paper - pathogenesis of diabetic nephropathy

Upload: cyro-vidal

Post on 04-Jun-2018

224 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    1/12

    Reviews in Endocrine & Metabolic Disorders 2004;5:237248C 2004 Kluwer Academic Publishers. Manufactured in The Netherlands.

    Pathogenesis of Diabetic NephropathyClaudia van Dijk and Tomas Berl

    University of Colorado Health Sciences Center, Division of Renal

    Diseases and Hypertension, 4200 E. 9th Ave., C-281, Denver, CO

    80262, USA

    Key Words. diabetes, kidney disease, mechanism

    Introduction

    Diabetes Mellitus (DM) is the most common cause of

    end-stage renal disease (ESRD) across racial and ethnic

    groups in the United States (US) and other developed

    countries. According to most recent USRDS reports, dia-

    betic nephropathy (DMN) represents 44% of new cases of

    ESRD in theUnited Statesand accounts forfar higherrates

    in black, Native American and Hispanic populations than

    it does in whites. The incidence of diabetic nephropathy

    (DMN) continues to rise far more rapidly than rates due to

    any other primary diagnosis of ESRD as a result of the in-

    creasing prevalence of DM, predominantly thosewith type

    2 diabetes (T2DM) [1]. A total of 131,173 patients with

    DMN were requiring renal replacement therapy: 77% of

    these were on hemodialysis, 6.2% are on peritoneal dial-

    ysis and 16.1% are transplant recipients according to the

    2002 data report.

    ESRD is associated with high mortality rates, and

    among these, those with DMN have the highest rates. Ad-

    justed relative risk of death in diabetic dialysis patients

    compared to their non-diabetic counterparts was 1.69 in a

    large cohort study [2]. The high mortality rates in the pre-

    dialysis period in these patients may underestimate the

    poor progression that the diagnosis of diabetic nephropa-

    thy imparts [3].

    Predisposing Factors for the Development

    of Diabetic Nephropathy

    As was summarized by Ritz and Orth [4], the risk factors

    associated with the development of DMN in the diabetic

    population include older age, non-caucasian race, male

    sex and poor glycemic, lipid and blood pressure controls.

    Genetic patterns have been established with familial

    occurrences of DMN and associated hypertension and

    cardiovascular diseases in family members of patients

    with DMN [5]. A clustering of DMN in some families

    with IDDM and NIDDM has lead to the belief that a

    genetic susceptibility predisposes to development of

    ESRD. Cumulative risk increase of DMN from 25 to

    72% has been observed if the index case in a diabetic

    family had persistent proteinuria. Linkage to at least one

    major gene on chromosome 3q, in vicinity of the AT1

    locus, has been observed, and polymorphisms in the

    angiotensinogen and angiotensin-1 converting enzyme

    genes appear to make minor contributions [6]. A genetic

    defect in regulation of glycosaminoglycans production by

    endothelial and mesangial cells has also been correlated

    with susceptibility of diabetics to progress to DMN [7].

    Pathophysiology of Hyperfiltration

    and Diabetic Nephropathy

    Although the pathogenesis of type 1 and type 2 DM

    is different, the pathophysiology of microvascular com-

    plications responsible for high morbidity and morality

    rates appears similar. Hyperglycemia underlies microvas-

    cular complications in all end-organ tissues and is at

    least partially responsible for glomerular hyperfiltration

    in DMN (Fig. 1). A very common pathologic feature of

    early DMN is the presence of glomerular hypertrophy,

    with mesangial expansion and glomerular basement mem-

    brane thickening. Likewise, the early hemodynamic al-

    terations include both low afferent and efferent arteriolar

    resistance, a markedly increased plasma flow, a moder-

    ately increased glomerular capillary pressure leading to

    an increased glomerular filtration rate (GFR). This earlystage of hyperfiltration precedes the eventual deteriora-

    tion of renal function. While the hyperfiltration and hy-

    pertrophy are at least in part markers of poor glycemic

    control, the pathogenetic contribution of hyperfiltration

    to the ultimate loss of renal function is not fully defined

    [8].

    The impact of numerous hormonal and vaso-active

    factors in the pathogenesis of the hyperfiltration in

    early stages of DM has been evaluated as to their

    E-mail: [email protected]

    237

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    2/12

    238 van Dijk and Berl

    Fig. 1. Pathophysiology of hyperfiltration.

    role. Prostaglandins, thromboxane, kalikrein and nore-

    pinephrine all do not appear to be associated with in-

    creases in GFR [8]. Findings of elevated atrial natriuretic

    peptide (ANP), fluid and sodium retention as a conse-

    quence of sodium-coupled reabsorption by the brush-

    border co-transporters in the proximal tubule [9], and re-

    duced renin-angiotensin-aldosterone system (RAAS) ac-

    tivity appear to be associated with induction of hyper-

    filtration in diabetics with poor glycemic control [8,9].

    Some of these changes may be related to the volume ex-

    pansion in the hyperglycemic state (secretion of ANP andin inhibition of the RAAS). A direct vasodilation medi-

    ated by the osmotic effect of hyperglycemia or sorbitol-

    induced activation of the polyol pathway has also been

    implicated.

    Insulin appears to ameliorate hyperfiltration by both

    reducing hyperglycemia and by causing systemic vasodi-

    latation that can concomitantly reduce blood pressure. In

    this regard the effect of insulin on mesangial cell con-

    tractility has been extensively investigated [1012]. These

    effects are independent of changes in renin, aldosterone

    or catecholamines. Impaired calcium uptake by vascular

    smooth muscles and impaired contractile response to an-giotensin II in insulin deficient states was noted in vitroin

    such cells, as well as in the evaluation of the hormones

    contribution to hyperfiltration in animal studies [10,12].

    A primary tubular ratherthan a vascularmechanism has

    also been postulated. In this conception it is an increase

    in reabsorption of sodium in proximal tubules that causes

    GFR to increase by the physiologic action of the tubulo

    glomerular feedback (TGF) [13]. The role of an arginine

    ornithine polyamine pathway has been implicated in both

    proximal tubular reabsorption and enlargement of the

    diabetic kidney. Use of difluoromethylornithine (DFMO),

    an inhibitor of the polyamine synthetic enzyme ornithine

    decarboxylase reduced reabsorption and filtration in

    diabetic rats along with attenuation of renal growth [13].

    Use of DFMO to ameliorate DMN must therefore be

    considered and investigation of this pathway is ongoing.

    A growing body of information indicates that vascular

    endothelium plays an important role in the regulation of

    smooth muscle tone and subsequent filtration. Production

    of substances known to influence vascular tone, such

    as endothelin and nitric oxide (NO), is altered under

    diabetic conditions causing an imbalance that may be

    at least partially responsible for the development of

    hyperfiltration [14,15]. Defective endothelial NO (eNOS)has been implicated to play a role in the pathogenesis of

    hyperfiltration. However, all isoforms of NO have been

    found to have significantly increased cortical but not

    medullary expression in streptozotocin (STZ)-induced

    diabetic rats [16]. A possible role for neuronal NO

    (nNOS) to counteract afferent vasoconstiction induced

    by TGF has been postulated [17,18].

    As noted above, nephromegaly is a commonly de-

    scribed feature in early diabetic changes in association

    with hyperfiltration. It is a poor prognostic factor for de-

    velopment of DMN [19]. Whether the development of

    nephromegally is a direct consequence of hyperfiltrationdue to a compensatory state similar to that seen after uni-

    lateral nephrectomy, is questioned in studies shown to

    ameliorate hyperfiltration but not kidney size during strict

    glycemic control [20,21]. The above mentioned tubu-

    loglomerular feedback mechanism argues that increased

    tubular reabsorption spurs filtration and results in ini-

    tial kidney growth. Growth factors such as GH, IGF,

    EGF, TGF and PDGF have been implicated in numer-

    ous reports, with upregulation of their genes and proteins

    noted in the diabetic kidney [22,23]. It must be noted

    that nephromegaly predominantly reflects tubulointersti-

    tial changes, accounting for more than 90% of kidney

    mass.

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    3/12

    Diabetic NephropathyPathogenesis 239

    Development of DMN; A Glomerular

    and Tubulointerstitial Process

    A dissociation between glomerular injury and renal dys-

    function prompted a shift of focus from glomeruloscle-

    rosis, with albuminuria and mesangial expansion, to the

    tubulointerstitial compartment with a strong correlation

    between the degree of tubulointerstitial damage and renal

    function [24]. It is increasingly recognized that tubuloin-

    terstitial changes are not simply a downstream reflection

    of glomerular damage, as evidenced by the high number

    of atubular glomeruli in progressive diabetic nephropathy

    [25]. Pathological examination of a large cohort of dia-

    betics with microalbuminuria showed that 29% had typi-

    cal DMN including balanced severity of glomerular scle-

    rosis, arteriolar hyalinosis and tubulointerstitial changes.In contrast 42% displayed predominant tubular atrophy

    and interstitial expansion [26]. Patients with patholog-

    ical changes had significantly worse glycemic control

    as evidenced by elevated HbA1C, but interestingly high

    prevalence of proliferative retinopathy occurred mainly

    in the group with typical diabetic glomerular changes.

    Glycemic, hypertensive and aging factors could all con-

    tribute to a varying picture of pathologic changes. Mecha-

    nisms proposed by which non-glomerular renal dysfunc-

    tion occurs may be related to simultaneous exposure of

    prosclerotic cytokines in glomeruli and tubulointerstitium

    and to the tubulotoxicity caused by the protein contentsin filtrate overloading the proximal tubules reabsorptive

    ability, which is the predominant site of tubular damage

    [27]. Increased filtered plasma protein reaches the proxi-

    mal tubule in the diabetic kidney characterized by abnor-

    mal permeability. Resultant endocytosis of these proteins

    by tubular epithelium exceeds lysosmal capacity when

    large quantities of protein are present in the filtrate. Rup-

    ture of lysosomes and phenotypical transformation of ep-

    ithelial cells to release endothelin and chemotactic factors

    results. Potent vasoconstriction and chemotaxis then leads

    to inflammation, ischemia and ultimately fibrosis, espe-

    cially in most heavily burdened region such as proximal

    tubules and surrounding interstitium [26].Finally, a postglomerular vasoconstriction has been

    postulated to occur with hyperglycemia which may re-

    sult in tubular ischemia and degeneration. The tubular

    ischemia leads to further interstitial leak of glomerular

    filtrate resulting in interstitial fibrosis [28]. A contribution

    of angiotensin II to tubulo-interstitial injury has also been

    proposed [29].

    Microalbuminuria/Proteinuria

    Lifetime risk of persistent proteinuria is 33%in type 1 DM,

    with a peak incidenceduring seconddecade of IDDM [30].

    Dependence on insulin had a significant influence on the

    risk of progression to overt nephropathy once microal-

    buminuria was diagnosed in a 10 year follow-up study

    [31]. Proteinuria was found to be a powerful predictor ofshortened survival in patients with both type 1 and 2 DM

    [32]. This likely reflects the correlation between microal-

    buminuria and vascular complications. When detected in

    early stages, a more recent study has shown that regres-

    sion of microalbuminuria is feasible, and this correlates

    with preservation of renal function [33].

    Proteinuria occurs as a result of abnormal charge

    permselectivity. This occurs due to reduction in glomeru-

    lar charge density as a consequence of non-enzymatic gly-

    cosylation of various basement membrane proteins and

    altered glycosaminoglycan (GAG) metabolism leading to

    reduced heparan sulfate content. GAG administration inanimal models of diabetes has been shown to exert an

    antiproliferative and antimitogenic effect on glomerular

    epithelial cells and increases negative electrical charge of

    the endothelium [34].

    The association between high protein diet and deterio-

    rationof renal function is controversial andstudiesdone on

    varying cohorts of patients infrequently included diabet-

    ics. Due to differences in design, sample size and methods

    used to assess progression of renal disease, studies have

    inconsistently shown the beneficial effect of dietary pro-

    tein restriction [35]. Pooled results suggest significant re-

    duction of decline in GFR and urinary albumin excretion

    as well as death in protein restricted patients. An initial

    reduction in single-nephron GFR was observed with sub-

    sequent slowing of progression of renal disease in several

    studies while other reports show correlation between pro-

    tein intakeand GFR, but not with albuminuria [36]. Evalu-

    ation of potential benefits of reduction of proteinuria with

    ACE-I therapy while maintaining normal-to-high protein

    intake in order to provide adequate nutritional support,

    has not been tested in humans. Animal studies showed a

    significant reduction in albuminuria during high protein

    intake with concomitant use of ACE-inhibition. Based on

    currently available data the recommended dietary protein

    intakeranges between 0.50.8 gm/kg of body weight, withapproximately 65% as high biological value protein and

    caloric intake sufficient to maintain body weight.

    Hypertension

    Hypertension and diabetes are co-morbid conditions with

    a nearly 100% correlation, associated as part of the

    metabolic syndrome which is growing in endemic pro-

    portions [37]. Increased incidence of atherosclerosis and

    association with left ventricular hypertrophy and diastolic

    dysfunction in the diabetic population has led to the de-

    scription of a diabetic cardiomyopathy. Many previously

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    4/12

    240 van Dijk and Berl

    published reports have observed the renoprotective effect

    of blood pressure reduction [38,39] leading to more ag-

    gressive recommendation for blood pressure control in di-

    abetics, aiming for a target of less than 130/80 (JNC VII)[40]. Independent benefits are shown with angiotensin-

    converting enzyme-inhibitors (ACE-I) and angiotensin re-

    ceptor blockers (ARB) for rate of loss of renal function,

    time to ESRD, stabilization of microalbuminuria, regres-

    sion to normoalbuminuria, and even for some cardiovas-

    cular outcomes and mortality for any given blood pres-

    sure [4143]. Non-hemodynamic benefits of these agents

    are likely underlying these observations. The role of an-

    giotensin receptor 1 (AT1) versus AT2 blockade and newer

    agents such as vasopeptidase inhibitors are currently un-

    derinvestigation[44,45]. In this review, we will not further

    discuss the RAAS system and its hemodynamic effects asthis has been extensively covered in the aforementioned

    published reports [3843].

    Non-Hemodynamic Mechanisms of DMN

    Role of advanced glycosylation endproducts(AGEs)Accumulation of nonenzymatically derived glycosylation

    products on proteins, lipids and amino acids results in

    formation of Amadori products. These products of a cova-

    lent, nonenzymatic Maillard reaction include circulating

    and tissue-structure proteins such as arterial-wall collagen

    and glomerular basement membrane proteins. Conversion

    of these Amadori proteins over months and years into

    AGEs with highly cross-linked nature, responsible for

    pathogenesis in diabetic complications, appears to occur

    more rapidly in tissues of diabetic patients compared to a

    similar phenomenon in aging [46,47]. Markedly increased

    circulating levels of AGE peptides were noted in diabetics

    with ESRD in a study comparing serum AGE levels to

    Fig. 2. Advanced glycosylation endproducts pathway.

    their diabetic counterparts without renalinvolvement [48].

    Accumulation of AGEs is characteristic of DMN, particu-

    larly in the mesangium and in nodular lesions. Staining for

    renal specific AGEs was reported to correlate with severityof DMN in human diabetic subjects [49].

    Extracellular matrix proteins, known to have a low

    turnover, are easily susceptible to AGE modification with

    formation of inter- and intracellular cross-links. The for-

    mation of crosslinks is responsible for structural changes,

    including increased stiffness and density and decreased

    thermal stability, resistance to proteolytic degradation and

    loss of epithelial phenotype. Changes in cellular adhesion

    and decreased affinity of laminin and fibronectin for type

    IV collagen and heparin sulfate proteoglycan result in al-

    tered protein permeability [50]. The prevention of AGE

    formation as well as inhibition of AGE-induced crosslinkshave shown to prevent the occurrence of albuminuria,

    confirming the importance of proposed mechanism in the

    pathogenesis of DMN [51]. Modification of lipoproteins

    (apo-B and LDL) resulting in their delayed clearance, en-

    hanced oxygen radical formation with subsequent nuclear

    factor-B (NFB) activation and cell proliferation or pro-

    grammed cell death are consequences of AGE accumula-

    tion believed to be implicated in pathogenesis of diabetic

    complications (Fig. 2).

    RAGE, a receptor for AGE that is present on

    macrophages, is a member of the immunoglobulin super-

    family. RAGE is responsible for the clearance of AGE-

    modified proteins. Renal clearance of breakdown prod-

    ucts is ultimately responsible for elimination; therefore

    the presence of renal insufficiency leads to a signifi-

    cantly increased plasma and tissue levels of AGEs and

    its breakdown products [48]. The accumulation of circu-

    lating AGEs is directly related to extent of remaining renal

    function and may further contribute to a more rapid dete-

    rioration towards ESRD.

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    5/12

    Diabetic NephropathyPathogenesis 241

    Balance of AGE/RAGE system. Various receptors for

    AGE have been identified on multiple cell types. The

    most extensively described receptor is RAGE. Affinity for

    AGEs with fondness for accumulation in renal tissue (N-carboxymethyllysine (CML) and pentosidine) to RAGE

    suggest importance of this receptor in the development

    of nephropathy. Studies have shown correlation of CML

    and pentosidine with glomerular lesions in both diabetic

    and non-diabetic renal disease; only in diabetic conditions

    however did this seem to be associated with upregula-

    tion of RAGE [49]. It seems therefore that the balance of

    AGE/RAGE system especially in diabetes becomes one of

    the predominant factors to consider in pathogenesis of re-

    nal disease. Interactions of AGEs with RAGE to generate

    reactive oxygen species (ROS) have been implicated as a

    mechanism underlying these findings [52]. Activation of arange of second messenger systems and increased produc-

    tion of cytokines including TGF, PDGF and IL-1 have

    also resulted from interaction between AGEs and RAGE.

    AGEs and tubular-epithelial myofibroblast transdif-

    ferentiation. The occurrence of transdifferentiation of

    tubuloepithelial cells into mesenchymal phenotype is

    observed and felt to be mediated through RAGE in a

    TGF-dependent fashion [53]. Activation of NFB and

    protein kinase C (PKC) is mediated through AGE in a

    self-sustaining mechanism. Binding sites of NFB arepresent on RAGE and this binding lowers the threshold

    for AGE to induce NFB during anti-oxidant depletion

    [54,55]. Exposure of proximal tubules to high levels of

    AGE-peptides due to active reabsorption in this portion

    of the nephron and interaction with RAGE which have

    shown to be distributed at this site [56], could explain the

    phenomenon of tubular-epithelial myofibroblast transdif-

    ferentiation (TEMT).

    AGEs and oxidative stress. Induction of endothelial dys-

    function as the result of accumulation of AGEs withdefective vasodilatation and reduced NO predisposes to

    vascular complications. This process is further enhanced

    by monocyte chemotaxis, adhesion-molecule expres-

    sion, decreased prostacycline formation and plasminogen-

    activator inhibitor-1 (PAI-1) induction [57]. AGE-induced

    increase in oxidative stress has beendemonstrated in DMN

    and ESRD [58] and attenuation of mitochondrial super-

    oxide production leads to diminished AGE accumulation

    pointing out the self-perpetuating mechanism of the devel-

    opment of DMNtrough oxidative andglycation pathways.

    Blockage of AGEs with aminoguanidine was equally ef-

    fective to ACE-I in animal models of diabetes to reduce

    proteinuria and tubular as well as glomerular nitrotyrosine

    levels to control levels,suggesting notonly theimportance

    of binding of Amadori compounds but also an additional

    blockade of inducible nitric oxide synthase and free oxy-

    gen radical propagation in the prevention of DMN [59].

    Animal studies. As described in previously mentioned

    experiments, AGEs has been implicated in the patho-

    genesis of DMN. Various mechanisms by which AGEs

    contribute to the occurrence of renal disease have been

    postulated including aforementioned interactions with its

    receptors, enlisting various growth and transcription fac-

    tors, cytokines and oxidative stress pathways as its mayor

    players. Induction of profibrotic cytokines and growth fac-

    tors, with increased expression of laminin and type IV

    collagen after AGE-injection into animals independentof glycemic control, leads credence to the existence of

    an AGE-mediated mechanism of fibrogenesis [60]. In-

    traperitoneal administration of AGE to normal SJL mice

    enhanced 1 type IV collagen, laminin B1 and TGF

    mRNA. These effect where abolished by co-treatment

    with aminoguanidine, an AGE-inhibitor, further implicat-

    ing AGE in causal relationship between AGE accumu-

    lation and renal injury [61]. Longterm exposure of rats

    to AGE albumin through tail vein injections resulted in

    doubling of plasma AGE levels and fourfold increase of

    urinary and kidney contents of AGE. These biochemical

    changes correlated with increase of glomerular volume,

    widening of basement membrane, expansion of mesangialmatrix and proteinuria. Deleterious effects on the vascular

    system were also noted.

    Human data. Evidence of TEMT (tubular-epithelial

    myofibroblast transdifferentiation) was noted on renal

    biopsies of humans with IDDM by positive immunostain-

    ing of-smooth muscle actin (-SMA) in tubular epithe-

    lial cells in early stages of nephropathy. Co-localization of

    AGE andRAGE in a Kimmelstiel-Wilson lesions of a post-

    mortem kidney further provides a potential pathogenic

    mechanism for development of tubulo-interstitial fibro-

    sis [53]. The presence of TGF in human specimens

    in association with TEMT however, has not yet been

    confirmed.

    Role of reactive oxygen species (Fig. 3)Hyperglycemia induces vascular injury through complex

    overlapping pathways including enzymatic and nonenzy-

    matic processes including formation of AGE, activated

    PKC and reactive oxygen species (ROS). The effect of

    anti-oxidant therapy in cell and animal studies strongly

    suggest an important role for ROS in theinitiation andpro-

    gression of DMN [62]. Lack of occurrence of nephropa-

    thy in normoglycemic insulin-resistant patients, where

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    6/12

    242 van Dijk and Berl

    Fig. 3. Reactive oxygen species pathway.

    increased superoxide production also is observed, along

    with paucity of evidence for beneficial anti-oxidant effects

    in humans may, however, suggest a supportive but not

    initiating role for ROS [63]. Reducing equivalents result-

    ing from glycolytic process in the metabolism of glucose

    drive the synthesis of adenosine triphosphate via oxidative

    phosphorylation in mitochondria (Fig. 3). Byproducts of

    this mitochondrial oxidative phosphorylation include free

    radicals. As a result of the abnormal metabolic milieu in

    DM (hyperglycemia, hyperlipidemia and increased FFA)

    ROS are thought to be increased. Increased production

    of glyco-oxidants, glycated compounds, oxidized LDL,superoxidants, nitrotyrosine and elevated levels of iso-

    prostanes, 8-hydroxydeoxy- guanosine and lipid perox-

    ides have been reported in cell, animal and human studies

    (reviewed in [62] Glucose auto-oxidation also results in

    free radical formation. Increases in oxidative stress dam-

    ages cellular proteins and promotes leukocyte adhesion

    to the endothelium while inhibiting its barrier function

    [64,65].

    H2O2 is increasingly recognized as an intracellular

    messenger produced in response to receptor stimulation.

    Propagation of its signal occurs by oxidizing cysteine

    residues in active sites of protein tyrosine phosphatase.The function of various proteins including protein ki-

    nases and transcription factors can be altered through

    oxidation of their H2O2-sensitive cysteine residues [66].

    Pathways affected by overproduction of superoxide in-

    clude the polyols, AGEs, PKC, TGF and NFB, all

    of which are known to mediate vascular damage in

    diabetics.

    An antioxidant defense system is employed in

    healthy cells that includes ROS scavengers such as uric

    acid, ascorbic acid, catalase, glutathione, glutathione

    peroxidase and superoxide dismutases. In pathologic

    circumstances failure of this scavenger system occurs

    due to overwhelming production of ROS. Irreversible

    modification of biologic macromolecules and altered NO

    bio-availability can lead to inflammation, endothelial

    dysfunction, fibrosis and apoptotic cell death. Resultant

    clinical syndromes include hypertension, renal disease

    and accelerated atherosclerosis.

    Animal studies versus human data. Major enzymatic

    sources of ROS generation are NADPH oxidases of the

    nonphagocytic cell types such as endothelial cells, vas-

    cular smooth muscle cells, renal mesangial and tubu-

    lar cells among others [67,68]. In response to a varietyof hormones, growth factors, cytokines and mechanical

    stress as well as its own end-product ROS, NADPH ox-

    idase can regulate signaling of ROS into a cascade of

    oxidative damage. A study on human arterial contents

    of NADPH oxidase reported marked elevations in car-

    diovascular risk groups compared to controls. Diabetes

    and hyperlipidemia were independently associated with

    NADPH oxidase-derived ROS generation in this study

    [69]. Elevated levels of markers of oxidative stress such

    as urinary isoprostanes and oxidized low-density lipopro-

    tein cholesterol also have been reported in diabetic pa-

    tients [70]. Besides the mulitfactorial sources of ROS,a renal derived NADPH oxidase termed renox has been

    shown to be expressed in renal parenchyma. This pro-

    vides a local source of ROS to renal tissue, which is vul-

    nerable to oxidative damage [68]. Only small amounts

    of ROS are generated in healthy renal mesangium and

    tubular cells. Animal studies suggest a role for the consti-

    tutively active NADPH oxidase leading to ROS produc-

    tion under normal physiologic conditions in regulation of

    medullary blood flow and arterial pressure. Under exces-

    sive oxidative stress this may lead to renovascular hyper-

    tension [71]. Modulation of local ROS production in renal

    parenchyma may be a promising step in the prevention of

    DMN.

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    7/12

    Diabetic NephropathyPathogenesis 243

    Fig. 4. Polyol pathway.

    Role of the polyol pathwaySubstantial contributions of the polyol pathway are pos-

    tulated in addition to previously discussed effects from

    glycated products and their receptors to induce oxidative

    stress as a result of hyperglycemia. The polyol pathway

    consists of aldose reductase (AR) which reduces glucose

    to sorbitol with the aid of co-factor nicotinamide adenine

    dinucleotide phosphate (NADPH), and sorbitol dehydro-

    genase (SDH) with co-factor NAD+, responsible for con-

    version of sorbitol to fructose (Fig. 4). Under normal phys-

    iologic conditions only a small amount of glucose is han-

    dled through this pathway; however, significant increases

    from 3 up to 30% can be seen in the diabetic state. Cellu-lar accumulation of sorbitol and depletion of myoinositol

    have been documented to happen selectively in tissues

    predisposed to developing diabetic complications, includ-

    ing the kidney [72]. Prevention of some of the pathologic

    changes in animal models of diabetic nephropathy as well

    as other diabetic complications by inhibition of the polyol

    pathway points out the importance of this enzyme system

    in the pathophysiology in DMN [7375].

    Mechanisms by which polyols contribute to the occur-

    rence of diabetic complications in addition to induction

    of oxidative stress possibly include osmotic-induced vas-

    cular damage through accumulation of sorbitol and freeradical scavenging functions. The poylol pathway plays a

    role in the induction of oxidative stress in several ways.

    Due to channeling of glucose into the polyol pathway un-

    der hyperglycemic conditions, a significant depletion of

    NADPH, co-factor to AR, causes a diminished regen-

    eration of glutathione (GSH) by glutathione reductase

    (Fig. 4). Reduced GSH stores are then responsible for de-

    creased antioxidant properties in the defence against ROS.

    In addition SDH activity under hyperglycemic conditions

    shunt NAD+ into substrate for the reaction of NADPH

    oxidase to form ROS. Also, fructose and its metabolites

    whenglycosylated are more potent glycation end-products

    fueling AGE-induced oxidative stress [76]. Increase flux

    through the pentose phosphate pathway (PPP), by deplet-

    ing its inhibitor NADPH and by oxidative reactions, al-

    tering the NADH/NAD+ratio further promotes oxidative

    stress [77].

    Animal data versus human studies. The fact that mice

    tend to have low levels of aldose reductase and diabetic

    mice appear to be resistant to the development of cataract,

    raised the question of polyol-induced damage to end-organ

    tissues common to human diabetic disease. Transgenic

    mice studies leading to overexpression of AR in combina-

    tion with streptozocin (STZ)-induced diabetes resulted in

    development of cataracts [78]. Evidence for increases ofoxidative stress in lenses of mice who developed cataract

    suggest that AR contributes to diabetes-associated oxida-

    tivestress. Similar findings of a role forAR in theinduction

    of diabetic neuropathy has been described. The role of AR

    in development of DMN has been studied extensively in

    the last decade as well.

    AR expression in rats predominates in the inner stripe

    of the outer medulla, the inner medulla and at the papillary

    tip. Relatively less expression of AR can be observed in

    the outer medulla and cortex, both in mesangial and prox-

    imal tubule cells as evidenced by accumulation of sorbitol

    and fructose [79]. Increases in AR mRNA in rat mesangialcells in response to elevated glucose levels and increased

    renal AR gene expression in STZ-induced diabetic rats

    implicate that hyperglycemia stimulates AR gene expres-

    sion [80]. The effect of hypertonicity was suggested to be

    the mechanism by which hyperglycemia may influence

    AR gene expression by activation of the osmotic response

    element (ORE). Glucose, however, more so than other os-

    molytes at both markedly and mildly hyperglycemic con-

    ditions was able to alter AR gene expression [81].

    In humans, a genetic variation in the AR gene (ALR2)

    of patients with type 1 DM has been shown, which may

    contribute to the genetic susceptibility to DMN [82].

    Emerging interest in the polyol pathway as a potential

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    8/12

    244 van Dijk and Berl

    target for modification of diabetic complications also fol-

    lows from findings of increased AR mRNA expression

    in patients with DMN compared to type 1 DM with-

    out nephropathy suggesting that the degree of AR geneexpression can modulate risk for development of DMN

    [83]. Controversy however surrounding the contribution

    of AR in DMN is the result of lack of efficacy of AR

    inhibitors in human trials. Findings of reduced hyperfil-

    tration and/or progression of microalbuminuria in some

    human trials are contradicted by trials without attenua-

    tion of renal microvascular complications [84,85]. This

    has been mainly attributed to dose-limiting side effects

    or inability to achieve adequate tissue levels of drug but

    may imply that AR is not the single predominant enzyme

    involved in the intricate pathways that lead to DMN but

    rather contributes to the early pathogenesis in the presenceof hyperglycemia [86].

    Final common pathways of diabetic microvasculardamageRegulation of vascular functions such as vasodilation, per-

    meability, endothelial activation and growth factor signal-

    ing occurs through intracellular signaling molecules such

    as MAPK, PKC and NFB. Activation of phospholipase

    C and subsequent increases in Ca2+, stimulate DAG and

    leads to activation of PKC. De novo synthesisof DAG with

    subsequent PKC activation can occur under metabolic

    stress, leading to chronically elevated states [87]. Oxi-

    dants, AGEs, hexosamine, flow abnormalitites and hy-

    pertension all have been shown to alter the activities of

    these kinases and upregulation of DAG, PKC, NFB and

    MAPK all have been demonstrated in various diabetic an-

    imal models [87] (Fig. 5).

    The MAPK cascade plays a central role in a range

    of biological processes relevant to DMN, including cell

    Fig. 5. Final common pathway.

    growth, differentiation and apoptosis. Three main groups

    of MAPK (extracellularsignal regulatedkinases (ERK), c-

    JunN-terminal kinases (JNK) andp38 kinases vary in their

    involvement in diabetic induced nephropathy. Classicallyp38 MAP kinase is linked to osmotic stress, JNK responds

    to forms of cellular stress, whereas ERKs primarily are re-

    garded as growth signalingkinases.Overlap between these

    different functions however has been described [87]. All

    three types are thought to potentially be part of the stress

    activated protein kinases (SAP). In concert all MAPKs

    produce the full range of cellular adaptation found as

    the basis of diabetic complications. Vascular damage due

    to increased vascular permeability, alterations in blood

    flow, NO dysregulation and leukocyte adhesion are the

    result of increased signaling molecules, hyperosmolar

    stress and dysregulation of oxidants and anti-oxidants.Associated induction of growth factors (TGF, VEGF)

    and cytokines (TNF, IL1, IGF-1) is also described and

    has been shown to stimulate proliferation of mesan-

    gial cells, contributing to nephromegally and fibrosis

    (Fig. 5).

    TGF is considered to be the pivotal cytokine in me-

    diating collagen formation of the kidney. Its uniquely

    powerful fibrogenic potential is the result of upregula-

    tion of matrix synthesis, inhibition of matrix degradation

    and modulation of matrix receptor expression to facilitate

    cell-matrix interactions. Induction of TGF by glucose

    and glycated proteins seems to be a PKC-dependent phe-

    nomenon (reviewed in 23).

    Since targeting any of the single pathways by selective

    inhibition causes attenuation but not complete reversion

    or halt of progression in diabetic complications, it seems

    that a change of expression of intracellular messengers

    more downstream to all these individual pathways may

    be of benefit. Development of inhibitors of intracellular

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    9/12

    Diabetic NephropathyPathogenesis 245

    Fig. 6. Hemodynamic and non-hemodynamic factors.

    signaling molecules, however, will need to be specific

    since many cellular processes are dependent on intact sig-

    naling of these messengers. In search of selective isotypes

    of PKC, associated with microvascular injury, 1 and

    2 emerged, both of which are expressed in glomeruli

    [88,89]. Specific inhibition of PKC have been shown to

    prevent mesangial expansion and glomerular dysfunction

    in diabetic mice [90,91].

    Interaction between hemodynamic

    and metabolic factors

    Interaction of hemodynamic factors such as increased

    systemic and intraglomerular pressures, activation of the

    RAAS with subsequent hemodymanic changes, increase

    in endothelin/EDRF and other vaso-active hormones with

    metabolic changes including ROS, AGE, polyols, an-

    giotensin, reduction of nephrin, cell growth stimulants

    and increase in cellular matrix, cytokine and intracellu-

    lar mediators compound the deleterious effects of the dia-

    betic milieu (Fig. 6). This constellation of factors reduces

    the threshold for injury via a final common pathway of

    increases in intracellular messengers (PKC, MAPK), nu-

    clear transcription factors (NFB) and growth factors (cy-

    tokine, TGFand VEGF). All these changes subsequently

    result in development of proteinuria, glomerulosclerosisand tubulo-interstitial fibrosis. It is of note that the inter-

    action of hemodynamic and nonhemodynamic pathways

    seems to involve TGF, making it a prime candidate for

    development of antagonists to treat diabetic nephropathy.

    References

    1. USRDS Reportincidence and prevalence of ESRD.Am J Kidney

    Dis2003;41(452): 4256.

    2. Fenton SS, Schaubel DE, Desmeules M, Morrison HI, Mao Y,

    Copleston P, Jeffery JR, Kjellstrand CM. Hemodialysis versus peri-

    toneal dialysis: A comparison of adjusted mortality rates. Am J Kid-

    ney Dis1997;30:334342.

    3. Chantrel F, Enache I, Bouiller M, Kolb I, Kunz K, Petitjean P,

    MoulinB, Hannedouche T. Abysmal prognosis of patients with type

    2 diabetes entering dialysis.Nephrol Dial Transplant1999;14:129

    136.

    4. Ritz E, Orth SR. Nephropathy in patients with type 2 diabetes mel-

    litus.N Engl J Med1999;341:11271133.

    5. Bowden DW, Sale M, Howard TD, Qadri A, Spray BJ, Rothschild

    CB, Akots G, Rich SS, Freedman BI. Linkage of genetic markers

    on human chromosomes 20 and 12 to NIDDM in Caucasian sib

    pairs with a history of diabetic nephropathy.Diabetes 1997;46:882

    886.

    6. Krolewski AS. Genetics of diabetic nephropathy: Evidence for ma-

    jor and minor gene effects.Kidney Int1999;55:15821596.

    7. Deckert T, Horowitz IM, Kofoed-Enevoldsen A, Kjellen L, Deckert

    M, Lykkelund C, Burcharth F. Possible genetic defects in regula-tion of glycosaminoglycans in patients with diabetic nephropathy.

    Diabetes1991;40:764770.

    8. Bank N. Mechanisms of diabetic hyperfiltration. Kidney Int

    1991;40:792807.

    9. Bank N, Aynedjian HS. Progressive increases in luminal glucose

    stimulate proximal sodium absorption in normal and diabetic rats.

    J Clin Invest1990;86:309316.

    10. Kreisberg JI. Insulin requirement for contraction of cultured rat

    glomerular mesangial cells in response to angiotensin II: Possible

    role for insulin in modulating glomerular hemodynamics. Proc Natl

    Acad Sci USA1982;79:41904192.

    11. Kamm KE, Stull JT. Regulation of smooth muscle contractile ele-

    ments by second messengers. Annu Rev Physiol1989;51:299313.

    12. Bank N, Lahorra MA, Aynedjian HS. Acute effect of calcium

    and insulin on hyperfiltration of early diabetes. Am J Physiol

    1987;252:E13E20.

    13. Thomson SC, Deng A, Bao D, Satriano J, Blantz RC, Vallon V.

    Ornithine decarboxylase, kidney size, and the tubular hypothesis of

    glomerular hyperfiltration in experimental diabetes. J Clin Invest

    2001;107:217224.

    14. Wakabayashi I, Hatake K, Kimura N, Kakishita E, Nagai K. Modu-

    lationof vascular tonusby the endotheliumin experimental diabetes.

    Life Sci 1987;40:643648.

    15. Veelken R, Hilgers KF, Hartner A, Haas A, Bohmer KP, Sterzel

    RB. Nitric oxide synthase isoforms and glomerular hyperfiltration

    in early diabetic nephropathy. J Am Soc Nephrol 2000;11:7179.

    16. Choi KC, Kim NH, An MR, Kang DG, Kim SW, Lee J. Alter-

    ations of intrarenal renin-angiotensin and nitric oxide systems in

    streptozotocin-induced diabetic rats.Kidney Int Suppl 1997;60:S23

    S27.

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    10/12

    246 van Dijk and Berl

    17. Ichihara A, Inscho EW, Imig JD, Navar LG. Neuronal nitric oxide

    synthase modulates rat renal microvascular function. Am J Physiol

    1998;274:F516F524.

    18. Komers R, Anderson S. Paradoxes of nitric oxide in the diabetickidney. Am J Physiol Renal Physiol2003;284:F1121F1137.

    19. Baumgartl HJ, Sigl G, Banholzer P, Haslbeck M, Standl E. On the

    prognosis of IDDMpatients withlarge kidneys.Nephrol Dial Trans-

    plant1998;13:630634.

    20. Wiseman MJ, Saunders AJ, Keen H, Viberti G. Effect of blood

    glucose control on increased glomerular filtration rate and kidney

    size in insulin-dependent diabetes. N Engl J Med 1985;312:617

    621.

    21. Tuttle KR, Bruton JL, Perusek MC, Lancaster JL, Kopp DT, De-

    Fronzo RA. Effect of strict glycemic control on renal hemodynamic

    response to amino acids and renal enlargement in insulin-dependent

    diabetes mellitus.N Engl J Med1991;324:16261632.

    22. Flyvbjerg A. Putative pathophysiological role of growth factors and

    cytokines in experimental diabetic kidney disease. Diabetologia

    2000;43:12051223.23. Cooper ME. Interaction of metabolic and haemodynamic fac-

    tors in mediating experimental diabetic nephropathy. Diabetologia

    2001;44:19571972.

    24. Taft JL, Nolan CJ, Yeung SP, Hewitson TD, Martin FI. Clinical

    and histological correlations of decline in renal function in diabetic

    patients with proteinuria.Diabetes1994;43:10461051.

    25. Gandhi M, Olson JL, Meyer TW. Contribution of tubular injury to

    loss of remnant kidney function.Kidney Int1998;54:11571165.

    26. Fioretto P, Mauer M, Brocco E, Velussi M, Frigato F, Muollo B,

    Sambataro M, Abaterusso C, Baggio B, Crepaldi G, Nosadini R.

    Patterns of renal injury in NIDDM patients with microalbuminuria.

    Diabetologia1996;39:15691576.

    27. Nuyts GD, Yaqoob M, Nouwen EJ, Patrick AW, McClelland P,

    MacFarlane IA, Bell GM, DeBroe ME. Human urinary intestinal

    alkaline phosphatase as an indicator of S3-segement-specific alter-ations in incipient diabetic nephropathy. Nephrol Dial Transplant

    1994;9:377381.

    28. Gilbert RE, Cooper ME. The tubulointerstitium in progressive dia-

    betic kidney disease: More than an aftermath of glomerular injury?

    Kidney Int1999;56:16271637.

    29. CaoZ, Cooper ME.Role of angiotensin II in tubulointerstitial injury.

    Semin Nephrol2001;21:554562.

    30. Krolewski AS, Warram JH, Christlieb AR, Busick EJ, Kahn CR.

    The changing natural history of nephropathy in type I diabetes. Am

    J Med1985;78:785794.

    31. Mogensen CE. Microalbuminuria as a predictor of clinical diabetic

    nephropathy.Kidney Int1987;31:673689.

    32. AdlerAI, Stevens RJ,Manley SE,Bilous RW, CullCA, HolmanRR.

    Development and progressionof nephropathyin type2 diabetes:The

    United Kingdom Prospective Diabetes Study (UKPDS 64). KidneyInt2003;63:225232.

    33. Perkins BA, Ficociello LH, Silva KH, Finkelstein DM, Warram JH,

    Krolewski AS. Regression of microalbuminuria in type 1 diabetes.

    N Engl J Med2003;348:22852293.

    34. Gambaro G, Cavazzana AO, Luzi P, Piccoli A, Borsatti A, Crepaldi

    G, Marchi E, Venturini AP, Baggio B. Glycosaminoglycans prevent

    morphological renal alterations and albuminuria in diabetic rats.

    Kidney Int1992;42:285291.

    35. Pedrini MT, Levey AS, Lau J, Chalmers TC, Wang PH. The ef-

    fect of dietary protein restriction on the progression of diabetic

    and nondiabetic renal diseases: A meta-analysis.Ann Intern Med

    1996;124:627632.

    36. Kalk WJ, Osler C, Constable J, Kruger M, Panz V. Influence of di-

    etary protein on glomerular filtration and urinary albumin excretion

    in insulin-dependent diabetes.Am J Clin Nutr1992;56:169173.

    37. Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syn-

    drome among US adults: Findings from the third National Health

    and Nutrition Examination Survey.JAMA2002;287:356359.

    38. Bakris G, Williams M, Dworkin L, Elliott W, Epstein M, Toto R,Tuttle K, Douglas J, Hsueh W, Sowers J. Preserving renal function

    in adultswith hypertension anddiabetes:A consensus approach.Am

    J Kidney 2000;36:646661.

    39. Lewis JB, Berl T, Bain RP, Rohde RD, Lewis EJ. Effect of intensive

    blood pressure control on the course of type 1 diabetic nephropathy.

    Collaborative Study Group.Am J Kidney Dis1999;34:809817.

    40. ChobanianAV, BakrisGL, BlackHR, CushmanWC,GreenLA,Izzo

    JL, Jr, Jones DW, Materson BJ, Oparil S, Wright JT Jr, Roccella EJ.

    The seventh report of the joint national committee on prevention,

    detection,evaluation, and treatment of highblood pressure:The JNC

    7 report.Jama2003;289:25602572.

    41. TaalMW, Chertow GM,Rennke HG,Gurnani A,JiangT,Shahsafaei

    A, Troy JL, Brenner BM, Mackenzie HS. Mechanisms underly-

    ing renoprotection during renin-angiotensin system blockade.Am J

    Physiol Renal Physiol2001;280:F343F355.42. Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis

    JB, Ritz E, Atkins RC, Rohde R, Raz I. Renoprotective effect

    of the angiotensin-receptor antagonist irbesartan in patients with

    nephropathy due to type 2 diabetes. N Engl J Med2001;345:851

    860.

    43. Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE,

    Parving HH,RemuzziG, Snapinn SM,ZhangZ, ShahinfarS. Effects

    of losartan on renal and cardiovascular outcomes in patients with

    type 2 diabetes and nephropathy. N Engl J Med2001;345:861869.

    44. Cao Z, Kelly DJ, Cox A, Casley D, Forbes JM, Martinello P, Dean

    R, GilbertRE, Cooper ME.Angiotensin type 2 receptor is expressed

    in the adult rat kidney and promotes cellular proliferation and apop-

    tosis.Kidney Int2000;58:24372451.

    45. Taal MW, Nenov VD, Wong W, Satyal SR, Sakharova O, Choi JH,

    Troy JL, BrennerBM. Vasopeptidase inhibition affords greater reno-protection than angiotensin-converting enzyme inhibition alone. J

    Am Soc Nephrol2001;12:20512059.

    46. Heidland A, SebekovaK, Schinzel R. Advanced glycationend prod-

    ucts and the progressive course of renal disease. Am J Kidney Dis

    2001;38:S100S106.

    47. Brownlee M, Cerami A, Vlassara H. Advanced glycosylation end

    products in tissue and the biochemical basis of diabetic complica-

    tions.N Engl J Med1988;318:13151321.

    48. Makita Z, Radoff S, Rayfield EJ, Yang Z, Skolnik E, Delaney

    V, Friedman EA, Cerami A, Vlassara H. Advanced glycosylation

    end products in patients with diabetic nephropathy. N Engl J Med

    1991;325:836842.

    49. TanjiN, MarkowitzGS, Fu C, Kislinger T, TaguchiA, Pischetsrieder

    M, Stern D, Schmidt AM, DAgati VD. Expression of advanced

    glycation end products and their cellular receptor RAGE in dia-betic nephropathy and nondiabetic renal disease. J Am Soc Nephrol

    2000;11:16561666.

    50. Charonis AS, Tsilbary EC. Structural and functional changes of

    laminin and typeIV collagen afternonenzymatic glycation.Diabetes

    1992;41(Suppl 2):4951.

    51. Forbes J,Thallas T, ThomasMC, FoundsHW, Burns WC,Jerums G,

    Cooper ME. The breakdown of preexisting advanced glycation end

    products is associated with reduced renal fibrosis in experimental

    diabetes. 2003FASEB J2003;17:17621764.

    52. Wautier MP, Chappey O, Corda S, Stern DM,Schmidt AM,Wautier

    JL. Activation of NADPH oxidase by AGE links oxidant stress to

    altered gene expression via RAGE.Am J Physiol Endocrinol Metab

    2001;280:E685E694.

    53. Oldfield MD, Bach LA, Forbes JM, Nikolic-Paterson D, McRobert

    A,ThallasV, AtkinsRC, OsickaT,Jerums G,CooperME. Advanced

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    11/12

    Diabetic NephropathyPathogenesis 247

    glycation end products causeepithelial-myofibroblast transdifferen-

    tiation via thereceptor for advanced glycation end products (RAGE).

    J Clin Invest2001;108:18531863.

    54. Lal MA, Brismar H, Eklof AC, Aperia A. Role of oxidative stress inadvanced glycation end product-induced mesangial cell activation.

    Kidney Int2002;61:20062014.

    55. Li J, Schmidt AM. Characterization and functional analysis of the

    promoter of RAGE, the receptor for advanced glycation end prod-

    ucts.J Biol Chem 1997;272:1649816506.

    56. Brett J, Schmidt AM, Yan SD, Zou YS, Weidman E, Pinsky

    D, Nowygrod R, Neeper M, Przysiecki C, Shaw A. Survey of

    the distribution of a newly characterized receptor for advanced

    glycation end products in tissues. Am J Pathol 1993;143:1699

    1712.

    57. Singh R, Barden A, Mori T, Beilin L. Advanced glycation end-

    products: A review.Diabetologia2001;44:129146.

    58. Miyata T, Wada Y, Cai Z, Iida Y, Horie K, Yasuda Y, Maeda K,

    KurokawaK, van Ypersele de Strihou C. Implication of an increased

    oxidative stressin the formationof advanced glycation endproductsin patients with end-stage renal failure. Kidney Int1997;51:1170

    1181.

    59. Forbes JM, Cooper ME, Thallas V, Burns WC, Thomas MC,

    Brammar GC, Lee F, Grant SL, Burrell LA, Jerums G, Osicka TM.

    Reduction of the accumulation of advanced glycation end products

    by ACE inhibition in experimental diabetic nephropathy. Diabetes

    2002;51:32743282.

    60. Yang CW, Vlassara H, Peten EP, He CJ, Striker GE, Striker LJ. Ad-

    vancedglycation end products up-regulate gene expression found in

    diabetic glomerular disease. ProcNatl Acad SciUSA 1994;91:9436

    9440.

    61. Vlassara H, Striker LJ, Teichberg S, Fuh H, Li YM, Steffes M.

    Advanced glycation end products induce glomerular sclerosis and

    albuminuriain normalrats. ProcNatl Acad SciUSA 1994;91:11704

    11708.62. Reactive Oxygen Species and Diabetic Nephropathy. Proceedings

    of the Hyonam Kidney Laboratory, Soon Chun Hyang University

    International Diabetes Symposium. Seoul, Korea, January 1819,

    2003.J Am Soc Nephrol2003;14:S209S296.

    63. Kuroki T, Isshiki K, King GL. Oxidative stress: The lead or support-

    ing actor in the pathogenesis of diabetic complications.J Am Soc

    Nephrol2003;14:S216S220.

    64. BaynesJW.Role of oxidativestressin development of complications

    in diabetes.Diabetes1991;40:405412.

    65. SuzukiS, Hinokio Y, Komatu K,OhtomoM, OnodaM, HiraiS, Hirai

    M,HiraiA, ChibaM, KasugaS, Akai H,ToyotaT.Oxidativedamage

    to mitochondrial DNAand its relationship to diabetic complications.

    Diabetes Res Clin Pract1999;45:161168.

    66. Rhee SG, Chang TS, Bae YS, Lee SR, Kang SW. Cellular reg-

    ulation by hydrogen peroxide. J Am Soc Nephrol 2003;14:S211S215.

    67. Griendling KK, Sorescu D, Ushio-Fukai M. NAD(P)H oxidase:

    Role in cardiovascular biology and disease. Circ Res2000;86:494

    501.

    68. ShioseA, KurodaJ, TsuruyaK, HiraiM, HirakataH, NaitoS, Hattori

    M, Sakaki Y, Sumimoto H. A novelsuperoxide-producingNAD(P)H

    oxidase in kidney.J Biol Chem 2001;276:14171423.

    69. Guzik TJ, West NE, Black E, McDonald D, Ratnatunga C, Pillai

    R, Channon KM. Vascular superoxide production by NAD(P)H ox-

    idase: Association with endothelial dysfunction and clinical risk

    factors.Circ Res 2000;86:E85E90.

    70. Devaraj S, Hirany SV, Burk RF, Jialal I. Divergence between LDL

    oxidative susceptibility and urinary F(2)-isoprostanes as measures

    of oxidative stress in type 2 diabetes. Clin Chem 2001;47:1974

    1979.

    71. Lerman LO, Nath KA, Rodriguez-Porcel M, Krier JD, Schwartz

    RS, Napoli C, Romero JC. Increased oxidative stress in exper-

    imental renovascular hypertension. Hypertension 2001;37:541

    546.72. Greene DA, Lattimer SA, Sima AA. Sorbitol, phospho-

    inositides, and sodium-potassium-ATPase in the pathogene-

    sis of diabetic complications. N Engl J M ed 1987;316:599

    606.

    73. Mauer SM, Steffes MW, Azar S, Brown DM. Effects of sorbinil on

    glomerular structure and function in long-term-diabetic rats. Dia-

    betes1989;38:839846.

    74. Yue DK, Hanwell MA, Satchell PM, Turtle JR. The effect of aldose

    reductase inhibition on motor nerve conduction velocity in diabetic

    rats.Diabetes1982;31:789794.

    75. Robison WG, Jr, Nagata M, Laver N, Hohman TC, Kinoshita

    JH. Diabetic-like retinopathy in rats prevented with an aldose

    reductase inhibitor. Invest Ophthalmol Vis Sci 1989;30:2285

    2292.

    76. Chung SS, Ho EC, Lam KS, Chung SK. Contribution of polyolpathway to diabetes-induced oxidative stress. J Am Soc Nephrol

    2003;14:S233S236.

    77. Steer KA, Sochor M, McLean P. Renal hypertrophy in experimental

    diabetes. Changes in pentose phosphate pathway activity.Diabetes

    1985;34:485490.

    78. Lee AY, Chung SK, Chung SS. Demonstration that polyol accumu-

    lation is responsible for diabetic cataract by the use of transgenic

    mice expressing the aldose reductase gene in the lens. Proc Natl

    Acad Sci USA1995;92:27802784.

    79. Terubayashi H, Sato S, Nishimura C, Kador PF, Kinoshita JH. Lo-

    calization of aldose and aldehyde reductase in the kidney. Kidney

    Int1989;36:843851.

    80. Ghahary A, Luo JM, Gong YW, Chakrabarti S, Sima AA, Murphy

    LJ. Increased renal aldose reductase activity, immunoreactiv-

    ity, and mRNA in streptozocin-induced diabetic rats. Diabetes1989;38:10671071.

    81. KanekoM, Carper D, Nishimura C, Millen J,Bock M,Hohman TC.

    Induction of aldose reductase expression in rat kidney mesangial

    cells and Chinese hamster ovary cells under hypertonic conditions.

    Exp Cell Res1990;188:135140.

    82. Heesom AE, Hibberd ML, Millward A, Demaine AG. Polymor-

    phism in the 5-end of the aldose reductase gene is strongly associ-

    ated with thedevelopment of diabeticnephropathy in type I diabetes.

    Diabetes1997;46:287291.

    83. Shah VO, Dorin RI, Sun Y, Braun M, Zager PG. Aldose reduc-

    tase gene expression is increased in diabetic nephropathy. J Clin

    Endocrinol Metab 1997;82:22942298.

    84. Ranganathan S, Krempf M, Feraille E, Charbonnel B. Short term

    effect of an aldose reductase inhibitor on urinary albumin excretion

    rate (UAER) and glomerular filtration rate (GFR) in type 1 diabeticpatients with incipient nephropathy. Diabete Metab 1993;19:257

    261.

    85. McAuliffe AV, Brooks BA, Fisher EJ, Molyneaux LM, Yue DK.

    Administration of ascorbic acid and an aldose reductase inhibitor

    (tolrestat) in diabetes: Effect on urinary albumin excretion.Nephron

    1998;80:277284.

    86. Sheetz MJ, King GL. Molecular understanding of hyperglycemias

    adverse effects for diabetic complications. JAMA 2002;288:2579

    2588.

    87. Tomlinson DR. Mitogen-activated protein kinases as glucose trans-

    ducers for diabetic complications. Dianetologia 1999;42:1271

    1281.

    88. Ishii H, Koya D, King GL. Protein kinase C activation and its role

    in the development of vascular complications in diabetes mellitus.

    J Mol Med1998;76:2131.

  • 8/13/2019 Paper - Pathogenesis of Diabetic Nephropathy

    12/12

    248 van Dijk and Berl

    89. Meier M, King GL. Protein kinase C activation and its pharmaco-

    logical inhibition in vascular disease.Vasc Med2000;5:173185.

    90. Ishii H, Jirousek MR, Koya D, Takagi C, Xia P, Clermont A, Bursell

    SE, Kern TS, Ballas LM, Heath WF, Stramm LE, Feener EP, KingGL. Amelioration of vascular dysfunctions in diabetic rats by an

    oral PKC beta inhibitor. Science1996;272:728731.

    91. Koya D, Haneda M, Nakagawa H, Isshiki K, Sato H, Maeda

    S, Sugimoto T, Yasuda H, Kashiwagi A, Ways DK, King GL,

    Kikkawa R. Amelioration of accelerated diabetic mesangial ex-

    pansion by treatment with a PKC beta inhibitor in diabetic db/dbmice, a rodent model for type 2 diabetes. Faseb J 2000;14:439

    447.