-AP have been shown to

mediate apoptosis in model systems like corneal epithelial cells (Wang et al.,

2003), 4-AP as well as TEA prevent apoptosis in neurons (Zhao et al., 2006). In

COS-7 cells overexpressing the voltage-gated K channel KCNA5, inhibition of

these K channels by 4-AP significantly reduced STP-induced apoptosis

(Brevnova et al., 2004). We observed that addition of Ba as well as the BK

channel inhibitor TEA did not prevent the increased Rb uptake after addition of

STP (Figure 36B, C). However, addition of 2 mM 4-AP to STP-treated cells

significantly reduced the increased Rb uptake caused by addition of STP alone in

the medium (Figure 36A).

In various cell culture models like the corneal epithelial cells, studies

demonstrate that 1 mM 4-AP incubation abrogates the RVD (Capo-Aponte et al.,

2006). However, to provide for the involvement of a 4-AP sensitive channel

during RVD in LEC, if one can draw parallels in the RVD behavior between

FHL124 and HLE-B3 cells, this important finding sets AVD apart from RVD

because previous studies published in FHL124 cells show that 4-AP did not

interfere with RVD (Lauf et al., 2008) and thus ruled out a major contribution of

voltage-gated type-A K channels in hyposmotic RVD. Even though 4-AP by itself

produced an increase in DNA fragmentation as compared to control-treated cells,

it was significantly lower than in cells treated with STP alone (Figure 37). The

inhibitory 4-AP effect on Rb uptake in STP-treated HLE-B3 cells effect was

Page 176: Ion Transport Mechanisms during Hyposmotic Regulatory and

165

paralleled by the significant decrease in the amount of DNA fragmentation in cells

treated with STP and 4-AP as compared to cells treated with STP alone (Figure

37), suggesting that A-type voltage-gated K channels are involved in AVD

conducive to subsequent apoptosis of HLE-B3 cells. Although 4-AP and TEA

have similar mechanisms of K channel blockage, the reason why only the former

is involved can be speculated to be that STP is a general kinase inhibitor and

probably inactivates the kinases required by TEA to inhibit the delayed rectifying

K channels, whereas such a presumable kinase inhibition is not required by 4-AP

to produce its action.

5.11. Effect of prolonged stress on IK channel gene expression

IK channels are stress-activated channels and are transcriptionally

upregulated in endothelial cells in response to shear stress after 24 h

(Brakemeier et al., 2003). It is known that several osmoregulatory genes have

altered transcription in response to long term cell volume perturbations,

especially proteins such as tonicity-responsive enhancer binding protein

(TonEBP) and transport proteins like aquaporins, NKCC1, and the Na/K pump

(Hoffman et al., 2009). However the transcriptional adaptation of the IK channel

gene to long term osmotic and apoptotic stress is unknown. We observed 88%

decrease in IK channel gene expression in response to hyposmotic stress for 24

h and 90% decrease in response to STP treatment for over 3 h (Figure 38, 39).

Preliminary RT-PCR data suggest that neither the bulk of the non-IK channels

shown in Figure 28A, nor the KCC isforms 1,3 and 4 present in HLE-B3 cells

Page 177: Ion Transport Mechanisms during Hyposmotic Regulatory and

166

(Figure 27, Misri et al., 2006) were affected. These data suggest that, certainly

during RVD, there is an early functional activation of IK channels leading to K and

water loss which is followed by transcriptional deactivation of IK channels on

prolonged stress,perhaps a compensatory mechanism elicited by the cell to

prevent further K loss and adapt to the new hyposmotic environment. This

interesting possibility reminds of adaptation of Hela and chick embryo cell

cultures to prolonged low concentrations of ouabain (>24 h) leading to down

regulation of Na/K pumps in terms of fewer free pump sites (Aiton et al., 1981).

The finding that IK channel expression is also down regulated in cells exposed to

STP warrants further studies, since at 3 h, there is a significant loss of cells

(Figure 18) and significant DNA fragmentation (Figure 17B), which in turn can

negatively affect the transcriptional properties of HLE-B3 cells. Interestingly,

incubating cultured cortical neuronal cells for 15 h in STP containing media,

significantly reduced gene expressions of 4-AP sensitive inwardly rectifying

voltage-gated K channels as well as of Na/K pumps as compared to untreated

control (Chen et al., 2008).

5.12. Membrane changes during AVD

Apoptosis is characterized by cell shrinkage, translocation of

phosphatidylserine (PS) to the cell surface, caspase activation and ultimately

DNA fragmentation and cell breakdown (Elliott and Higgins, 2002). The exposure

of PS on the outer leaflet of the plasma membrane is a known crucial marker for

cells undergoing apoptosis to facilitate recognition, engulfment and degradation

Page 178: Ion Transport Mechanisms during Hyposmotic Regulatory and

167

by phagocytes (Savil J and Fadok V, 2000). In normal cells, PS is present on the

inner leaflet of the cell membrane. During cell injury or cell death, the PS

translocation to the outer leaflet, leading to a loss of membrane symmetry, is

mediated by phospholipid scramblases, in the presence of cytosolic Ca (Lang et

al., 2004, Sims P and Wiedmer T, 2000). It is known that externalization of PS

during apoptosis precedes caspase activation and DNA fragmentation by several

hours during the apoptotic cascade and can be detected by annexin binding in

the presence of cytosolic Ca (Martin et al., 1995). Translocation of PS,

determined by positive annexin staining was observed at 3 h in primary cultures

of bovine lens epithelial cells treated with STP (Andersson et al., 2000) and at 4 h

in SRA 01-04 human lens epithelial cells exposed to UVB irradiation (Long et al.,

2004), consistent with our findings of positive membrane labeling at 2 h (Figure

40). However, these studies did not examine PS exposure at earlier time points.

Annexin V binding was observed in HLE-B3 cells as early as 15 min (Figure 41

B) and distinct membrane labeling was observed at 35 min (Figure 41D)

consistent with previously published reports of significant number of annexin

positive CD4+ lymphocytes 30 min after treatment with calcimycin (Elliott and

Higgins , 2002). Given the diffusional rate of 0.3 x 10-9 cm2/sec for PS within the

plasma membrane (el Hage Chahine et al.,1993), PS externalization can be

estimated to be complete within minutes and hence it can be speculated to

precede the major loss of intracellular water, Ki, and possibly [Cl]i . Thus

membrane property changes are temporally well in advance of caspase

Page 179: Ion Transport Mechanisms during Hyposmotic Regulatory and

168

activation and associated DNA fragmentation, paving the way for increased K

and water permeability and for AVD.

5.13. Caspase independence of AVD

Caspase independent AVD was described in earlier studies on U937 cells

where administration of the broad-spectrum caspase inhibitor Z-VAD-FMK

prevented cell death and DNA laddering, but not AVD (Maeno et al., 2000). To

test whether PS externalization and therefore changes in the cell membrane

structure occurs prior to caspase activation, the number of annexin-labeled cells

was counted after pre-incubation with a broad spectrum caspase inhibitor (Q-VD-

OPh NM) for 30 min prior to STP treatment (Figure 42). No significant

quantitative change occurred in this parameter, supporting our hypothesis that

the early positive annexin labeling is independent of caspase activation and most

likely precedes and occurs as a result of change in membrane permeability.

Similar studies in bovine lens epithelial cells pre-incubated with broad-spectrum

caspase inhibitors demonstrate PS translocation by treatment with STP in a

caspase-independent manner (Andersson et al., 2000).

5.14. Apoptosis vs. necrosis in STP- treated cells

In contrast to the characteristics of apoptotic cell death described earlier,

necrotic cell death is characterized by cell swelling followed by rupture of plasma

membrane, leading to increased permeability of the cell plasma membrane. Cell

membrane permeability was demonstrated by uptake of propidium iodide (PI) in

STP-treated cells as a function of time, which in turn stains nucleic acids. HLE-B3

Page 180: Ion Transport Mechanisms during Hyposmotic Regulatory and

169

cell nuclei stained positive for PI as early as 35 min (Figure 41D) indicating an

increased permeability. At the same time points, the cell cytoplasm stained

positive for annexin, which suggests an increased membrane permeability

causing annexin to enter the cell and bind to the internal cytoplasmic PS along

with the externalized PS (Figure 41 D, arrows). These observations suggest the

occurrence of necrosis along with apoptosis in HLE-B3 cells after STP treatment.

As seen in Figure 17B, the amount of fragmented DNA after incubation with STP,

quantified by Cell Death ELISA, decreased significantly at 4 h as compared to

values at 3 h, which can be attributed to a mixed apoptotic and necrotic

population in cells. During quantification of apoptosis by Cell Death ELISA, the

apoptotic and necrotic populations are separated and hence at 4 h time point, the

measured amount of fragmented DNA is lowered, as the necrotic cell population

is not measured. Similar findings of PI -positive cells were reported in studies on

SV-40 transformed SRA 01-04 LEC where cell death due to oxidative stress was

induced by treatment with H2O2 and t-butyl hydroperoxide (Long et al., 2004).

However, our study demonstrates the occurrence of AVD mediated by a 4-AP-

sensitive K channel after STP treatment by K and water loss, and DNA

fragmentation (detected by Cell Death ELISA), a hallmark for apoptotic cell death.

The positive annexin and PI staining of the cells, in addition to the functional

studies suggest a “mixed population” of apoptotic and necrotic cells after STP

treatment, with apoptosis being the dominant cause of cell death. The presence

of this mixed cell population could be the reason why a distinct DNA ladder was

Page 181: Ion Transport Mechanisms during Hyposmotic Regulatory and

170

not observed during the DNA fragmentation assay, whereas DNA fragmentation

was quantified by cell death ELISA, which separates the apoptotic and necrotic

cell populations. In addition, although 4-AP significantly inhibited Rb influx, the

drug did not completely prevent the STP mediated activation of Rb influx, further

supporting the concept of cell-heterogeneity.

Page 182: Ion Transport Mechanisms during Hyposmotic Regulatory and

171

VI. Summary

In summary, this study demonstrates that during hyposmotically elicited RVD,

loss of K, water and possibly Cl were early events mediated by IK channels,

without obvious major participation by other Ca-activated K channels present in

HLE-B3 cells. However, during AVD, the efflux rates of K and water were about

an order of magnitude slower in the early phases of AVD following a major pre-

apoptotic stimulus, such as application of STP. The sluggish loss of water and K

during the first 30 min of AVD appears to be preceded by an early change of

membrane phospholipids presumably due to PS externalization which, in

conjunction with a general STP-induced dephosphorylation, probably triggers 4-

AP-sensitive voltage-gated K channels prior to caspase activation leading to

subsequent DNA fragmentation and cell death. This mechanism of AVD is

summarized in Figure 43. Only at later times, i.e. after 1 h, K and water loss were

more significant before further cellular deterioration occurred and such

measurements became less precise. These findings can .provide insight into the

mechanism of apoptosis of LEC, responsible for the pathology of cataract

formation in the human lens.

Page 183: Ion Transport Mechanisms during Hyposmotic Regulatory and

172

Figure 43: Proposed scheme of events during AVD

STP inhibits kinases, especially the serine threonine kinases (1), which are

dephosphorylated and in turn, possibly randomize phospholipid symmetry

through activation of scramblases (2) causing externalization of PS from the inner

cell surface (3,4) (5). The resulting membrane lipid asymmetry causes activation

of 4-AP-sensitive voltage-gated K (Kv) channels (6A), which are also activated by

depolarization (6B), and lead to K efflux from the cell (7,8). The loss of

intracellular K (8) is followed by activation of effector caspases (9), DNA

fragmentation (10) and cell death.

Page 184: Ion Transport Mechanisms during Hyposmotic Regulatory and

173

Figure 43

Page 185: Ion Transport Mechanisms during Hyposmotic Regulatory and

174

VII. References: Adragna, N. (1991). Cation transport in vascular endothelial cells and aging. The Journal of membrane biology, 124(3), 285-91. Adragna, N., White, R., Orlov, S., & Lauf, P. (2000). K-Cl cotransport in vascular smooth muscle and erythrocytes: possible implication in vasodilation. American Journal of Physiology: Cell Physiology, 278(2), C381-90. Aiton, J., Lamb, J., & Ogden, P. (1981). Down-regulation of the sodium pump following chronic exposure of HeLa cells and chick embryo heart cells to ouabain. British Journal of Pharmacology, 73(2), 333-40. Andersson, M., Sjöstrand, J., Petersen, A., Honarvar, A., & Karlsson, J. (2000). Caspase and proteasome activity during staurosporin-induced apoptosis in lens epithelial cells. Investigative Ophthalmology and Visual Science, 41(9), 2623-32. Andersson, M., Honarvar, A., Sjöstrand, J., Peterson, A., & Karlsson, J. (2003). Decreased caspase-3 activity in human lens epithelium from posterior subcapsular cataracts. Experimental Eye Research, 76(2), 175-82. Andley, U. (2008). The lens epithelium: focus on the expression and function of the alpha-crystallin chaperones. The International Journal of Biochemistry & Cell Biology, 40(3), 317-23. Andley, U., Song, Z., Wawrousek, E., Fleming, T., & Bassnett, S. (2000). Differential protective activity of alpha A- and alphaB-crystallin in lens epithelial cells. The Journal of biological chemistry, 275(47), 36823-31 Andley, U., Song, Z., Wawrousek, E., & Bassnett, S. (1998). The molecular chaperone alphaA-crystallin enhances lens epithelial cell growth and resistance to UVA stress. The Journal of biological chemistry, 273(47), 31252-61 Andley, U., Rhim, J., Chylack LT Jr,& Fleming, T. (1994). Propagation and immortalization of human lens epithelial cells in culture. Investigative Ophthalmology and Visual Science, 35(7), 3094-102. Arrebola, F., Zabiti, S., Cañizares, F., Cubero, M., Crespo, P. et al. (2005). Changes in intracellular sodium, chlorine, and potassium concentrations in staurosporine-induced apoptosis. Journal of Cellular Physiology, 204(2), 500-7.

Page 186: Ion Transport Mechanisms during Hyposmotic Regulatory and

175

Awasthi, N., & Wagner, B. (2004). Interferon-gamma induces apoptosis of lens alphaTN4-1 cells and proteasome inhibition has an antiapoptotic effect. Investigative Ophthalmology & Visual Science, 45(1), 222-9. Balasubramanian, D. (2000). Ultraviolet radiation and cataract. Journal of ocular pharmacology and therapeutics : the official journal of the Association for Ocular Pharmacology and Therapeutics, 16(3), 285-97 Beebe, D., Parmelee, J., & Belcher, K. (1990). Volume regulation in lens epithelial cells and differentiating lens fiber cells. Journal of Cellular Physiology, 143(3), 455-9. Begenisich, T., Nakamoto, T., Ovitt, C., Nehrke, K., Brugnara, C., Alper, S. et al. (2004). Physiological roles of the intermediate conductance, Ca2+-activated potassium channel Kcnn4. The Journal of biological chemistry, 279(46), 47681-7. Bhat, S. (2001). The ocular lens epithelium. Bioscience Reports, 21(4), 537-63 Bodero, A., Ye, R., & Lees-Miller, S. (2003). UV-light induces p38 MAPK-dependent phosphorylation of Bcl10. Biochemical and Biophysical Research Communications, 301(4), 923-6. Bortner, C., & Cidlowski, J. (1996). Absence of volume regulatory mechanisms contributes to the rapid activation of apoptosis in thymocytes. American Journal of Physiology, 271(3 Pt 1), C950-61. Bortner, C., & Cidlowski, J. (1998). A necessary role for cell shrinkage in apoptosis. Biochemical Pharmacology, 56(12), 1549-59. Bortner, C., & Cidlowski, J. (2000). Volume regulation and ion transport during apoptosis. Methods in Enzymology, 322, 421-33. Bortner, C., & Cidlowski, J. (2002). Apoptotic volume decrease and the incredible shrinking cell. Cell Death and Differentiation, 9(12), 1307-10. Bortner, C., & Cidlowski, J. (2003). Uncoupling cell shrinkage from apoptosis reveals that Na+ influx is required for volume loss during programmed cell death. The Journal of Biological Chemistry, 278(40), 39176-84. Bortner, C., & Cidlowski, J. (2004). The role of apoptotic volume decrease and ionic homeostasis in the activation and repression of apoptosis. Pflugers Archiv : European journal of physiology, 448(3), 313-8. Bortner, C., & Cidlowski, J. (2007). Cell shrinkage and monovalent cation fluxes: role in apoptosis. Archives of Biochemistry and Biophysics, 462(2), 176-88.

Page 187: Ion Transport Mechanisms during Hyposmotic Regulatory and

176

Bossy-Wetzel, E., Talantova, M., Lee, W., Schölzke, M., Harrop, A. et al. (2004). Crosstalk between nitric oxide and zinc pathways to neuronal cell death involving mitochondrial dysfunction and p38-activated K+ channels. Neuron, 41(3), 351-65. Brakemeier, S., Kersten, A., Eichler, I., Grgic, I., Zakrzewicz, A., Hopp, H. et al., (2003). Shear stress-induced up-regulation of the intermediate-conductance Ca(2+)-activated K(+) channel in human endothelium. Cardiovascular Research, 60(3), 488-96. Brevnova, E., Platoshyn, O., Zhang, S., & Yuan, J. (2004). Overexpression of human KCNA5 increases IK V and enhances apoptosis. American Journal of Physiology: Cell Physiology, 287(3), C715-22. Capo-Aponte, J., Iserovich, P., & Reinach, P. (2005). Characterization of regulatory volume behavior by fluorescence quenching in human corneal epithelial cells. The Journal of membrane biology, 207(1), 11-22. Candia, O. (2004). Electrolyte and fluid transport across corneal, conjunctival and lens epithelia. Experimental Eye Research, 78(3), 527-35. Caserta, T., Smith, A., Gultice, A., Reedy, M., & Brown, T. (2003). Q-VD-OPh, a broad spectrum caspase inhibitor with potent antiapoptotic properties. Apoptosis : an international journal on programmed cell death, 8(4), 345-52. Chambers, C., & Russell, P. (1993). Sequence of the human lens beta B2-crystallin-encoding cDNA. Gene (Amsterdam), 133(2), 295-9. Chen, J-M., Sepramaniam, S., Armugam, A., Shyan Choy, M., Manikandan, J., Melendez, A. et al. (2008). Water and ion channels: crucial in the initiation and progression of apoptosis in central nervous system? Current Neuropharmacology, 6(2), 102-16. Cidlowski, J., Bortner, C., Gomez-Angelats, M., Mann, C., Scoltock, A., Huang, J. et al. (2000). The cell and molecular biology of apoptosis in T-lymphocytes. Annales d'Endocrinologie, 61(2), 138. Cui, G., Dean, W., & Delamere, N. (2002). The influence of cycloheximide on Na,K-ATPase activity in cultured human lens epithelial cells. Investigative Ophthalmology & Visual Science, 43(8), 2714-20. Coca-Prados, M., Angíta, J., Chalfant, M., & Civan, M. (1995). PKC-sensitive Cl- channels associated with ciliary epithelial homologue of pICln. American Journal of Physiology, 268(3 Pt 1), C572-9.

Page 188: Ion Transport Mechanisms during Hyposmotic Regulatory and

177

Deng, X., Lau, C., Lai, K., Cheung, K., Lau, G., & Li, G. (2007). Cell cycle-dependent expression of potassium channels and cell proliferation in rat mesenchymal stem cells from bone marrow. Cell Proliferation, 40(5), 656-70. Eichler, I., Wibawa, J., Grgic, I., Knorr, A., Brakemeier, S., Pries, A. et al. (2003). Selective blockade of endothelial Ca2+-activated small- and intermediate-conductance K+-channels suppresses EDHF-mediated vasodilation. British Journal of Pharmacology, 138(4), 594-601. Ekhterae, D., Platoshyn, O., Zhang, S., Remillard, C., & Yuan, J. (2003). Apoptosis repressor with caspase domain inhibits cardiomyocyte apoptosis by reducing K+ currents. American Journal of Physiology: Cell Physiology, 284(6), C1405-10. el Hage Chahine, J., Cribier, S., & Devaux, P. (1993). Phospholipid transmembrane domains and lateral diffusion in fibroblasts. Proceedings of the National Academy of Sciences, 90(2), 447-51. Elliott, J., & Higgins, C. (2003). IKCa1 activity is required for cell shrinkage, phosphatidylserine translocation and death in T lymphocyte apoptosis. EMBO reports, 4(2), 189-94. Fischbarg, J. (2003). On the mechanism of fluid transport across corneal endothelium and epithelia in general. Journal of experimental zoology. Part A, Comparative experimental biology, 300(1), 30-40. Flynn, J., & Cammarata, P. (2006). Estradiol attenuates mitochondrial depolarization in polyol-stressed lens epithelial cells. Mol Vis, 12, 271-82. Franco, R., Bortner, C., & Cidlowski, J. (2006). Potential roles of electrogenic ion transport and plasma membrane depolarization in apoptosis. The Journal of membrane biology, 209(1), 43-58. Fujise, H., Higa, K., Kanemaru, T., Fukuda, M., Adragna, N., & Lauf, P. (2001). GSH depletion, K-Cl cotransport, and regulatory volume decrease in high-K/high-GSH dog red blood cells. American Journal of Physiology: Cell Physiology, 281(6), C2003-9. Futter, C., Crowston, J., & Allan, B. (2005). Interaction with collagen IV protects lens epithelial cells from Fas-dependent apoptosis by stimulating the production of soluble survival factors. Investigative Ophthalmology & Visual Science, 46(9), 3256-62.

Page 189: Ion Transport Mechanisms during Hyposmotic Regulatory and

178

Gagnon, K., Fyffe, R., Adragna, N., & Lauf, P. (2007). Characterization of an extracellular epitope antibody to the neuronal K-Cl cotransporter, KCC2. Clinical & Experimental Pharmacology & Physiology, 34(7), 566-73. Gasbjerg, P., & Brahm, J. (1991). Kinetics of bicarbonate and chloride transport in human red cell membranes. The Journal of general physiology, 97(2), 321-49. Grgic, I., Eichler, I., Heinau, P., Si, H., Brakemeier, S., Hoyer, J. et al. (2005). Selective blockade of the intermediate-conductance Ca2+-activated K+ channel suppresses proliferation of microvascular and macrovascular endothelial cells and angiogenesis in vivo. Arteriosclerosis, Thrombosis, and Vascular Biology, 25(4), 704-9. Grissmer, S., Nguyen, A., & Cahalan, M. (1993). Calcium-activated potassium channels in resting and activated human T lymphocytes. Expression levels, calcium dependence, ion selectivity, and pharmacology. The Journal of general physiology, 102(4), 601-30. Han, X., Patters, A., Jones, D., Zelikovic, I., & Chesney, R. (2006). The taurine transporter: mechanisms of regulation. Acta physiologica (Oxford, England), 187(1-2), 61-73. Heimlich,G.,Bortner,C., & Cidlowski, J (2004) Apoptosis and Cell Volume Regulation the importance of ions and ion channels . Chapter 18,Advances in Experimental Medicine and Biology,Ed: Lauf ,P.K., Adragna ,N.C.,Springer NY, Vol. 559,189-203 Heimlich, G., & Cidlowski, J. (2006). Selective role of intracellular chloride in the regulation of the intrinsic but not extrinsic pathway of apoptosis in Jurkat T-cells. The Journal of biological chemistry, 281(4), 2232-41. Hightower, K., Reddan, J., McCready, J., & Dziedzic, D. (1994). Lens epithelium: a primary target of UVB irradiation. Experimental Eye Research, 59(5), 557-64. Hoffmann, E., & Simonsen, L. (1989). Membrane mechanisms in volume and pH regulation in vertebrate cells. Physiological Reviews, 69(2), 315-82. Hoffmann, E., Schettino, T., & Marshall, W. (2007). The role of volume-sensitive ion transport systems in regulation of epithelial transport. Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology, 148(1), 29-43. Hoffmann, E., Lambert, I., & Pedersen, S. (2009). Physiology of cell volume regulation in vertebrates. Physiological Reviews, 89(1), 193-277.

Page 190: Ion Transport Mechanisms during Hyposmotic Regulatory and

179

Hughes FM Jr,, Bortner, C., Purdy, G., & Cidlowski, J. (1997). Intracellular K+ suppresses the activation of apoptosis in lymphocytes. The Journal of biological chemistry, 272(48), 30567-76. Inoue, H., & Okada, Y. (2007). Roles of volume-sensitive chloride channel in excitotoxic neuronal injury. Journal of Neuroscience, 27(6), 1445-55. Ito, C., Tecchio, C., Coustan-Smith, E., Suzuki, T., Behm, F., Raimondi, S. et al. (2002). The antifungal antibiotic clotrimazole alters calcium homeostasis of leukemic lymphoblasts and induces apoptosis. Leukemia : 16(7), 1344-52. Jablonski, E., Webb, A., McConnell, N., Riley, M., & Hughes FM Jr, . (2004). Plasma membrane aquaporin activity can affect the rate of apoptosis but is inhibited after apoptotic volume decrease. American journal of physiology.,Cell physiology, 286(4), C975-85. Jayaraman, S., & Verkman, A. (2000). Quenching mechanism of quinolinium-type chloride-sensitive fluorescent indicators. Biophysical Chemistry, 85(1), 49-57. Karaman, M., Herrgard, S., Treiber, D., Gallant, P., Atteridge, C., Campbell, B. et al. (2008). A quantitative analysis of kinase inhibitor selectivity. Nature Biotechnology, 26(1), 127-32. Khanna, R, Chang MC, Joiner WJ, Kaczmarek LK, and Schlichter LC. (1999) hSK4/hIK1, a calmodulin-binding KCa channel in human T lymphocytes. J Biol Chem 274, 14838-14849 Köhler, R., Wulff, H., Eichler, I., Kneifel, M., Neumann, D. et al. (2003). Blockade of the intermediate-conductance calcium-activated potassium channel as a new therapeutic strategy for restenosis. Circulation, 108(9), 1119-25. Koncz, C., & Daugirdas, J. (1994). Use of MQAE for measurement of intracellular [Cl-] in cultured aortic smooth muscle cells. American Journal of Physiology, 267(6 Pt 2), H2114-23. Krick, S., Platoshyn, O., McDaniel, S., Rubin, L., & Yuan, J. (2001). Augmented K+ currents and mitochondrial membrane depolarization in pulmonary artery myocyte apoptosis. American Journal of Physiology - Lung Cellular and Molecular Physiology, 281(4), L887-94.

Page 191: Ion Transport Mechanisms during Hyposmotic Regulatory and

180

Lai, Z., Chen, Y., & Nishi, K. (2003). Modulation of intracellular Cl- homeostasis by lectin-stimulation in Jurkat T lymphocytes. European Journal of Pharmacology, 482(1-3), 1-8. Lang, F., Gulbins, E., Szabo, I., Lepple-Wienhues, A., Huber, S., Duranton, C. et al. (2004). Cell volume and the regulation of apoptotic cell death. Journal of Molecular Recognition, 17(5), 473-80. Lang, P., Kaiser, S., Myssina, S., Wieder, T., Lang, F., & Huber, S. (2003). Role of Ca2+-activated K+ channels in human erythrocyte apoptosis. American Journal of Physiology: Cell Physiology, 285(6), C1553-60. Langford, M., Chen, D., Gosslee, J., Misra, R., Redens, T., & Texada, D. (2006). Intracameral toxicity of bacterial components muramyl dipeptide and staurosporine: ciliary cyst formation, epithelial cell apoptosis and necrosis. Cutaneous and Ocular Toxicology, 25(2), 85-101. Lauf, P., Zhang, J., Delpire, E., Fyffe, R., Mount, D., & Adragna, N. (2001). K-Cl co-transport: immunocytochemical and functional evidence for more than one KCC isoform in high K and low K sheep erythrocytes. Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology, 130(3), 499-509. Lauf, P., Warwar, R., Brown, T., & Adragna, N. (2006). Regulation of potassium transport in human lens epithelial cells. Experimental Eye Research, 82(1), 55-64. Lauf, P., Misri, S., Chimote, A., & Adragna, N. (2008). Apparent intermediate K conductance channel hyposmotic activation in human lens epithelial cells. American Journal of Physiology: Cell Physiology, 294(3), C820-32. Lee, J., Wan, X., Song, J., Kang, J., Chung, W., Lee, E. et al. (2002). TGF-beta-induced apoptosis and reduction of Bcl-2 in human lens epithelial cells in vitro. Current Eye Research, 25(3), 147-53. Levine, A. J. (2009). The common mechanisms of transformation by the small DNA tumor viruses: The inactivation of tumor suppressor gene products: p53. Virology, 384(2), 285-293. Li, W., Kuszak, J., Dunn, K., Wang, R., Ma, W., Wang, G. et al. (1995). Lens epithelial cell apoptosis appears to be a common cellular basis for non-congenital cataract development in humans and animals. The Journal of cell biology, 130(1), 169-81. Li, W., & Spector, A. (1996). Lens epithelial cell apoptosis is an early event in the

Page 192: Ion Transport Mechanisms during Hyposmotic Regulatory and

181

development of UVB-induced cataract. Free Radical Biology and Medicine, 20(3), 301-11. Liyanage, N., Fernando, M., & Lou, M. (2007). Regulation of the bioavailability of thioredoxin in the lens by a specific thioredoxin-binding protein (TBP-2). Experimental Eye Research, 85(2), 270-9. Long, A., Colitz, C., & Bomser, J. (2004). Apoptotic and necrotic mechanisms of stress-induced human lens epithelial cell death. Experimental biology and medicine (Maywood, N.J.), 229(10), 1072-80. Lou, M. (2003). Redox regulation in the lens. Progress in Retinal and Eye Research, 22(5), 657-82. Lu, L., Wang, L., & Shell, B. (2003). UV-induced signaling pathways associated with corneal epithelial cell apoptosis. Investigative Ophthalmology & Visual Science, 44(12), 5102-9. Maeno, E., Ishizaki, Y., Kanaseki, T., Hazama, A., & Okada, Y. (2000). Normotonic cell shrinkage because of disordered volume regulation is an early prerequisite to apoptosis. Proceedings of the National Academy of Sciences, 97(17), 9487-92. Maeno, E., Takahashi, N., & Okada, Y. (2006). Dysfunction of regulatory volume increase is a key component of apoptosis. Febs Letters, 580(27), 6513-7. Martin, S., Reutelingsperger, C., McGahon, A., Rader, J., van Schie, R., LaFace, D. et al., (1995). Early redistribution of plasma membrane phosphatidylserine is a general feature of apoptosis regardless of the initiating stimulus: inhibition by overexpression of Bcl-2 and Abl. The Journal of experimental medicine, 182(5), 1545-56. Maruno, K., Lovicu, F., Chamberlain, C., & McAvoy, J. (2002). Apoptosis is a feature of TGF beta-induced cataract. Clinical & experimental optometry : journal of the Australian Optometrical Association, 85(2), 76-82 Mathias, R., Kistler, J., & Donaldson, P. (2007). The lens circulation. The Journal of membrane biology, 216(1), 1-16. Mathias, R., & Rae, J. (2004). The lens: local transport and global transparency. Experimental Eye Research, 78(3), 689-98. Misri, S., Chimote, A., Adragna, N., Warwar, R., Brown, T., & Lauf, P. (2006). KCC isoforms in a human lens epithelial cell line (B3) and lens tissue extracts. Experimental Eye Research, 83(5), 1287-94.

Page 193: Ion Transport Mechanisms during Hyposmotic Regulatory and

182

Miyazaki, H., Shiozaki, A., Niisato, N., & Marunaka, Y. (2007). Physiological significance of hypotonicity-induced regulatory volume decrease: reduction in intracellular Cl- concentration acting as an intracellular signaling. American Journal of Physiology - Renal Physiology, 292(5), F1411-7. Mongin, A., & Orlov, S. (2001). Mechanisms of cell volume regulation and possible nature of the cell volume sensor. Pathophysiology : the official journal of the International Society for Pathophysiology / ISP, 8(2), 77-88. Nietsch, H., Roe, M., Fiekers, J., Moore, A., & Lidofsky, S. (2000). Activation of potassium and chloride channels by tumor necrosis factor alpha. Role in liver cell death. The Journal of biological chemistry, 275(27), 20556-61. Nobel, C., Aronson, J., van den Dobbelsteen, D., & Slater, A. (2000). Inhibition of Na+/K(+)-ATPase may be one mechanism contributing to potassium efflux and cell shrinkage in CD95-induced apoptosis. Apoptosis : an international journal on programmed cell death, 5(2), 153-63. Numata, T., Sato, K., Okada, Y., & Wehner, F. (2008). Hypertonicity-induced cation channels rescue cells from staurosporine-elicited apoptosis. Apoptosis : an international journal on programmed cell death, 13(7), 895-903. Okada, Y. (2004). Ion channels and transporters involved in cell volume regulation and sensor mechanisms. Cell Biochemistry and Biophysics, 41(2), 233-58. Okada, Y., & Maeno, E. (2001). Apoptosis, cell volume regulation and volume-regulatory chloride channels. Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology, 130(3), 377-83. Okada, Y., Maeno, E., Shimizu, T., Dezaki, K., Wang, J., & Morishima, S. (2001). Receptor-mediated control of regulatory volume decrease (RVD) and apoptotic volume decrease (AVD). The Journal of Physiology, 532(Pt 1), 3-16. Orlov, S., Pchejetski, D., Taurin, S., Thorin-Trescases, N., Maximov, G., Pshezhetsky, A. et al., (2004). Apoptosis in serum-deprived vascular smooth muscle cells: evidence for cell volume-independent mechanism. Apoptosis, 9(1), 55-66. Orlov, S., Thorin-Trescases, N., Kotelevtsev, S., Tremblay, J., & Hamet, P. (1999). Inversion of the intracellular Na+/K+ ratio blocks apoptosis in vascular smooth muscle at a site upstream of caspase-3. The Journal of biological chemistry, 274(23), 16545-52.

Page 194: Ion Transport Mechanisms during Hyposmotic Regulatory and

183

Oya-Ito, T., Liu, B., & Nagaraj, R. (2006). Effect of methylglyoxal modification and phosphorylation on the chaperone and anti-apoptotic properties of heat shock protein 27. Journal of Cellular Biochemistry, 99(1), 279-91. Pilas, B., & Durack, G. (1997). A flow cytometric method for measurement of intracellular chloride concentration in lymphocytes using the halide-specific probe 6-methoxy-N-(3-sulfopropyl) quinolinium (SPQ). Cytometry, 28(4), 316-22. Porcelli, A., Ghelli, A., Zanna, C., Valente, P., Ferroni, S., & Rugolo, M. (2004). Apoptosis induced by staurosporine in ECV304 cells requires cell shrinkage and upregulation of Cl- conductance. Cell Death & Differentiation, 11(6), 655-62. Rhodes, J., Collison, D., & Duncan, G. (2003). Calcium activates SK channels in the intact human lens. Investigative Ophthalmology and Visual Science, 44(9), 3927-32. Roucou, X., Antonsson, B., & Martinou, J. (2001). Involvement of mitochondria in apoptosis. Cardiology Clinics, 19(1), 45-55. Rüegg, U., & Burgess, G. (1989). Staurosporine, K-252 and UCN-01: potent but nonspecific inhibitors of protein kinases. Trends in Pharmacological Sciences, 10(6), 218-20. Savill, J., & Fadok, V. (2000). Corpse clearance defines the meaning of cell death. Nature (London), 407(6805), 784-8. Shimizu, T., Maeno, E., & Okada, Y. (2007). Prerequisite role of persistent cell shrinkage in apoptosis of human epithelial cells. Sheng li xue bao : [Acta physiologica Sinica], 59(4), 512-6. Shimizu, T., Wehner, F., & Okada, Y. (2006). Inhibition of hypertonicity-induced cation channels sensitizes HeLa cells to shrinkage-induced apoptosis. Cellular Physiology & Biochemistry, 18(6), 295-302. Shiraishi, J., Tatsumi, T., Keira, N., Akashi, K., Mano, A., Yamanaka, S. et al. (2001). Important role of energy-dependent mitochondrial pathways in cultured rat cardiac myocyte apoptosis. American Journal of Physiology: Heart & Circulatory Physiology, 281(4), H1637-47. Sims, P., & Wiedmer, T. (2001). Unraveling the mysteries of phospholipid scrambling. Thrombosis and Haemostasis, 86(1), 266-75.

Page 195: Ion Transport Mechanisms during Hyposmotic Regulatory and

184

Small, D., Tauskela, J., & Xia, Z. (2002). Role for chloride but not potassium channels in apoptosis in primary rat cortical cultures. Neuroscience Letters, 334(2), 95-8. Stewart, S., Duncan, G., Marcantonio, J., & Prescott, A. (1988). Membrane and communication properties of tissue cultured human lens epithelial cells. Investigative Ophthalmology and Visual Science, 29(11), 1713-25. Szabo, I., Lepple-Wienhues, A., Kaba, K., Zoratti, M., Gulbins, E., & Lang, F. (1998). Tyrosine kinase-dependent activation of a chloride channel in CD95-induced apoptosis in T lymphocytes. Proceedings of the National Academy of Sciences, 95(11), 6169-74. Tao, R., Lau, C., Tse, H., & Li, G. (2008). Regulation of cell proliferation by intermediate-conductance Ca2+-activated potassium and volume-sensitive chloride channels in mouse mesenchymal stem cells. American Journal of Physiology: Cell Physiology, 295(5), C1409-16. Thompson, G., Langlais, C., Cain, K., Conley, E., & Cohen, G. (2001). Elevated extracellular [K+] inhibits death-receptor- and chemical-mediated apoptosis prior to caspase activation and cytochrome c release. Biochemical Journal, 357(Pt 1), 137-45 Thuret, G., Chiquet, C., Herrag, S., Dumollard, J., Boudard, D., Bednarz, J. et al. (2003). Mechanisms of staurosporine induced apoptosis in a human corneal endothelial cell line. British Journal of Ophthalmology, 87(3), 346-52. Vázquez, E., Nobles, M., & Valverde, M. (2001). Defective regulatory volume decrease in human cystic fibrosis tracheal cells because of altered regulation of intermediate conductance Ca2+-dependent potassium channels. Proceedings of the National Academy of Sciences, 98(9), 5329-34. Verkman, A., Sellers, M., Chao, A., Leung, T., & Ketcham, R. (1989). Synthesis and characterization of improved chloride-sensitive fluorescent indicators for biological applications. Analytical Biochemistry, 178(2), 355-61. Vermes, I., Haanen, C., Steffens-Nakken, H., & Reutelingsperger, C. (1995). A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V. Journal of Immunological Methods, 184(1), 39-51. Wang, J., Morishima, S., & Okada, Y. (2003). IK channels are involved in the regulatory volume decrease in human epithelial cells. American Journal of Physiology: Cell Physiology, 284(1), C77-84.

Page 196: Ion Transport Mechanisms during Hyposmotic Regulatory and

185

Wang, X., Garcia, C., Shui, Y., & Beebe, D. (2004). Expression and regulation of alpha-, beta-, and gamma-crystallins in mammalian lens epithelial cells. Investigative Ophthalmology and Visual Science, 45(10), 3608-19. Wang, L., Chen, L., Zhu, L., Rawle, M., Nie, S., Zhang, J. et al. (2002). Regulatory volume decrease is actively modulated during the cell cycle. Journal of Cellular Physiology, 193(1), 110-9. Wang, L., Chen, L., & Jacob, T. (2000). The role of ClC-3 in volume-activated chloride currents and volume regulation in bovine epithelial cells demonstrated by antisense inhibition. The Journal of Physiology, 524 Pt 1, 63-75. Wang, L., Xu, D., Dai, W., & Lu, L. (1999). An ultraviolet-activated K+ channel mediates apoptosis of myeloblastic leukemia cells. The Journal of biological chemistry, 274(6), 3678-85. Wang, L., & Lu, L. (2007). Pathway-specific effect of caffeine on protection against UV irradiation-induced apoptosis in corneal epithelial cells. Investigative Ophthalmology and Visual Science, 48(2), 652-60. Wang, L., Li, T., & Lu, L. (2003). UV-induced corneal epithelial cell death by activation of potassium channels. Investigative Ophthalmology and Visual Science, 44(12), 5095-101. Wang, L., Dai, W., & Lu, L. (2005). Ultraviolet irradiation-induced K(+) channel activity involving p53 activation in corneal epithelial cells. Oncogene, 24(18), 3020-7. Wehner, F. (2006). Cell volume-regulated cation channels. Contributions to Nephrology, 152, 25-53. Wei, L., Xiao, A., Jin, C., Yang, A., Lu, Z., & Yu, S. (2004). Effects of chloride and potassium channel blockers on apoptotic cell shrinkage and apoptosis in cortical neurons. Pflugers Archiv : European journal of physiology, 448(3), 325-34. Wei, A., Gutman, G., Aldrich, R., Chandy, K., Grissmer, S., & Wulff, H. (2005). International Union of Pharmacology. LII. Nomenclature and molecular relationships of calcium-activated potassium channels. Pharmacological Reviews, 57(4), 463-72. Weskamp, M., Seidl, W., & Grissmer, S. (2000). Characterization of the increase in [Ca2+]i during hypotonic shock and the involvement of Ca2+-activated

Page 197: Ion Transport Mechanisms during Hyposmotic Regulatory and

186

K+channels in the regulatory volume decrease in human osteoblast-like cells. The Journal of membrane biology, 178(1), 11-20. West, M., & Molloy, C. (1996). A microplate assay measuring chloride ion channel activity. Analytical Biochemistry, 241(1), 51-8. Wulff, H., Gutman, G., Cahalan, M., & Chandy, K. (2001). Delineation of the clotrimazole/TRAM-34 binding site on the intermediate conductance calcium-activated potassium channel, IKCa1. The Journal of biological chemistry, 276(34), 32040-5. Wyllie, A., Kerr, J., & Currie, A. (1980). Cell death: the significance of apoptosis. International Review of Cytology, 68, 251-306. Yu, S., Canzoniero, L., & Choi, D. (2001). Ion homeostasis and apoptosis. Current Opinion in Cell Biology, 13(4), 405-11. Yu, H., Li, Z., Zhang, H., & Wang, X. (2006). Role of potassium channels in Abeta(1-40)-activated apoptotic pathway in cultured cortical neurons. Journal of Neuroscience Research, 84(7), 1475-84. Yue, T., Wang, C., Romanic, A., Kikly, K., Keller, P., DeWolf WE Jr, . et al. (1998). Staurosporine-induced apoptosis in cardiomyocytes: A potential role of caspase-3. Journal of Molecular and Cellular Cardiology, 30(3), 495-507. Yurinskaya, V., Goryachaya, T., Guzhova, I., Moshkov, A., Rozanov, Y., Sakuta, G. et al. (2005). Potassium and sodium balance in U937 cells during apoptosis with and without cell shrinkage. Cellular Physiology & Biochemistry, 16(4-6), 155-62. Zhang, J., Misri, S., Adragna, N., Gagnon, K., Fyffe, R., & Lauf, P. (2005). Cloning and expression of sheep renal K-CI cotransporter-1. Cellular Physiology & Biochemistry, 16(1-3), 87-98. Zhang, J., & Jacob, T. (1997). The role of chloride in the lens of the eye. Experimental Physiology, 82(2), 245-59. Zhao, Y., Sun, L., Zhou, H., & Wang, X. (2006). Voltage-dependent potassium channels are involved in glutamate-induced apoptosis of rat hippocampal neurons. Neuroscience Letters, 398(1-2), 22-7.

Page 198: Ion Transport Mechanisms during Hyposmotic Regulatory and

187

VIII. Appendix : List of Abbreviations Abbreviation Word

[Cl]i Intracellular chloride concentration

[Cl]o Extracellular chloride concentration

[K]i Intracellular potassium concentration

[K]o Extracellular potassium concentration

4-AP 4-Aminopyridine

Apaf-1 Apoptotic peptidase activating factor 1

AVD Apoptotic volume decrease

Ba Barium

BCA Bicinchoninic acid assay

BID BH3 interacting domain death agonist

BK Large conductance K channels

BSA Bovine serum albumin

BSS Balanced salt solution

Ca Calcium

CAD Caspase-activated DNAse

Cl Chloride

Cl/HCO3 Chloride-bicarbonate exchanger

CLC-2 Chloride channel -2

CLC-3 Chloride channel-3

Cli Intracellular chloride content

CsCl Cesium chloride

Page 199: Ion Transport Mechanisms during Hyposmotic Regulatory and

188

Abbreviation Word

CTD C-terminus domain

CTZ Clotrimazole

DAPI 4',6-diamidino-2-phenylindole

DIDS 4,4'-diisothiocyano-2,2'-stillbene-disulfonic acid

DIOA [(dihydronindenyl)oxy] alkanoic acid

DMSO Dimethyl sulfoxide (DMSO)

DWT Dry weight

ECL Extracellular loop

ECl Nernst potential for chloride

Em Membrane potential

FBS Fetal bovine serum

GAPDH Glyceraldehyde 3-phosphate dehydrogenase

GSH Glutathione

h Hours

H2O2 Hydrogen peroxide

HCE Human corneal epithelium

HEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

HLE Human lens epithelium

HRP Horseradish peroxidase

IK Intermediate conductance calcium activated K channel

IKVOL Swelling-induced K current

JNK c-Jun N-terminal kinase

K Potassium

Ki Intracellular potassium content

Page 200: Ion Transport Mechanisms during Hyposmotic Regulatory and

189

Abbreviation Word

KCl Stern-volmer constant

Kv Voltage activated K channel

KCC Potassium chloride cotransporter

LEC Lens epithelial cells

LFC Lens fiber cells

M molar

MAP kinase Mitogen-activated protein kinase

MEM Minimum essential medium

MOPS 3-[N-morpholino] propane sulfonic acid

MQAE [N - (Ethoxycarbonylmethyl) - 6 - methoxyquinolinium bromide]

Na Sodium

Na/H Sodium proton exchanger

NaOH Sodium hydroxide

NHE Sodium proton (hydrogen) exchanger

NKCC Sodium -potassium-chloride cotransporter

NMDA N-methyl-D-aspartic acid

NPPB 5-Nitro-2-(3-phenylpropylamino)benzoic acid

ORCC Outwardly rectifying chloride currents

PBS Phosphate buffered saline

PCA Perchloric acid

PCR Polymerase chain reaction

PDGF Platelet derived growth factor

PI Propidium iodide

PS Phosphatidyl serine

Page 201: Ion Transport Mechanisms during Hyposmotic Regulatory and

190

Abbreviation Word

qPCR Quantitative PCR

Rb Rubidium

RbCl Rubidium chloride

Rbi Intracellular rubidium content

RFU Relative fluorescence units

RQ Relative quantity

RT-PCR Reverse transcription polymerase chain reaction

RVD Regulatory volume decrease

RVI Regulatory volume increase

SITS 4-acetamido-4'-isothiocyanostilbene

SK Small conductance K channel

STP Staurosporine

TdT Terminal Deoxynucleotidyl Transferase

TEA Tetraethyl ammonium

TGF-beta Transforming growth factor-beta

TNF-alpha Tumor necrosis alpha

TRAM 1-[(2-Chlorophenyl)diphenylmethyl]-1H-pyrazole

UV Ultraviolet

UVA Ultraviolet A radiations

UVB Ultraviolet B radiations

UVC Ultraviolet C radiations

VRAC Volume regulated anion channels

VSOR Volume sensitive outwardly rectifying Cl channels

WWT Wet weight

Page 202: Ion Transport Mechanisms during Hyposmotic Regulatory and

191