is inflammation a useful target for the treatment of metabolic syndrome?

29
  Is inflammation a useful target for the treatment of metabolic syndrome? PA30035  ANNA MATTHE WS, 2015

Upload: anna-matthews

Post on 01-Nov-2015

10 views

Category:

Documents


0 download

DESCRIPTION

The focus of this literature review is primarily on the significance of the involvement of inflammatory cytokines in the two major aspects of metabolic syndrome: obesity and insulin resistance / type 2 diabetes mellitus (T2DM); and potential novel therapies for the metabolic syndrome with respect to the inflammatory roles in the associated conditions. In particular, the effects of TNF-α, IL-1β and IL-6 secretion on the metabolic syndrome are elucidated; as well as T-lymphocytes, oxidative stress and the link to the JNK pathway. All information used has been collected from articles obtained via Embase, and cited using EndNote in the Harvard-Bath style. As this is a literature review, data from original case studies has been used and appropriately referenced.The findings of the research undertaken suggest that inflammation may indeed be a useful target for the treatment of the metabolic syndrome, with more and more studies identifying promising results in the control of glucose homeostasis in recent years. The main conclusion to be drawn is that inflammatory cytokines TNF-α, IL-1β and IL-6 are likely prospective candidates for new targets in treating the metabolic syndrome, with some anti-diabetic progress already being observed in anti-inflammatory treatments such as chloroquine.

TRANSCRIPT

  • Is inflammation a useful target for the treatment of metabolic

    syndrome? PA30035

    ANNA MATTHEWS, 2015

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Abstract The focus of this literature review is primarily on the significance of the involvement of

    inflammatory cytokines in the two major aspects of metabolic syndrome: obesity and

    insulin resistance / type 2 diabetes mellitus (T2DM); and potential novel therapies for the

    metabolic syndrome with respect to the inflammatory roles in the associated conditions.

    In particular, the effects of TNF-, IL-1 and IL-6 secretion on the metabolic syndrome

    are elucidated; as well as T-lymphocytes, oxidative stress and the link to the JNK

    pathway. All information used has been collected from articles obtained via Embase, and

    cited using EndNote in the Harvard-Bath style. As this is a literature review, data from

    original case studies has been used and appropriately referenced.

    The findings of the research undertaken suggest that inflammation may indeed be a

    useful target for the treatment of the metabolic syndrome, with more and more studies

    identifying promising results in the control of glucose homeostasis in recent years. The

    main conclusion to be drawn is that inflammatory cytokines TNF-, IL-1 and IL-6 are

    likely prospective candidates for new targets in treating the metabolic syndrome, with

    some anti-diabetic progress already being observed in anti-inflammatory treatments

    such as chloroquine.

    Introduction

    Metabolic syndrome is a condition that is characterised by differing clusters of three out

    of four health abnormalities, depending on which criteria is used. WHO defines the

    metabolic syndrome as diagnosis of type 2 diabetes plus any two other risk factors:

    including central obesity, hypertension and dyslipidaemia (Yasein et al. 2010); however,

    the National Cholesterol Education Program (NCEP) Adult Treatment Panel (ATP) III

    characterises it as a combination of any three of the above disorders: with the difference

    that instead of there being a requirement for diagnosis of type 2 diabetes, impaired

    insulin tolerance will suffice, with a fasting blood glucose of 110mg/dL (6.1mmol/L) or

    more. The latter explanation seems to be the more generally accepted and so when

    discussing Metabolic Syndrome, I will be referring to the NCEP ATP-III classification.

    It is becoming an increasing global health issue, as each of the above factors alone carry

    a risk of developing cardiovascular disease, and current statistics show approximately

    51% of T2DM patients die from CVD and associated complications (Morrish et al. 2001).

    Obesity is considered to be a central feature that increases the risk for the development

    of metabolic syndrome-associated diseases such as cancer, asthma, sleep apnoea,

    osteoarthritis, neurodegeneration and gall bladder disease (Hotamisligil 2006).

    1

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    According to Daskalopoulou et al, the treatment of metabolic syndrome outlined by the

    NCEP ATP-III focuses on improving the underlying insulin resistance, and this is

    managed by modifying lifestyle such as diet and exercise and possibly by drugs;

    though monotherapy is often ineffective (Daskalopoulou et al. 2004). Potential drug

    therapies include high-dose statins for lowering LDL-C and triglycerides; fibrates for

    increasing HDL-C levels to >40mg/dL and thiazolidinediones, as these are thought to

    also improve dyslipidaemia and blood pressure (Table 1).

    Healthy lifestyle promotion Drug Intervention Small weight loss

    Moderate physical activity

    Dietary restriction in calorie

    intake

    Change in dietary composition

    Atherogenic dyslipidaemia: Statins; fibrates;

    nicotinic acid

    Elevated blood pressure: No particular agent is

    preferred, although ACE inhibitors and

    angiotensin receptor blockers may carry

    advantage in patients with metabolic disorders

    Insulin resistance and hyperglycaemia:

    Metformin; thiazolidinediones; acarbose; orlistat

    Table 1 Primary Prevention of Cardiovascular Events associated with Metabolic Syndrome according to

    NICE (Paoletti et al. 2006)

    Ahmad et al concurs with insulin sensitivity being a main target for treatment of the

    symptoms of the abnormalities associated with metabolic syndrome, at least as an initial

    step; and goes on to highlight that insulin resistance has been known to exacerbate an

    increase in lipids in diabetic patients (Ahmad et al. 2011). This connection suggests that

    by targeting insulin resistance, other symptoms may be positively addressed

    consequently, such as obesity and hypercholesteraemia. The targets for effective

    treatment of the metabolic syndrome are outlined in Table 2:

    Table 2 The target levels from therapy for metabolic syndrome (Expert Panel on Detection et al. 2001)

    LDL-C

    (Main target of therapy for all

    high-risk patients)

    Patients with Coronary Heart Disease: 2) risk factors: < 130mg/dL

    0-1 risk factor: < 160mg/dL

    Weight control -10% from baseline

    Physical activity 30 45 minutes / day for 3 5 days / week

    Treatment of hypertension < 140/90mmHg

    2

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    However, the lack of effective drug treatment for tackling the metabolic syndrome as a

    whole highlights an obvious clinical need for novel approaches and targets. If we were

    to discover a common underlying cause for all the conditions associated with metabolic

    syndrome - and target that - we may be able to develop an over-arching treatment, which

    would increase patient compliance if taken alone; or improve the effectiveness of current

    treatments if used in combination.

    There has recently been interest in exploring whether or not there is a connection

    between metabolic syndrome and inflammation. I will be reviewing literature on the link

    between the immune system and T2DM primarily, as well as the potential benefits of

    using anti-inflammatory agents as treatment for metabolic syndrome, which are still being

    elucidated. Furthermore, I will discuss the anti-inflammatory aspects of current

    treatments and whether these can be isolated and potentially improved into new,

    targeted treatments.

    The main aspects of metabolic syndrome I will be researching as potential links to

    inflammation are T2DM and obesity, as these are the two that are most poorly controlled.

    The inflammatory cytokines I will be focussing on are T-cells, IL-1, IL-6, and TNF-

    cytokines on the pathogenesis of metabolic syndrome; although there are many to

    investigate, and I will make reference to the other inflammatory factors involved without

    discussing them in great detail.

    Inflammatory Aspects of Insulin Resistance and Obesity

    T2DM occurs when insulin resistance is uncompensated for by the pancreatic -cells,

    due to a deterioration in -cell function (Maedler et al. 2003). Researching the potential

    reasons for -cell dysfunction and death in T2DM produced results linking to

    inflammation.

    TNF-, IL-1 and IL-6 In 1997, Pickup et al published a study on non-insulin dependent diabetes mellitus

    (NIDDM) as a disease of the innate immune system. Three different groups of patients

    were used in the study, all similar ages and of the same ethnicity (white European).

    Those with four or five of the following were considered to be metabolic syndrome

    positive (NIDDM syndrome X positive):

    3

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Hypertriglyceridaemia (VLDL triglyceride > 1.5mmol/L);

    Low serum HDL cholesterol ( 160 and/or diastolic pressure >95mmHg

    and/or receiving anti-hypertensives);

    Coronary heart disease (as assessed by the WHO cardiovascular questionnaire);

    Obesity (calculated by BMI > 30kg/m2)

    Those with nought or one of the above

    features as well as type 2 diabetes mellitus

    were considered to be metabolic syndrome

    negative (NIDDM syndrome X negative).

    Healthy subjects without diabetes acted as a

    control group. The results of fasting blood

    glucose tests are displayed in Figure 1.

    It is evident from these results that the patients

    with metabolic syndrome displayed higher levels

    of acute-phase immune response, with

    increases in C-reactive protein (CRP)

    concentrations. Furthermore, a measure of serum IL-6 concentrations in the groups

    displayed a significant increase in the NIDDM syndrome X positive group compared with

    others (Fig. 2).

    It is also useful to note that the scale of the

    graph here in Figure 2 is logarithmic: therefore,

    the changes are more significant than they may

    first appear.

    Overall, Pickup et als study provides evidence

    that T2DM is associated with increased plasma

    levels of the cells and markers associated with

    acute immune response (Pickup et al. 1997). As a fairly

    early study in this area, the conclusions drawn from it

    were merely that inflammation required more research

    with regards to T2DM pathogenesis, as it was unclear

    whether or not inflammation is the causative agent.

    Figure 2 Comparisons of serum

    concentrations of IL-6 in non-

    diabetics, non-insulin dependent

    diabetes mellitus (NIDDM) syndrome

    X negative and NIDDM syndrome X

    positive. (Pickup et al, 1997)

    Figure 1 Serum C-reactive protein levels of

    non-diabetic, non-insulin dependent diabetes

    mellitus (NIDDM) syndrome X negative and

    NIDDM syndrome X positive patient groups

    (Modified from Pickup et al, 1997)

    4

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    One of the first areas of interest concerning inflammatory mediators in the pathogenesis

    of T2DM was the involvement of IL-1 on -cell destruction, initially seen in T1DM.

    Two studies on the effect of IL-1 on -cell function in fact precede Pickup et als study.

    In 1986, Mandrup-Poulsen deduced that IL-1 appears to play a significant role in the

    inhibition of -cell function symbolic of both types of diabetes mellitus (Mandrup-Poulsen

    1996). Indeed, a study by

    Bendtzen et al showed inhibition

    of insulin secretion from the -

    cells, followed by destruction,

    when rat islets were treated with

    IL-1 and cultured for six

    days (Bendtzen et al. 1986). The

    amount of insulin secretion was

    detected via radioimmunoassay

    during the six subsequent days

    of culture, and the results

    showed that with increasing

    concentrations of IL-1, insulin

    secretion decreased similarly (Fig. 3).

    The mechanism of action of IL-1 on -cell function is thought to be via promotion of

    Fas-triggered apoptosis by activating NF-B in human islets (Maedler et al. 2003) and

    indeed Fas has already been labelled as playing an important role in the pathogenesis

    of T1DM. Maedler et al performed a study in 2001 to ascertain whether or not Fas

    expression constituted a similar effect in T2DM from hyperglycaemia (Maedler et al.

    2001).

    The 2001 study was undertaken using islets that were isolated from the pancreases of

    eight organ donors who had no previous history of diabetes or metabolic disorder. These

    tissues were exposed to elevated glucose concentrations for five days and analysed for

    DNA fragmentation, using the dUTP nick-end labelling (TUNEL) technique to identify the

    nuclei of the -cells, thus indicating the presence of any degradation that occurred.

    TUNEL labels the terminal end of nucleic acids and relies on the presence of nicks in the

    DNA, which can be identified by terminal deoxynucleotidyl transferase (TdT). TdT

    catalyses the addition of the labelled dUTPs (Gavrieli et al. 1992). The results of the

    study were that even when human islets were only transiently exposed to glucose for

    three days, the number of proliferating -cells decreased; therefore indicating

    involvement of glucose-induced -cell death in T2DM (Fig. 4).

    Figure 3 - Comparison between islet-inhibitory and IL-1 effect

    regarding insulin levels in rat islets after being cultured with IL-1

    for six days. The relationship between IL-1 and insulin levels is

    established: increased IL-1 correlated with decreased

    insulin. (Bendtzen et al. 1986)

    5

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Figure 4 - Effect of elevated glucose concentrations on human -cell apoptosis: measured using the

    TUNEL technique. The cells were isolated from the pancreases of eight heart-beating human cadavers.

    Elevated glucose is an important control because T2DM is associated with increased blood glucose levels.

    Panel 1 shows the relative number of TUNEL positive -cells per islet after culturing for 5 days in 5.5, 11.1

    and 33.3mM glucose or 5.5mM D-glucose + 27.8mM L-glucose normalised to a control of 5.5mM glucose

    incubations alone. Panel 2 shows TUNEL-positive -cells per islet during a 10 day culture at 5.5 or

    33.3mM glucose. (Maedler et al, 2001)

    Maedler et al then went on to study the mechanism involved to identify any potential for

    Fas-receptor involvement.

    Figure 5 - Glucose effect on stimulating Fas in the -cells of eight human organ donors as assessed by

    Western Blotting after culturing the islets in suspension containing 5.5mM, 11.1mM and 3.3mM of glucose

    and incubating for 36 hours. Human foreskin fibroblast was used as a positive control for Fas. (Modified

    from Maedler et al, 2001)

    Fig. 5 shows that glucose has a proportional relationship with expression of Fas and Fas-

    ligand. According to Stennicke and Salvesen, the underlying mechanism concerning

    apoptosis of -cells is to do with Fas receptor up-regulation, initiated by glucose. More

    Fas receptors means more interactions with the constitutively active Fas-ligand on

    neighbouring -cells, leading to cleavage of procaspase-8 to caspase-8 and activation

    of caspase-3, resulting in DNA fragmentation (Stennicke and Salvesen 2000).

    6

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Fig. 6 supports Stennicke and Salvesens explanation of -cell death, portraying the

    effect of increasing glucose concentration on activated caspase 8 and 3.

    Addition of an antagonistic anti-Fas antibody ZB4 inhibited the effect of both IL-1 and

    glucose on degradation of -cells (Fig. 7)

    Figure 7 - Effect of Fas receptor blockade on glucose and IL-1 induced -cell DNA fragmentation in

    human islets cultured for 5 days on extracellular-matrix-coated dishes in 5.5 or 33.3mM glucose alone (the

    control) or in the presence of interleukin-1 (IL-1), antagonistic Fas antibody ZB4 or both. Results were

    normalised to control incubations at 5.5mM glucose alone. (Maedler et al, 2001)

    It is evident that the response to both concentrations of glucose are affected similarly by

    ZB4, and -cell degradation levels are greatly reduced with ZB4 added to tissue

    expressing IL-1 compared to the tissue expressing IL-1 alone. From this, we can deduce

    that inhibition of the Fas pathway may be a potential therapeutic target for T2DM, and

    success can be gauged by assessing IL-1 expression in the pancreatic islets.

    Figure 6 The effect of differing concentrations of glucose on procaspase-8 and activated caspase-8

    expression in the pancreases of eight human organ donors after 36 hours incubation (Modified from

    Maedler et al, 2001)

    7

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    A study performed by Spranger et al was designed to examine the effect of inflammatory

    cytokines on the development of T2DM. The format of the study was a nested case-

    control consisting of 27,548 individuals who were free of T2DM at the beginning of the

    study. The case subjects were defined as individuals who then developed T2DM within

    a 2.3 year follow-up period, and these numbered 192 vs 384 control subjects (Spranger

    et al. 2003).

    Spranger et al found IL-6 to be an independent predictor of T2DM development after

    analysis; however, the increase in TNF- and CRP were found to be insignificant when

    adjusting for body mass index (BMI) and waist-to-hip ratio (WHR).

    TNF- and IL-1

    Odds Ratio 95% Confidence Interval

    Reference (low TNF- /

    undetectable IL-1)

    1 n/a

    High TNF- 0.89 0.45 1.73

    Undetectable IL-1 0.95 0.54 1.67

    Interaction term (high TNF- /

    undetectable IL-1)

    2.51 0.84 7.57

    TNF- and IL-6 Reference

    (low TNF- / low IL-6)

    1 n/a

    High TNF- 1.32 0.68 2.52

    High IL-6 2.15 1.12 4.11

    Interaction term (both high) 0.7 0.23 2.16

    IL-1 and IL-6 Reference

    (low IL-6 / undetectable IL-

    1)

    1 n/a

    Undetectable IL-1 0.8 0.43 1.44

    High IL-6 1.14 0.55 2.32

    Interaction term

    (detectable IL-1 / high IL-6)

    3.31 1.14 9.87

    Table 4 - Interaction between IL- IL-1, IL-6 and TNF- on diabetes risk (Spranger et al, 2003)

    8

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    41.0% of case subjects compared to 36.6% of control subjects showed detectable levels

    of IL-1, which is not statistically significant. Nevertheless, when taking into

    consideration the presence of other cytokines alongside IL-1 expression, it was found

    that the combined effects modified the risk of T2DM development (Table 4).

    Therefore, the presence of two or more of the inflammatory cytokines IL-1, IL-6 and

    TNF- may promote the development of diabetes, and could conceivably suggest a

    reason for why traditional Chinese medicines such as Radix Astragali and Radix Ginseng

    which have both hypoglycaemic and anti-inflammatory properties are effective at

    combating diabetes (Xie and Du 2011).

    Dr Paul Lacy first proposed that local generation of cytokines

    in the islets may be responsible for -cell damage; although

    this was initially only thought to be the case in T1DM (Lacy

    1994). However, with increasing studies being undertaken to

    examine potential links between inflammation and T2DM, this

    pathway was analysed with regards to T2DM pathogenesis.

    Westwell-Roper et al performed a study using RMPI cultured

    mouse islets to determine whether macrophages are present

    and might contribute to islet gene expression. The markers

    they looked for in the cells (after 1 and 5 days in RPMI

    suspension i.e. media utilising a bicarbonate and buffering

    system and alterations in the amount of amino acids and

    vitamins) were F4/80 and CD11b. However, their results

    indicate that there was little expression of macrophage genes Emr1 and Itgam; and the

    -cell genes Pdx1, Ins1 and Ins2 did not change during the culture period either (not

    shown).

    This indicates that if there is a link to inflammation in T2DM, it is not these genes that are

    over-expressed.

    Looking at human islet amyloid poplypeptide (hIAPP) in islets, Westwell-Roper et al

    discovered that after treating isolated mouse islets with synthetic IAPP for 4 hours, both

    hIAPP and the toll-like receptor 2 (TLR2) ligand FSL-1 induced expression of genes that

    encode pro-inflammatory cytokines TNF-, IL-1 and IL-6.

    Isolating islet macrophages with synthetic hIAPP and rIAPP showed that hIAPP-treated

    cells formed amyloid fibrils after 2 hours of exposure; but not rIAPP. Pro-IL-1 was

    significantly increased in the hIAPP-treated cells compared with rIAPP-treated cells (Fig.

    9). This is proposed to be due to cross-species recognition.

    Figure 8 - Levels of macrophage and

    -cell genes in mouse islets cultured

    in suspension RPMI at 37oC for 1 day

    and 5 days respectively. (Westwell-

    Roper et al, 2014)

    9

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    This is the first evidence to implicate that resident macrophages are the major islet cell

    type in which hIAPP stimulates IL-1 synthesis and secretion (Westwell-Roper et al.

    2014).

    Chawla et al agree that macrophage infiltration into adipose tissues in a state of obesity

    is significant: in the adipose tissue of lean mice, only 10-15% of cells express the

    macrophage marker F4/80, compared to 45-60% in obese animals (Chawla et al. 2011).

    Further regarding inflammatory markers in T2DM, Donath and Shoelson propose that

    high glucose intake increases the production of IL-1 (Fig. 10); resulting in further

    inflammation of the pancreatic -cells, and impaired insulin secretion.

    Figure 9 Effect of human IAPP (hIAPP) and rat IAPP (rIAPP) on stimulating the synthesis and secretion

    of IL-1 from pancreatic islets. A: hIAPP or rIAPP was added to bone marrow derived macrophages

    following dissolution, and the cells were lysed at the indicated time points for analysis of the expression of

    IL-1 by Western Blotting, using an antibody that detects pro-IL-1 and mature IL-1. B: Expression of pro-

    IL-1 relative to actin quantified by band densitometry. C: IAPP-induced secretion of IL-1 by BMDMs

    and islets evaluated by ELISA after 24 hours. (Modified from Westwell-Roper et al, 2014).

    BMDM = Bone Marrow Derived Macrophages.

    A

    B C

    10

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    In 2010, Badawi et al published an article which focuses on the association of

    inflammatory mediators and obesity-related insulin resistance. It describes that in the

    case of obesity, both animal and human models have shown an increase in adipose

    tissues of TNF- and macrophages. In addition, those who have risk factors associated

    with T2DM exhibit increased serum levels of pro-inflammatory cytokines than those who

    do not (Badawi et al. 25 May 2010). Petersen et al undertook a study concerning the

    insulin-resistant offspring of patients with T2DM to examine the mitochondrial activity

    and identify whether or not it was impaired in the overweight, insulin-resistant children

    compared to lean, insulin-sensitive controls. It was discovered that in insulin-resistant

    subjects, the rate of glucose uptake by myocytes stimulated by insulin was around 60%

    lower in the insulin-resistant subjects compared to the control. This was associated with

    an increase of intra-myocellular lipid content of 80% and a reduction of mitochondrial

    phosphorylation of 30% in the insulin-resistant subjects; indicating that insulin resistance

    could be due to a defect in mitochondrial oxidative phosphorylation (Petersen et al.

    2004).

    Donath and Shoelson also allude to TNF- having some role to play in obesity and

    T2DM, as displayed in Figure 10 above; similarly Xie and Du highlight that the

    inflammatory cytokines released during a state of inflammation and/or obesity result in

    insulin resistance via activation of the JNK pathway (Xie and Du 2011), which will be

    further discussed later.

    Figure 10 - Development of

    Inflammation in type 2 diabetes. High

    glucose intake promotes local

    production of inflammatory cytokines,

    and a decrease in production of

    interleukin-1 receptor antagonist (IL-

    1RA), resulting in immune cell

    recruitment to the islets and ultimately

    tissue inflammation. (Donath and

    Shoelson 2011)

    11

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    The above evidence suggests that inflammation may well play a part in the pathogenesis

    of T2DM: in essence, the cellular stresses indicated in T2DM are thought to either induce

    inflammatory response or be associated with inflammation. These include oxidative

    stress; ectopic lipid deposition in the muscle, liver and pancreas, lipotoxicity and

    glucotoxicity as well as amyloid deposition in the pancreas (Donath and Shoelson 2011).

    Kaneto et al propose that oxidative stress is indeed involved in insulin resistance, as

    seen in a knock-out study concerning -cell derived cell lines and isolated rat islets

    exposed to oxidative stress. The result was a dramatic decrease in insulin gene

    expression seen in the knock-out rats (Kaneto et al. 2004) and may be due to activation

    of the JNK pathway (Fig. 11). This will be further discussed in The JNK Pathway in

    Metabolic Syndrome.

    T-lymphocytes and Inflammation in Metabolic Syndrome Many inflammatory cytokines are products of Th2 lymphocytes (such as IL-4 and IL-13).

    Fracchia and Walsh propose that activated T lymphocytes require glucose transport via

    GLUT1 receptors to accommodate their metabolic demands (Fracchia and Walsh 2015).

    Indeed, Caro-Maldonado et al name the main supply of metabolic fuel for lymphocytes

    to be glucose, lipids and amino acids all of which are increased in diet-induced

    obesity (Caro-Maldonado et al. 2012). It is well known that T-cell activation results in

    metabolic reprogramming. Therefore, if levels of glucose, lipids and amino acids are

    altered, T-cell production will be affected.

    Th1, 2 and 17 cells showed increased proliferation when glucose uptake via GLUT1

    transporters was increased; but TReg cells were not affected, as they utilise lipids rather

    Figure 11 The proposed effects of Oxidative Stress on

    Insulin Resistance. Reactive Oxygen Species (ROS)

    have been shown to interfere with cellular signalling

    pathways. The JNK pathway in the liver is activated by

    ROS, free fatty acids (FFAs) and TNF-. Under obese

    diabetic conditions, levels of these factors rise. Serine

    phosphorylation of insulin receptor substrate-1 (IRS-1)

    inhibits insulin-stimulated tyrosine phosphorylation of the

    receptor, resulting in increased insulin resistance and

    therefore glucose intolerance. (Kaneto et al, 2004)

    12

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    than glucose (Shepherd and Kahn 1999). This indicates that exposure to high levels of

    fuels such as glucose, as is evident in obesity, may lead to over-expression of T-

    lymphocytes, altering immune response.

    Chatzigeorgiou et al portray TNF- and IL-6 in adipocyte interference regarding insulin

    signalling locally in the white adipose tissues (WAT) as well as systemically (Table 5).

    Lymphocyte subset

    Marker Secreted molecules

    Classical inflammatory

    response

    Function in WAT inflammation

    CD8+ T-cells CD3

    CD8

    IFN- Cytotoxic function for

    destruction of virus-

    infected or cancer

    cells

    Activation of WAT

    macrophages; IFN-

    -related WAT

    inflammation

    Th1 cells CD3

    CD4

    IL-12R

    IFN-

    IL-2

    Activate

    macrophages for

    phagocytosis and

    pathogen killing

    Promotion of WAT

    inflammation

    through IFN- and

    RANTES

    Th2 cells CD3

    CD4

    IL-4

    IL-5

    IL-10

    IL-13

    Stimulation of B

    cells, promote

    humoral antibody

    responses &

    eosinophil activation

    Anti-inflammatory

    effects through

    activation of IL-10-

    producing

    macrophages

    Table 5 - Lymphocyte subsets and their implicated roles in white adipose tissue (WAT)

    inflammation (Chatzigeorgiou et al. 2012)

    Chronic Inflammation and Insulin Resistance Konner and Bruning state that the link between obesity, insulin resistance and T2DM has

    been extensively researched; and this research has increased our understanding of the

    interrelation between them (Konner and Bruning 2011). Inflammation is now considered

    to be one of the crucial mechanisms in the development of obesity-associated T2DM

    and insulin resistance (Hotamisligil 2006).

    There are further, more recent studies that suggest chronic inflammation may be

    associated with insulin resistance; and ultimately metabolic syndrome and/or

    T2DM (Dregan et al. 2014). One, conducted by Zuliani et al, measures the levels of low

    13

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    grade systemic inflammation in subjects with metabolic syndrome compared with healthy

    individuals and gave the following results:

    38.3% of subjects with metabolic syndrome had high levels of both insulin resistance

    (IR) and low grade systemic inflammation (LGSI) compared with 17.6% of healthy

    individuals. This shows that although IR and LGSI are not exclusive to metabolic

    syndrome, increased levels are indicative of a predisposition to metabolic syndrome.

    However, we must question whether or not inflammation is a significant enough

    contributing factor to consider anti-inflammatory agents as treatments for metabolic

    syndrome. Indeed, only a slightly increased percentage of subjects had LGSI with

    metabolic syndrome than without (55% vs 41%). (Zuliani et al. 2015)

    A figure outlining the role of inflammatory markers in pathologic conditions from

    overweight to T2DM is redacted from Badawi et al (Fig. 12):

    Figure 12 - Relationship of inflammatory markers to disease states

    FFA = Free fatty acids; TNF- = Tumour necrosis factor ; CRP = C Reactive Protein; LDL = Low density

    lipoproteins; HDL = high density lipoproteins; TG = Triglycerides; IGT = Impaired glucose tolerance.

    (Badawi et al. 25 May 2010)

    This implies that there are many other inflammatory markers related to disease factors

    which have the potential to be explored other than those I am discussing in this literature

    review.

    14

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    The JNK Pathway and Metabolic Syndrome A study by Kaneto et al addressed the involvement of the JNK pathway in the

    development of insulin resistance. Obesity was induced both in a control group of mice

    with wild type JNK and knock-out mice lacking JNK. When the mice were placed on a

    high-fat diet, the obese wild type mice developed mild hyperglycaemia but the blood

    glucose levels in obese JNK-KO mice were significantly lower. These results indicate

    that the knock-out mice were able to avoid obesity-induced hyperglycaemia, and so the

    JNK pathway must play a role in insulin resistance. Fig. 13 proposes a potential

    mechanism for this pathway, with the suggestion that oxidative stress may induce

    nucleocytoplasmic translocation of PDX-1 through activation of the JNK pathway,

    resulting in -cell dysfunction found in both types of diabetes.

    Nguyen et al found that FFA treatment impaired insulin signalling in mice via decreased

    GLUT-4 translocation when compared to mice not injected with FFAs (Fig. 14).

    This was due to inhibition of

    phosphorylation of key

    downstream components such

    as IRS-1 in 3T3-L1 adipocytes

    that were used as an in vitro

    model system (Fig. 15).

    Figure 13 - Possible mechanism for impaired

    -cell function under hyperglycaemic

    conditions in rodents. Pancreatic and

    duodenal homeobox 1 (PDX-1, a.k.a. insulin

    promotor factor 1) is translocated from the

    nuclei to the cytoplasm in response to

    oxidative stress, which in turn can be caused

    by hyperglycaemia seen in diabetes.

    Translocation results in lower levels of PDX-1

    in the nucleus, which is detrimental to

    pancreatic development as it is necessary for

    -cell maturation. (Kaneto et al, 2004)

    Figure 14 Free Fatty Acid (FFA) treatment effect on impaired

    insulin signalling in mice, measured via decreased GLUT-4

    translocation using secreted TNF-. (Nguyen et al. 2005)

    15

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Figure 15 Effects of Insulin Receptor Substrate 1 (IRS-1) from various tissues of obese rats on insulin

    stimulated insulin resistance autophosphorylation (Hotamisligil et al. 1996)

    It was also shown that inhibitory serine phosphorylation of insulin receptor substrate

    (IRS)-1 is responsible for both TNF- and free fatty acid (FFA) induced insulin resistance

    (as alluded to above when discussing Nguyens study); and this phosphorylation was

    markedly increased in obese wild type mice compared to lean wild type mice and JNK-

    KO mice. This demonstrates that the absence of JNK1 leads to an enhancement of IRS-

    1 due to lack of phosphorylation (Kaneto et al. 2004).

    Anti-Inflammatory Effects of Current Treatments of Metabolic Syndrome

    Statins and ACE Inhibitors Two medications used for control of the hypercholesteraemic and hypertensive aspects

    of Metabolic Syndrome were assessed for anti-inflammatory activity when used in

    combination. The study was performed by Han et al and compared the decrease in

    atheroma size when treated with a rosuvastatin and ramipril combination and

    rosuvastatin alone.

    The study revealed that the anti-inflammatory effect observed was an important factor

    for the change in atheroma volume, suggesting that anti-inflammatory mechanisms can

    be elucidated by combining a statin with an ACE inhibitor. Although this study was

    designed to explore the effect of this combination of therapies on reducing the size of

    atheromas, the underlying conditions associated with development of atheromas are

    also seen in metabolic syndrome, and so the benefits seen in metabolic syndrome when

    statins and ACE inhibitors are used for the individual aspects of the condition may

    actually be due, in part, to the anti-inflammatory effect seen in this case of polypharmacy.

    The proposed mechanism of effect of the combined therapy of a statin and an ACE

    inhibitor is one similar to that of methotrexate: ACE inhibitors decrease the amount of

    reactive oxygen species, which as we discussed earlier, may contribute to the

    development of insulin resistance seen in metabolic syndrome; and statins exert a variety

    of effects other than reducing low density cholesterol.

    16

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    According to Marini et al, the pathophysiology of the anti-inflammatory effects of statins

    reside in the inhibition of hydroxymethylglutaryl CoA (HMG CoA) reductase, in turn

    decreasing the production of isoprenoid intermediates farnesyl-PP and geranylgeranyl-

    PP (Marini et al. 2014) and therefore decreasing inflammation.

    Blanco-Colio et al clarify the anti-inflammatory and immunomodulatory effects of such

    HMG CoA reductase inhibitors in Table 6.

    In essence, the anti-inflammatory effects of statins in combination with the anti-ROS

    effects of ACE inhibitors may be responsible for some therapeutic benefit when used in

    metabolic syndrome, and HMG CoA reductase inhibitors could potentially be an area of

    research to go into regarding novel treatments for the metabolic syndrome.

    Thiazolidinediones Thiazolidinediones are peroxisome proliferator-activated receptor (PPAR-) agonists

    that can be used in patients with insulin resistance or T2DM as well as kidney failure.

    According to Szeto and Li, induction of PPAR- results in an inhibition of inflammatory

    cytokines being produced by macrophages (Szeto and Li 2008).

    Indeed, Jiang et al demonstrated the inhibition of TNF- release by troglitazone in a

    study using monocytes prepared from unrelated donors, using PMA- and okadaic acid-

    induced cytokine synthesis to establish the effects (Jiang et al. 1997).

    The result was a clear decrease in expression of the pro-inflammatory cytokine TNF-

    in lipopolysaccharides when troglitazone is added to the monocyte, as established via

    ELISA.

    As we have already elucidated, TNF- plays a significant role in the pathogenesis of

    obesity and T2DM in particular, which are prominent features of the metabolic syndrome.

    This group of anti-diabetic agents, thiazolidinediones, appears to have more than just a

    hypoglycaemic effect contributing to its therapeutic mechanism of action.

    Anti-Inflammatory Effects Immunomodulatory Effects Adhesion molecules Proliferation of lymphoid cells

    Chemoattractant proteins Natural Killer activity

    Pro-inflammatory transcription factors Major histo-compatibility class II

    antigens

    Inflammatory serum markers Organ rejection

    Table 6 Immunomodulatory and anti-inflammatory effects of 3-hydroxy-3-methyl-glutaryl CoA reductase

    (HMG-CoA reductase) inhibitors (Blanco-Colio et al. 2003)

    17

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Biguanides According to Lockwood, therapeutic metformin concentrations of 25M have

    chloroquine-mimetic anti-lysosomal action (which will be discussed later): in a study

    performed by Lockwood on rat heart cells, 25M of metformin alone caused a 20-25%

    inhibition of total protein degradation (reducing ROS and pro-inflammatory cytokine

    release) (Fig. 17A). This action was completely stopped by prior addition of chloroquine,

    indicating that the proteolysis inhibited by metformin is due to the same lysosomal

    subcomponent as is the target for chloroquine (Lockwood 2010).

    Metformins anti-inflammatory effects may contribute to its efficacy in the treatment of

    insulin resistance / T2DM in metabolic syndrome.

    Figure 17 Results from a study performed on rat heart cells. A) Percent inhibition of perfused myocardial

    protein degradation by 25M metformin; B) Synergy of metformin anti-proteolytic action by Zn2+ (Lockwood

    2010)

    It also appears that Zn2+ has synergistic activity when combined with metformin, with

    regards to the proteolytic effect (Fig. 21B). This could show promise as a novel treatment

    for the inflammation associated with T2DM or insulin resistance in metabolic syndrome:

    metformin and zinc.

    18

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Effects of Anti-Inflammatory Treatments on T2DM

    Interleukin-1 Receptor Antagonist A study published in the New England Journal of Medicine in 2007 by Larsen et al

    contains evidence that interleukin-1 receptor antagonist (IL-1RA) is reduced in the

    pancreatic islets of patients with T2DM.

    Larsen et als study involved a double-blind, parallel-group trial where 34 of 70 patients

    with T2DM received 100mg of a recombinant human IL-1RA (Anakinra) via

    subcutaneous injection once a day for 13 weeks; and the remaining 36 patients received

    a placebo.

    Figure 18: Changes in glycated haemoglobin and fasting plasma glucose levels during the study whereby

    100mg of recombinant human IL-1RA (Anakinra) was injected into human subjects once a day for 13

    weeks. (Larsen et al, 2007)

    Fig. 18 depicts the difference in glycated haemoglobin levels from baseline and 13 weeks

    from the study beginning in the Anakinra group and the placebo group.

    19

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    The results were a reduction of 0.33 percentage point in the Anakinra group; and an

    increase of 0.13 percentage point in the placebo group a difference of 0.46 percentage

    point between groups (95% confidence interval 0.01 0.90 and p = 0.03).

    With regards to -cells (Fig. 19), function was increased in the group receiving Anakinra,

    and decreased in the group receiving placebo.

    Figure 19 - -cell function differences observed during the study whereby 100mg of recombinant human

    IL-1RA (Anakinra) was injected into human subjects once a day for 13 weeks. (Larsen et al, 2007)

    Furthermore, Fig. 20 displays that hs-CRP and IL-6 levels were consistently and

    significantly decreased in women who had developed T2DM and were receiving

    Anakinra compared to placebo (Larsen et al. 2007).

    Figure 20 - Markers of Systemic Inflammation observed during the study whereby 100mg of recombinant

    human IL-1RA (Anakinra) was injected into human subjects once a day for 13 weeks (Larsen et al, 2007)

    20

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Put together, these results indicate that the ILRA Anakinra both decreases inflammatory

    markers CRP and IL-6 and improves the symptoms of T2DM with regards to -cell

    function, glycated haemoglobin and fasting plasma glucose (Fig. 22, 23 and 24).

    However, neither changes in the baselines of CRP or IL-6 correlated with the change in

    glycated haemoglobin levels. This suggests that reduced systemic inflammation does

    not play a large part in improved insulin secretion; however, it may prevent -cell

    destruction and promote its regeneration in T2DM.

    Targeting hIAPP with Clodronate Westwell-Roper et al treated isolated mouse islets with clodronate-containing liposomes

    to assess the contribution of macrophages to hIAPP-induced inflammation of the islets;

    and found, via immunostaining, that clodronate reduced the number of F4/80 positive

    islet cells as well as macrophage markers in IAPP, but not -cell markers (Fig.

    21) (Westwell-Roper et al. 2014).

    This was due to phagocyte depletion mediated by clodronate, which in turn prevented

    the expression of pro-inflammatory cytokines normally initiated by hIAPP.

    Figure 21 Effect of treatment with clodronate-containing liposomes (CLOD-lip; 1mg/mL clodronate) on

    expression of inflammatory genes by IAPP in isolated 12-week-old mice compared to control liposomes

    (PBS-lip) for 36 hours. Results assessed by RT-qPCR. (Westwell-Roper et al 2014)

    21

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Anti-TNF- As discussed previously, the study by Hotamisligil in 1993 (using rodents) was one of

    the first that demonstrated the link between TNF- and insulin resistance (Hotamisligil et

    al. 1993). Since then, according to Donath, the results have been repeated in a large

    number of animal models (Donath 2014).

    A double-blind clinical trial by Ofei et al was designed to examine any effect of a

    recombinant-engineered human TNF--neutralising antibody (CDP571) on blood sugar

    control, in obese patients who are diabetic but not insulin dependent. This was a

    relatively short and small study, involving

    only 21 patients over a 2-week period

    receiving intravenous (IV) injections of

    either CPD571 5mg/kg or normal saline.

    The results from this study indicate that

    TNF- neutralisation over a short period of

    time had no effect on treating insulin

    resistance in obese NIDDM patients (Ofei

    et al. 1996).

    However, a more recent and prolonged

    study performed in 2010 by Stanley et al

    focused on the link between obesity and

    TNF- in forty patients with metabolic syndrome but without diabetes. The randomised

    clinical trial included administration of either: the anti-TNF- drug Etanercept 50mg twice

    weekly for 3 months followed by 50mg once weekly for three months; or a placebo.

    The results showed significant improvement in fasting blood glucose in the patients who

    took Etanercept compared to placebo (Fig. 22).

    At the start of the trial, five of the Etanercept group and four of the placebo group had a

    fasting blood glucose of at least 100mg/dL. Three of the five receiving Etanercept and

    one of the four receiving the placebo had normalised fasting glucose after six months

    administration (Stanley et al. 2011).

    These results imply that anti-TNF- treatments have only been proven to be effective for

    metabolic syndrome-related insulin resistance when used long-term.

    Figure 22 - Effect of twice-weekly injections of 50mg

    Etanercept for three months followed by once weekly

    for three months on fasting glucose in obese patients

    with metabolic syndrome compared to placebo

    (Stanley et al 2011)

    22

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Hydroxychloroquine Chloroquine is used against various inflammatory conditions, such as rheumatoid

    arthritis (RA) (Joint Formulary Committee, 2013). Its mechanism of action is via

    interference with vacuolar transport; swelling lysosomal vesicles and making them more

    alkaline. However, it is little known that chloroquine has anti-diabetic actions, and was

    proposed for use in diabetes but cast aside due to safety issues (Lockwood 2010). Its

    hypoglycaemic activity was demonstrated in a controlled trial performed by Wasko et al,

    where hydroxychloroquine was administered to patients with RA and they were

    monitored for development of T2DM, against patients with RA not receiving

    hydroxychloroquine; and the results showed improvement in the hydroxychloroquine

    group (the number of reports of incident diabetes were 54 patients from 1808 in the

    hydoxychloroquine group vs 171 from 3097 in the control group) (Wasko et al. 2007).

    After adjusting for time and confounding factors, it was distinguished that the relative risk

    of developing diabetes was 0.23 when the patient had 4 years of treatment with

    hydroxychloroquine compared to without (Fig. 23).

    Figure 23 - Probability of developing type 2 diabetes mellitus (T2DM) in rheumatoid arthritis (RA) patients

    according to hydroxychloroquine use (Wasko et al 2007)

    This implies that the anti-inflammatory drug chloroquine can have a beneficial effect on

    the prevention of T2DM development; in turn suggesting that the inflammatory aspect of

    T2DM is worth targeting for therapy.

    23

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Conclusions

    Role of Inflammation in Metabolic Syndrome With regards to the role of inflammation in metabolic syndrome, there appears to be

    significant evidence that this may be the case.

    As determined by Stennicke and Salvesen, Fas receptor upregulation by glucose plays

    a role in -cell death: and addition of an anti-Fas antibody to pancreatic cells has already

    exhibited positive effects on reducing -cell apoptosis. The benefit of this therapy is that

    effectiveness is independent of glucose concentration, which also indicates that Fas

    receptor may be over-expressed in patients with metabolic syndrome and could be a

    cause of insulin resistance rather than just over-consumption of glucose.

    A plethora of studies, including Spranger et al, elucidate that inflammatory cytokines are

    increased in T2DM, in particular IL-6 and TNF-.

    Lacy also highlighted the effect that the increased infiltration of macrophages into

    adipose tissue has on cytokine secretion, and ruled out pathways that are not involved;

    and consequently the outcome of hIAPP on cytokine secretion and activation has also

    been addressed. 30-50% more obese mice express the IL-1 secreting macrophage

    F4/80 than lean mice, which indicates that the problems associated with obesity (seen

    in Fig. 1) must be in some part due to increased inflammation: and indeed, as Badawi et

    al discovered, patients with T2DM risk factors exhibited increased serum levels of

    inflammatory cytokines than patients without. Linking this to the fact that Fas receptor

    and IL-1 are upregulated in T2DM; TNF-, FFA and IL-6 levels increased in obesity;

    and low grade systemic inflammation combined with insulin resistance noted in patients

    with metabolic syndrome, the evidence points to inflammation as a propagator of

    metabolic syndrome development.

    The evidence of links between anti-inflammatory and anti-diabetic effects of current

    treatments such as statins, ACE inhibitors, biguanides and thiazolidinediones further

    support this. Furthermore, the evidence of effectiveness of anti-inflammatory treatments

    insofar on glucose homeostasis (for example Anakinra, the IL-1R antagonist) conveys

    insulin resistance to be a result of inflammation rather than vice versa.

    24

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Potential Future Targets The notable potential targets for novel treatments focusing on inflammation include Fas

    receptors; IL-1; IL-6; TNF-; hIAPP and IL-1R, as all have been consistently indicated

    in increased risk of the conditions associated with metabolic syndrome.

    The main issue involved with developing such therapies is expense, but taking into

    account the cost of treating all the individual components of the metabolic syndrome

    separately, plus the eventual cost of consequential heart failure / cancer / liver failure

    may just equate.

    In conclusion, inflammation does appear to link together the conditions associated with

    metabolic syndrome to a fairly significant extent, and so appears to be a reasonable

    target for addressing the symptoms involved as a whole. It is an option worth exploring.

    25

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    References

    Joint Formulary Committee, 2013. British National Formulary. 66 ed. London: British Medical Association and Royal Pharmaceutical Society of Great Britain. Ahmad, M., Hassan, S., Hafeez, F. & Jajja, A., 2011. Prevalence of Various Components of Metabolic Syndrome in our Younger Population. Pakistan Journal of Physiology, 7(2), pp. 46-49. Badawi, A., Klip, A., Haddad, P., Cole, D.E., Garcia Bailo, B., El-Sohemy, A. & Karmali, M., 25 May 2010. Type 2 diabetes mellitus and inflammation: Prospects for biomarkers of risk and nutritional intervention. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy, (3), pp. 173-187. Bendtzen, K., Mandrup-Poulsen, T., Nerup, J., Nielsen, J.H., Dinarello, C.A. & Svenson, M., 1986. Cytotoxicity of human pl 7 interleukin-1 for pancreatic islets of Langerhans. Science, (232), pp. 1545-1547. Blanco-Colio, L.M., Tunon, J., Martin-Ventura, J.L. & Egido, J., 2003. Anti-inflammatory and immunomodulatory effects of statins. Kidney Int, 63(1), pp. 12-23. Caro-Maldonado, A., Gerriets, V.A. & Rathmell, J.C., 2012. Matched and mismatched metabolic fuels in lymphocyte function. Seminars in Immunology, 24(6), pp. 405-413. Chatzigeorgiou, A., Karalis, K.P., Bornstein, S.R. & Chavakis, T., 2012. Lymphocytes in obesity-related adipose tissue inflammation. Diabetologia, 55(10), pp. 2583-2592. Chawla, A., Nguyen, K.D. & Goh, Y.P.S., 2011. Macrophage-mediated inflammation in metabolic disease. Nat Rev Immunol, 11(11), pp. 738-749. Daskalopoulou, S.S., Mikhailidis, D.P. & Elisaf, M., 2004. Prevention and Treatment of the Metabolic Syndrome. Angiology, 55(6), pp. 589-603. Donath, M. & Shoelson, S., 2011. Type 2 diabetes as an inflammatory disease. Nature Reviews: Immunology, (11), pp. 98-107. Donath, M.Y., 2014. Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat Rev Drug Discov, 13(6), pp. 465-476. Dregan, A., Charlton, J., Chowienczyk, P. & Gulliford, M.C., 2014. Chronic inflammatory disorders and risk of type 2 diabetes mellitus, coronary heart disease, and stroke : A population-based cohort study. Circulation, 130(10), pp. 837-844. Expert Panel on Detection, -., Evaluation and Treatment of, -. & High Blood Cholesterol in Adults, -. 2001. Executive Summary of the Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA, 285(19), pp. 2486-2497. Fracchia, K.M. & Walsh, C.M., 2015. Metabolic mysteries of the inflammatory response: T cell polarization and plasticity. International Reviews of Immunology, 34(1), pp. 3-18. Gavrieli, Y., Sherman, Y. & Ben-Sasson, S.A., 1992. Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. The Journal of Cell Biology, 119(3), pp. 493-501. Hotamisligil, G.S., 2006. Inflammation and metabolic disorders. Nature, 444(7121), pp. 860-867. Hotamisligil, G.S., Peraldi P Fau - Budavari, A., Budavari A Fau - Ellis, R., Ellis R Fau - White, M.F., White Mf Fau - Spiegelman, B.M. & Spiegelman, B.M., 1996. IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science, 271(5249), pp. 665-668.

    26

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Hotamisligil, G.S., Shargill Ns Fau - Spiegelman, B.M. & Spiegelman, B.M., 1993. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science, 259, pp. 87-91. Jiang, C., Ting At Fau - Seed, B. & Seed, B., 1997. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Letters to Nature, 391, pp. 82-86. Kaneto, H., Kawamori, D., Nakatani, Y., Gorogawa, S.-i. & Matsuoka, T.-a., 2004. Oxidative Stress and the JNK Pathway as a Potential Therapeutic Target for Diabetes. Drug News & Perspectives, 17(7), p. 447. Konner, A.C. & Bruning, J.C., 2011. Toll-like receptors: linking inflammation to metabolism. Trends in Endocrinology and Metabolism, 22(1), pp. 16-23. Lacy, P.E., 1994. The intraislet macrophage and type 1 diabetes. Mt Sinai J Med, 61, pp. 170-174 Larsen, C.M., Faulenbach, M., Vaag, A., Vlund, A., Ehses, J.A., Seifert, B., Mandrup-Poulsen, T. & Donath, M.Y., 2007. Interleukin-1Receptor Antagonist in Type 2 Diabetes Mellitus. New England Journal of Medicine, 356(15), pp. 1517-1526. Lockwood, T.D., 2010. The lysosome among targets of metformin: new anti-inflammatory uses for an old drug? Expert Opinion on Therapeutic Targets, 14(5), pp. 467-478. Maedler, K., Sergeev, P., Ris, F., Oberholzer, J., Joller-Jemelka, H.I., Spinas, G.A., Kaiser, N., Halban, P.A. & Donath, M.Y., 2003. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. Journal of Clinical Investigation, 110(6), pp. 851-860. Maedler, K., Spinas, G.A., Lehmann, R., Sergeev, P., Weber, M., Fontana, A., Kaiser, N. & Donath, M.Y., 2001. Glucose Induces -Cell Apoptosis Via Upregulation of the Fas Receptor in Human Islets. Diabetes, 50(8), pp. 1683-1690. Mandrup-Poulsen, T., 1996. The role of interleukin-1 in the pathogenesis of IDDM. Diabetologia, (39), pp. 1005-1029. Marini, M.G., Sonnino, C., Previtero, M. & Biasucci, L.M., 2014. Targeting Inflammation: Impact on Atherothrombosis. Journal of Cardiovascular Translational Research, 7, pp. 9-18. Morrish, N.J., Wang Sl Fau - Stevens, L.K., Stevens Lk Fau - Fuller, J.H., Fuller Jh Fau - Keen, H. & Keen, H., 2001. Mortality and causes of death in the WHO Multinational Study of Vascular Disease in Diabetes. Diabetologia, 44(2), pp. 14-21. Nguyen, M.T.A., Satoh, H., Favelyukis, S., Babendure, J.L., Imamura, T., Sbodio, J.I., Zalevsky, J., Dahiyat, B.I., Chi, N.-W. & Olefsky, J.M., 2005. JNK and Tumour Necrosis Factor-alpha Mediate Free Fatty Acid-induced Insulin Resistance in 3T3-L1 Adipocytes. The Journal of Biological Chemistry, 280, pp. 35361-35371. Ofei, F., Hurel, S., Newkirk, J., Sopwith, M. & Taylor, R., 1996. Effects of an Engineered Human AntiTNF- Antibody (CDP571) on Insulin Sensitivity and Glycemic Control in Patients With NIDDM. Diabetes, 45(7), pp. 881-885. Paoletti, R., Bolego, C., Poli, A. & Cignarella, A., 2006. Metabolic Syndrome, Inflammation and Atherosclerosis. Vascular Health and Risk Management, 2(2), pp. 145-152. Petersen, K.F., Dufour S Fau - Befroy, D., Befroy D Fau - Garcia, R., Garcia R Fau - Shulman, G.I. & Shulman, G.I., 2004. Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. New England Journal of Medicine, 350(7), pp. 664-671.

    27

  • Is inflammation a useful target for the treatment of metabolic syndrome? | Anna Matthews

    Pickup, J.C., Mattock, M.B., Chusney, G.D. & Burt, D., 1997. NIDDM as a disease of the innate immune system: association of acute-phase reactants and interleukin-6 with metabolic syndrome X. Diabetologia, 40, pp. 1286-1292. Shepherd, P.R. & Kahn, B.B., 1999. Glucose Transporters and Insulin Action Implications for Insulin Resistance and Diabetes Mellitus. New England Journal of Medicine, 341(4), pp. 248-257. Spranger, J., Kroke A Fau - Mohlig, M., Mohlig M Fau - Hoffmann, K., Hoffmann K Fau - Bergmann, M.M., Bergmann Mm Fau - Ristow, M., Ristow M Fau - Boeing, H., Boeing H Fau - Pfeiffer, A.F.H. & Pfeiffer, A.F., 2003. Inflammatory cytokines and the risk to develop type 2 diabetes: results of the prospective population-based European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Study. Diabetes, 52(3), pp. 812-817. Stanley, T.K., Zanni, M.V., Johnsen, S., Rasheed, S., Makimura, H., Lee, H., Khor, V.K., Ahima, R.S. & Grinspoon, S.K., 2011. TNF-alpha Antagonism with Etanercept Decreases Glucose and Increases the Proportion of High Molecular Weight Adiponectin in Obese Subjects with Features of the Metabolic Syndrome. J Clin Endocrinol Metab, 96(1), pp. E136-E150. Stennicke, H.R. & Salvesen, G.S., 2000. Caspase-controlling intracellular signals by protease zymogen activation. Biochimica et Biophysica Acta, 1477, pp. 299-306. Szeto, C.-C. & Li, P.K.-T., 2008. Antiproteinuric and anti-inflammatory effects of thiazolidinedione (Review Article). Nephrology, 13(1), pp. 53-57. Wasko, M.C., Hubert Hb Fau - Lingala, V.B., Lingala Vb Fau - Elliott, J.R., Elliott Jr Fau - Luggen, M.E., Luggen Me Fau - Fries, J.F., Fries Jf Fau - Ward, M.M. & Ward, M.M., 2007. Hydroxychloroquine and risk of diabetes in patients with rheumatoid arthritis. JAMA, 298(2), pp. 187-193. Westwell-Roper, C.Y., Ehses Ja Fau - Verchere, C.B. & Verchere, C.B., 2014. Resident macrophages mediate islet amyloid polypeptide-induced islet IL-1beta production and beta-cell dysfunction. Diabetes, 16(5), pp. 1698-1711. Xie, W. & Du, L., 2011. Diabetes is an inflammatory disease: evidence from traditional Chinese medicines. Diabetes, Obesity and Metabolism, (13), pp. 289-301. Yasein, N., Ahmad, M., Matrook, F., Nasir, L. & Froelicher, E.S., 2010. Metabolic syndrome in patients with hypertension attending a family practice clinic in Jordan. Eastern Mediterranean Health Journal, 16(4), pp. 375-379. Zuliani, G., Morieri, M.L., Volpato, S., Maggio, M., Cherubini, A., Francesconi, D., Bandinelli, S., Paolisso, G., Guralnik, J.M. & Ferrucci, L., 2015. Insulin resistance and systemic inflammation, but not metabolic syndrome phenotype, predict 9 years mortality in older adults. Atherosclerosis, 235(2), pp. 538-545.

    28