Sindromes de Insulino Resistencia

Embed Size (px)

DESCRIPTION

SDSD

Citation preview

  • Genetic Syndromes of Severe Insulin Resistance

    Robert K. Semple, David B. Savage, Elaine K. Cochran, Phillip Gorden,and Stephen ORahilly

    University of Cambridge Metabolic Research Laboratories (R.K.S., D.B.S., S.O.), Institute of Metabolic Science,Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom; and Clinical Endocrinology Branch (E.K.C., P.G.),National Institute of Diabetes, Digestive and Kidney Diseases, Bethesda, Maryland 20892

    Insulin resistance is among themost prevalent endocrine derangements in theworld, and it is closely associatedwithmajor diseases of global reach including diabetesmellitus, atherosclerosis, nonalcoholic fatty liver disease,and ovulatory dysfunction. It is most commonly found in those with obesity but may also occur in an unusuallysevere form in rare patients with monogenic defects. Such patients may loosely be grouped into those withprimary disorders of insulin signaling and those with defects in adipose tissue development or function (lipo-dystrophy). The severe insulin resistanceofboth subgroupsputspatients at riskofacceleratedcomplicationsandposes severe challenges in clinical management. However, the clinical disorders produced by different geneticdefects are often biochemically and clinically distinct and are associatedwith distinct risks of complications. Thismeans that optimal management of affected patients should take into account the specific natural history ofeach condition. In clinical practice, they are often underdiagnosed, however, with low rates of identification ofthe underlying genetic defect, a problem compounded by confusing and overlapping nomenclature and clas-sification. We now review recent developments in understanding of genetic forms of severe insulin resistanceand/or lipodystrophy and suggest a revised classification based on growing knowledge of the underlyingpathophysiology. (Endocrine Reviews 32: 498514, 2011)

    I. IntroductionII. Definition and Prevalence of Severe IRIII. Generic Clinical Features of Severe IR

    A. Abnormal glucose homeostasisB. Ovarian dysfunctionC. Acanthosis nigricans

    IV. Clinical Features Limited to Some Severe IR SubtypesA. Dyslipidemia and hepatic steatosisB. Abnormal adipose development or topographyC. Growth disorders

    V. Sexual Dimorphism in Severe IRVI. Biochemical Subphenotyping of Severe IRVII. Monogenic IR Classification/NomenclatureVIII. The INSR SpectrumIX. Downstream Insulin Signaling DefectsX. Disorders of Adipose Tissue Development/Function

    (Lipodystrophies)XI. Digenic IRXII. Complex SyndromesXIII. Therapy

    A. Dietary and lifestyle modificationB. Insulin sensitization and replacementC. Adipose tissue offloading

    XIV. Summary

    I. Introduction

    Insulin resistance (IR), or more precisely the reduced re-sponsiveness of the body to the glucose-lowering ac-tivity of insulin, is closely associatedwith someof themostprevalent chronic clinical disorders, namely type 2 diabe-tes, atherosclerosis, polycystic ovarian syndrome, and he-patic steatosis. The population-wide toll of morbidity andmortality attributable to IR is large and growing, in par-ticular due to the consequences of coronary artery disease(1), type2diabetes (2), polycystic ovary syndrome (3), andfatty liver disease, and indeed IR is a cardinal feature of themetabolic syndrome itself (4).

    Although IR is a traitwith significant heritability (58),it is usually only clinically expressed in the context of obe-sity, especially where this has a predominantly centripetaldistribution. In a small number of patients, however, IR ofan unusually severe degree developswithout obesity, or inassociation with generalized or regional lack of adiposetissue. Many such patients harbor pathogenic single gene

    ISSN Print 0021-972X ISSN Online 1945-7197Printed in U.S.A.Copyright 2011 by The Endocrine Societydoi: 10.1210/er.2010-0020 Received October 1, 2010. Accepted March 28, 2011.First Published Online May 2, 2011

    * R.K.S. and D.B.S. contributed equally to this work.Abbreviations:AR,Autosomal recessive;CIDEC,cell death-inducingDNAfragmentation factorA-like effector family-C; DS, Donohue syndrome; IGFBP1, IGF binding protein-1; INSR, insulinreceptor; IR, insulin resistance; mTORC1, mammalian target of rapamycin complex 1; PTRF,polymerase 1 and transcript release factor; RMS, Rabson Mendenhall syndrome.

    R E V I E W

    498 edrv.endojournals.org Endocrine Reviews, August 2011, 32(4):498514

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • mutations, and several of these have been identified inrecent years. These genetic defects may currently begrouped into those affecting insulin signaling and thoseaffecting adipocyte development and/or function. De-tailed physiological study of such patients with definedgenetic defects has begun to identify distinct subpheno-types of IR and has yielded insights into the mechanisticbasis of more prevalent forms of IR.

    Severe IR may also arise through acquired, immune-mediated mechanisms. These include antibodies againsteither insulin or the insulin receptor leading to blockade ofinsulin action (9) or autoimmune destruction of adi-pocytes leading to lipodystrophy (10, 11). Many, but notall, of these patients have coexisting autoimmune disease,alerting physicians to the possibility of acquired severe IR.These disorders have been recently reviewed (1215).

    The prevailing clinical nomenclature in the field of se-vere IR dates from early work on these syndromes in the1970s (9, 16).Wenowdescribe the current state of knowl-edge of genetic forms of severe IR, suggest a refined clas-sification based on recent findings, and review currentlyavailable treatments.

    II. Definition and Prevalence of Severe IR

    Plasma insulin, whether determined in the fasting state orafter a glucose challenge, is a continuous variable, and sothresholds used to diagnose IR and severe IR are arbi-trary. Furthermore, such thresholds are only reliable be-fore -cell decompensation has occurred. In entirely insu-lin-deficient individuals, severe IRmaybedefined solely interms of the body mass-adjusted requirements for exoge-nous insulin to maintain euglycemia, whereas in nondia-betic patients with compensated IR, severe IR may be de-fined solely in terms of plasma insulin levels before and/orafter a glucose challenge, with reference to data from acontrol population. However, between these extremes, inpatients with relative rather than absolute insulin defi-ciency, diagnosis of severe IR is based on semiquantitativeassessment of the biochemical abnormality coupled withclinical evidence of severe IR. A further complication isthat severe IR is most commonly seen in obese patients,and yet they are a group far less likely to harbor single genedefects. Similarly, puberty is a time of physiological IR,and so ideally biochemical assessment of possible severeIR should be made with reference to normative data de-rived from people of similar adiposity and developmentalstage. Subject to these caveats, a suggested working diag-nostic scheme for likely monogenic severe IR is shown inFig. 1. This emphasizes that, whereas numerical determi-nants of severe IR have utility in the settings of normal

    glycemia and absolute insulin deficiency, diagnosis in thecontext of partial -cell decompensation, which is themost common scenario, relies heavily on interpretation ofphysical signs and clinical history. Although not widelyemployed in current clinical practice, we have also founda nomogram derived from a large, nondiabetic popula-tion, showing the relationship between body mass indexand insulin levels to be helpful in discriminating degrees ofIR in overweight/obese patients that are manifestly dispro-portionate to thedegreeof adiposity andare thusmore likelyto have a contribution from a single gene defect (Fig. 1B).

    Thesediagnostic complexitiesmeanthat severe IR isoftennot recognized, especially in men. Furthermore, patientspresent to many different clinical services according to theirdominant clinical problem, and for these reasons no popu-lation-based prevalence figures exist. However, clinical ex-perience suggests that approximately 0.10.5% of patientsin hospital-based diabetes practices may have monogenicforms of severe IR.

    III. Generic Clinical Features of Severe IR

    A. Abnormal glucose homeostasisClinical awareness of IR is greatest among those car-

    ing for patients with established diabetes mellitus, whereit is recognizedmost commonly by a requirement for large

    Fig. 1. Diagnosis of possible monogenic severe IR. A, Suggested(arbitrary) diagnostic criteria. B, Relationship between body mass index(B.M.I.) and fasting plasma insulin in a healthy European nondiabeticpopulation (n 800). The solid line represents the 50th centile, andthe dashed lines the 5th and 95th centiles. [Figure courtesy of Prof.Nicholas J. Wareham.]

    Endocrine Reviews, August 2011, 32(4):498514 edrv.endojournals.org 499

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • doses of exogenous insulin. However, hyperglycemia isnot usually the earliest clinical manifestation of severe IR.It may be recognized much earlier by the presence of ac-anthosis nigricans and/or ovarian hyperandrogenism inwomen. Furthermore, symptomatic hypoglycemia oftenprecedes hyperglycemia, sometimes bymany years. Char-acteristically, hypoglycemia related to severe IR occurspostprandially, with autonomic symptoms sometimesprogressing toneuroglycopeniaandseizures ifnotabrogatedby oral carbohydrate. Such severe postprandial hypoglyce-miamaybeseen inpatientswith insulinreceptordefects (17),insulin signal transduction defects, or primary lipodystro-phies (18, 19). Its mechanism is unclear, but it most likelyrelates to severe impairment of hepatic insulin clearance dueprimarily to a insulin receptor defect or secondarily to theconsequences of hepatic steatosis (20).

    Even in the context of severe loss of insulin receptorfunction, hyperplasia of pancreatic-cells, with attendantextreme hyperinsulinemia, may prevent hyperglycemiaformany years, indicating that the receptor on islets them-selves is not a prerequisite for -cell expansion, as sug-gested by some murine studies (21). However, in mostcases pancreatic -cell hyperplasia does eventually fail tocompensate for severe IR, and hyperglycemia ensues.Commonly, hyperglycemia diagnostic of diabetes is onlyseen after an oral glucose challenge, contrasting with fast-ing hypoglycemia or normoglycemia, making fasting glu-cose alone an inadequate diagnostic test for diabetes in thecontext of severe IR. Compounding this, glycosylated he-moglobin may be normal, or even low, at the time whenpostload glucose levels are diagnostic of diabetes. Al-though these observations suggest that the various currentdiagnostic criteria for diabetes may have different utilitiesin predicting risk of diabetic complications in severe IR,this has not yet been studied. The time taken to -celldecompensation varies substantially, with diabetes devel-oping in the neonatal period in the most severe cases andin the fourth decade or beyond at the milder end of thespectrum, especially in men.

    B. Ovarian dysfunctionSevere IRmost commonly presents to clinical attention

    first as oligomenorrhea and severe hyperandrogenism inyoung women after menarche, although the underlyinghyperinsulinemia is often not recognized. Ovarian ultra-sonography usually reveals multiple peripheral cysts asseen in idiopathic polycystic ovary syndrome. In severecases, cystsmay become very large and vulnerable to hem-orrhage or torsion, and surgical removal may be required,sometimes in infancy (Fig. 2). Hyperandrogenism in IRmaybe severe,with testosterone levels above 10nmol/litersometimes seen, well in excess of thresholds commonly

    reported to discriminate virilizing tumors from nontu-moral hyperandrogenism (22).

    The ovarian hyperandrogenism of IR is driven by syn-ergy between gonadotropin and insulin action on theovary (23, 24). Thus, it may be clinically apparent duringboth infancy and postpubertal life, when the hypothalam-ic-pituitary-gonadal axis is fully active, and it may alsoacceleratepuberty (25). Severeovarianhyperandrogenismmay occur postmenopausally, in which case ovarian his-tology characteristically reveals hyperthecosis, or hyper-plasia of the androgen-secreting theca cells (26).However,hyperandrogenism is not seen, despite extreme hyperin-sulinemia, when insulin receptor function is lost for thefirst time postmenopausally (12), suggesting that hyper-insulinemia around the time of the menopause may benecessary to sustain androgen-secreting theca cells.

    Themain differential diagnosis of IR-related severe hy-perandrogenism is congenital adrenal hyperplasia or anandrogen-secreting tumor, but in these cases acanthosisnigricans is not usually prominent unless the patient is alsooverweight or obese.Although congenital adrenal hyperpla-sia is easily diagnosed biochemically, discriminating auton-omous androgen secretion and hence virilizing tumors maybe more challenging (27). A complicating consideration isthat ovarian tumors may arise in the context of sustainedsevere hyperinsulinemia, most likely due to chronic activa-tion of IGF-I receptor-mediated signaling (28).

    C. Acanthosis nigricansAnother feature of nearly all known forms of severe IR

    is acanthosis nigricans, a velvety thickening of the skin. It

    Fig. 2. Ovarian appearances in severe IR. A, Ultrasound appearanceof an ovary in a 14-yr-old patient with severe IR due to a heterozygousmutation in the insulin receptor. B, Perioperative appearance of largeovarian and Fallopian tube cysts in a patient with digenic severe IR dueto heterozygous mutations in the PPARG and PPP1R3A genes (121). Cand D, Ovarian histology of the same patient, showing prominentsclerosis of the superficial ovarian cortex associated with multiplefollicle cysts (C) and stromal hyperthecosis with nests of eosinophilicluteinized cells (arrows) (D) embedded in hyalinized ovarian stroma.[Histological images courtesy of Dr. Merche Jimenez-Linan.]

    500 Semple et al. Severe Insulin Resistance Syndromes Endocrine Reviews, August 2011, 32(4):498514

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • is usually found in the axillae, nape of the neck, and groin,but it can occur in any flexures and in the most extremecases may be periocular, perioral, perianal, or even occuron planar surfaces (Fig. 3). It is commonly associatedwithacrochordons (skin tags). Histologically, acanthosis nig-ricans is characterized by hyperkeratosis, sometimes withhyperpigmentation, as well as mild papillomatosis, sug-gesting that both keratinocytes and dermal fibroblasts areaffected (29). The precise pathogenesis is unclear, but itmay also rarely be found in congenital syndromeswithoutIR (29) or as a paraneoplastic syndrome (30), and severallines of evidence suggest that enhanced signaling throughmitogenic tyrosine kinase-type receptors including theIGF-I receptor plays a central role (29, 31). In IR, acan-thosis depends on hyperinsulinemia, and this is not seen inthe rare situation of pre receptor IR due to unusuallyhigh levels of anti-insulin antibodies in those receivingexogenous insulin therapy (32). Fading of acanthosis in-dicates a reduction in insulin levels either due to lesseningof IR or, conversely, worsening of -cell failure. Acantho-sis nigricans may become excoriated and/or infected, andin concert with IR-related hyperandrogenism may con-tribute to hydradenitis suppurativa (33).

    IV. Clinical Features Limited to Some SevereIR Subtypes

    All the above are seen in severe IR irrespective of under-lying etiology, whether congenital or acquired, and

    whether due to a primary signaling defect or due to lipo-dystrophy. Some features of severe IR syndromes, in con-trast, are seen in only some subtypes.

    A. Dyslipidemia and hepatic steatosis

    Hypertriglyceridemia and lowhigh-density lipoproteincholesterol levels (hereafter designated metabolic dyslip-idemia) are closely associated with prevalent forms of IR(3437) and are seen in more severe form also in patientswith severe monogenic IR. Indeed, in some cases, hyper-triglyceridemia may be complicated by pancreatitis anderuptive xanthomata, and hepatic steatosis may progressto steatohepatitis, cirrhosis, andhepatocellular carcinoma(38). The presence of significant dyslipidemia and hepaticsteatosis is a sensitive but nonspecific clinical indicator ofunderlying lipodystrophy. Their absence in a patient withsevere IR is suggestive of a primary insulin receptoropathy(39, 40). Recently published mouse data (4143), sup-ported by our own observations in patients with severe IR(39), suggest that hepatic steatosis and dyslipidemia are aconsequence of selective postreceptor (or partial-) hepaticIR (44).

    B. Abnormal adipose development or topography

    Lipodystrophy is a common cause of severe IR andshould be considered in all cases. The fact that fat mass inlean women is close to double that in lean men, coupledwith the readily recognizable femorogluteal depot usuallypresent in women, generally means that lipodystrophy is

    Fig. 3. Acanthosis nigricans (AN) in severe IR. A, Severe AN on the neck in a prepubertal patient with autosomal dominant IR of unknown cause.B, AN associated with exuberant axillary acrochordons in a 50-yr-old male with severe IR of unknown cause. CF, AN in abdominal skin flexures ofa 15-yr-old boy with severe IR due to a heterozygous INSR mutation (C), on the foot of a patient with congenital generalized lipodystrophy andsevere IR due to homozygous AGPAT2 mutations (D), on the knuckles in a prepubertal patient with severe IR of unknown cause (E), and on theneck of a prepubertal girl with RMS due to a homozygous INSR mutation (F). G, Histological appearances from a nuchal skin biopsy showingcharacteristic papillomatosis (solid arrows), hyperkeratosis, and some acanthosis (open arrow). [Histological image courtesy of Dr. Ed Rytina.]

    Endocrine Reviews, August 2011, 32(4):498514 edrv.endojournals.org 501

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • more readily detected in women than men. Lean athleticmen can be very difficult to distinguish from lipodystro-phic men, particularly those with partial lipodystrophy.

    C. Growth disordersSevere IR per se may be associated with a range of

    growth disorders, including linear growth impairment(40, 45), prepubertal linear growth acceleration (46), orpseudoacromegalic soft tissue overgrowth in adult-hood (4750).Although the precisemolecular basis of thisclose association between severe IR and growth abnor-malities has yet to be determined, it may well relate toperturbation of the cross talk between the endocrine axiscontrolling growth and insulin action, or perhaps to ab-normal paracrine action of IGF-I or IGF-II (45). BothIGF-I, themajor hormone driving linear growth, and IGF-II, which is an important determinant of growth in utero,but which remains present at high levels in serum in post-natal life in humans but not rodents, exert mitogenic ef-fects through the IGF-I receptor and also have significantability to stimulate the insulin receptor. Insulin, con-versely, is able to stimulate the IGF-I receptor at concen-trations seen in extreme hyperinsulinemia (51, 52). Addedto this direct cross talk, insulin or IR has been shown in avariety of contexts to influence action of IGF through al-terations in expression of the ligands themselves, of theirbinding proteins, or of their receptors (53). These obser-vations have yet to be synthesized into a coherent modelaccounting for growth abnormalities in severe IR; how-ever, improving understanding of these phenomena willnot only be of relevance to these rare conditions, but mayalso give mechanistic insights into the link between com-mon obesity/IR and both the prevalence and prognosis ofa variety of different cancers (52, 54, 55).

    V. Sexual Dimorphism in Severe IR

    Aprominent feature of severe IR is the earlier presentationand more severe metabolic derangement seen in affectedwomen (56). Amajor contributor to this is hyperinsuline-mia-driven ovarian dysfunction, with hyperandrogenismand oligomenorrhea serving as clinical red flags that leadto early presentation (57). Men, in contrast, exhibit onlyacanthosis nigricans, and sometimes symptomatic post-prandial hypoglycemia. Even if noticed, these are muchless likely to lead to medical consultation. However,women also havemuchmore severe hyperinsulinemia anddyslipidemia than men. In lipodystrophy, this is almostcertainly explained at least in part by the larger amount ofwhite adipose tissue as a proportion of total body mass inhealthy women.

    VI. Biochemical Subphenotyping of Severe IR

    Growing evidence suggests that some syndromes of se-vere IR exhibit different patterns of IR among insulin-responsive tissues and pathways. Thus, the generalized IRof insulin receptor (INSR) defects is associatedwith a nor-mal lipid profile and relative lack of fatty liver (39, 40),suggesting that some insulin signaling is needed to drivehepatic fat synthesis and secretion. This is quite unlike thesituation in patients with lipodystrophy or with defects inthe insulin signal transducer AKT2, all of whom showsevere dyslipidemia and fatty liver (39).

    The mechanisms linking hyperinsulinemia in prevalentforms of IR and fatty liver/dyslipidemia are of particularimportance, given the enormous associated prevalence ofatherosclerosis, and are the subject of intense investiga-tion. Full consideration of progress in the field is beyondthe scope of this article; however, recent cell-based studieshave strongly implicated activation of themammalian tar-get of rapamycin complex 1 (mTORC1), hitherto widelyperceived to be predominantly a mediator of insulinsactions on cell growth, in driving hepatic de novo lipo-genesis in response to insulin (5860). Such up-regu-lation of hepatic lipogenesis has been suggested to be asignificant contributor to fatty liver and atherogenicdyslipidemia in humans (61, 62), whereas aberrant ac-tivation of mTORC1 is well documented in IR (63),potentially explaining why lipogenesis appears to be in-creased in these states. However, many critical questionsremain to be answered, including whether enhanced denovo lipogenesis in IR may truly be explained as a hepa-tocyte autonomous phenomenon related to resistance toonly some of insulins cellular effects, or whether it mayinstead reflect parallel hyperactivation of mTORC1 byincreased delivery to hepatocytes of, for example,branched chain amino acids, which have been reported tobe elevated in many forms of IR (59, 64, 65).

    INSR defects may also be discriminated from otherforms of IR by unexpectedly high adiponectin (6668),SHBG (69), and IGF binding protein-1 (IGFBP1) (70) lev-els, providing further evidence that in prevalent forms ofIR (7173), in lipodystrophies (15, 67), and in nonrecep-toropathy severe IR (70, 74), where the levels of theseproteins are usually reduced, hyperinsulinemia is able toexert effects through intact elements of the cellular insulinsignal transduction network to suppress gene expressionin both adipose tissue and liver (62, 75). Parenthetically,because adiponectin is nearly exclusively expressed in ad-ipose tissueandbecause circulatingSHBGand IGFBP1areproducts of hepatic expression, determination of thesemarkers in states of IR may also allow assessment to bemade of the insulinization of distinct insulin target tissues

    502 Semple et al. Severe Insulin Resistance Syndromes Endocrine Reviews, August 2011, 32(4):498514

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • in future, enhancing clinical ability to discern any organ-selective forms of severe IR.

    This concept of IR subphenotypes has been exploiteddiagnostically by using it to subclassify IR and target ge-netic screening, greatly enhancing efficiency of moleculardiagnosis of INSR defects (68, 70). Thus, in the context ofsevere IR, we have found that adiponectin levels above 7mg/liter have a 97% positive predictive value for insulinreceptoropathy (70), although the precise cutoff is assay-specific. SHBG and IGFBP1 levels have lesser, althoughstill significant, utility.

    VII. Monogenic IR Classification/Nomenclature

    The nomenclature in the field of severe IR dates from the1970s. Then, in seminal publications, Kahn and col-leagues (9, 16) designated severe IR in nonobese patientsas type A or type B, the latter discriminated by thepresence of anti-insulin receptor antibodies. In a series ofindependent publications around the same time, the termHAIR-AN came to be used commonly (76). However,HAIR-AN (hyperandrogenism, insulin resistance, andacanthosis nigricans) is an entirely generic description ofsevere IR inwomen. If it has anyutility, it iswhere it is usedto discriminate women with severe IR who also have abodymass index above 30 kg/m2,who aremuch less likelyto harbor pathogenic single gene defects than their nono-bese counterparts. However, the imprecise and overlap-ping usage of these different diagnostic terms and increas-ing understanding of subgroups of severe IR suggest thata reclassification of syndromes of severe IRmay be timely.Based on the above observations, such a classification issuggested in Table 1.

    The key subdivision in this proposedmechanism-basedclassification is between those disorders in which there isa primary defect in canonical insulin signal transductionand those in which severe IR is a consequence of adiposetissue abnormalities, or adipose failure. Primary IR isthen subdivided into generalized IR, in which there is adefect at the level of the insulin receptor, and biochemi-cally distinguishable partial IR, in which there is a sig-naling defect that is limited either to only some parts of thepostreceptor signal transduction pathway or to some tis-sues. Few examples of this group currently exist, but it isto be anticipated that increased recognition of this group,allied tomodern sequencing technologies, will lead to fur-ther examples and refinement of this subgroup.

    Adipose failure may also be subdivided into a groupwithmanifest lipodystrophy, inwhich there is a deficiencyin generating adipose tissue, leading to severe IR despitelow or normal adipose tissue mass, and a group in whichthe dominant defect is unrestrained accumulation of ad-ipose tissue, most commonly due to hyperphagia, suchthat even a relatively normal capacity safely to accrue tri-glyceride in adipose tissue is overcome.

    Severe IR is also seen in a groupof complex disorders(see Table 3), in association with other defects; however,because the IR in these conditions is subjected to mecha-nistic study with the above framework in mind, we antic-ipate that they may be accommodated within one of thetwo main groups.

    VIII. The INSR Spectrum

    The first defects in the INSR were reported in 1988 (77,78), shortly after cloning of the human gene (79), and

    TABLE 1. Proposed new classification for syndromes of severe IR

    Discriminating features

    I. Primary insulin-signaling defectsA. Generalizeda INSR mutations (40) or anti-INSR antibodies (12) Extreme hyperinsulinemia but normal lipid profile (39, 40),

    preserved or elevated adiponectin, SHBG, and IGFBP1 (70)B. Partialb AKT2 (19), AS160 (91, 169), others to be defined Likely to depend upon precise signaling defect

    II. Secondary to adipose tissue abnormalitiesc

    A. Severe obesity e.g., MC4R (170), POMC (171), LEP (126), LEPR (172),SH2B1 (173)

    Early onset severe, hyperphagic obesityTall stature (MC4R)Hypogonadotropic hypogonadism (LEP)Red hair and hypoadrenalism (POMC)Disproportionate IR SH2B1 (173)

    B. Lipodystrophy (generalized or partial, Table 2) Congenitally absent adipose tissue, or regional deficiency ofadipose tissue

    Usually severe dyslipidemia, fatty liverLow adiponectin and leptin levels

    a An alternative term for this cluster of disorders is insulin receptoropathies.b Affecting only some intracellular arms of the insulin signaling pathway, or variable among tissues.c In addition to frank obesity or lipodystrophy, there is a less well-defined group of disorders having clinical and biochemical evidence of adipose tissue failure andsevere dyslipidemia despite grossly normal whole body adipose tissue mass.

    Endocrine Reviews, August 2011, 32(4):498514 edrv.endojournals.org 503

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • more than 100 allelic variants have now been described.These genetic insulin receptoropathies form a continuumof clinical severity but are best divided into two groups(Fig. 4). The first consists of rare and severe autosomalrecessive (AR) disorders presenting in the first decade oflife and usually classified, arbitrarily, as Donohue syn-drome (DS; formerly leprechaunism) or Rabson Men-denhall syndrome (RMS), based on the original clinicaldescriptions (80, 81). These syndromes have been welldescribed (40, 82). As well as fasting hypoglycemia, post-prandial hyperglycemia, and extreme hyperinsulinemia,their dominant features are markedly retarded lineargrowth, impairedmuscle and adipose tissue development,and overgrowth or precocious development of sex hor-mone-dependent tissues such as genitalia and nipples, andof other tissues including hair, skin, and viscera (Fig. 4).When -cells decompensate, hyperglycemia may becomerefractory to treatment. InDS, deathusually occurs duringintercurrent infection in infancy, whereas in RMS, ad-vanced microvascular complications or diabetic ketoaci-dosis are the commonest modes of death, usually in thesecond or third decade (40).

    Some aspects of the DS phenotype remain to be fullyexplained. In particular, it remains to be determined de-finitivelywhy affected infants are resistant to ketoacidosisat least in the first year of life, even in those with no func-tional insulin receptor, although suggestions include con-tinued action of extremely elevated insulin on persistinghepatic IGF-I receptors in the immature liver or deficiencyof GH secretion or action (83).

    More commonly, INSR defects present peripubertallyas oligomenorrhea and hyperandrogenism with acantho-sis nigricans (40). At presentation, hyperglycemia has of-ten yet to develop. In the prediabetic phase, males exhibitonly acanthosis nigricans and sometimes hypoglycemia,and they often remain undiagnosed even after the devel-opment of symptomatic diabetes, which may not occuruntil the fourth decade or beyond.

    In some patients with DS or RMS, loss of INSR ex-pression from one allele has been identified and has beeninferred to be due to a mutation in a regulatory sequencesuch as the promoter (84). However, four patients withsevere IR and extremely low expression of both alleles ofthe INSR gene have also been described harboring eithera heterozygous deletion ormutation of theHMGA1 gene,but no mutations in the INSR gene (85). HMGA1 is anarchitectural transcription factor that binds to key sites inthe promoter of the INSR gene to facilitate its transcrip-tion. Based on the single report to date, these patientsappeared to share many characteristics of patients withINSR defects, consistent with the notion that a key func-tion for HMGA1 is the stimulation of INSR expression.

    IX. Downstream Insulin Signaling Defects

    Rapid progress in elucidating the key components of theinsulin signaling pathway in the early 1990s raised hopethat defects may be found in the genes encoding thesesignaling elements in patients with severe IR but withoutINSR mutations. However, few such sequence variantshave been reported in severe IR, and in most cases theresulting signaling defects in vitro have been subtle at best(8690).

    One exceptionwas a single family in which threemem-bers carried a nonfunctional, heterozygous mutation inAKT2, encoding a critical serine/threonine kinase down-stream from the INSR in the signal transduction pathway(19). Clinical features seen in affected family membersincluded acanthosis nigricans, ovarian hyperandro-genism, diabetes mellitus presaged by several years ofpostprandial hypoglycemia, metabolic dyslipidemia, andfatty liver (19). The female proband also exhibited partiallipodystrophy, highlighting the role of insulin in adipo-genesis and the need for awareness that primary defects inthe insulin signaling cascade may impair adipose tissuegeneration in vivo. However, although a generalized de-ficiency in adipose tissue development is seen in severeinsulin receptoropathies, the metabolic characteristics ofthis differ dramatically fromthefat failurephenotypeofprimary generalized lipodystrophy, and they may moreappropriately be regarded as states of nonlipidated ratherthan absent adipose tissue.

    More recently, a heterozygous nonsense mutation inAS160, a smallGTPase-activatingprotein that formsakeylink between insulin signaling and glucose uptake by theGLUT4 transporter, has been described in a family inwhomaffectedmembers had acanthosis nigricans anddis-proportionate hyperinsulinemia after a glucose challenge(91). Surprisingly, no other mutations have been found to

    Fig. 4. Clinical spectrum of insulin receptoropathies.

    504 Semple et al. Severe Insulin Resistance Syndromes Endocrine Reviews, August 2011, 32(4):498514

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • date in other, more distal signaling components involvedin GLUT4 transporter translocation to the cell membranein response to insulin.

    X. Disorders of Adipose Tissue Development/Function (Lipodystrophies)

    Far more success has been had in identifying primary de-fects in adipose tissue that lead to severe IR as a secondaryconsequence. This probably reflects the fact that lipodys-trophy, in contrast to many other forms of severe IR, isrelatively easily recognized clinically, facilitating identifi-cation of extended families with multiple affected mem-bers for genetic studies (9296). The possibility of lipo-dystrophy should be carefully considered in all severelyinsulin-resistant patients with dyslipidemia and/or nonal-coholic fatty liver disease.

    Lipodystrophy is a heterogeneous disorder character-ized by pathological adipose tissue deficiency. The lack offatmay be partial or generalized and inherited or acquiredin origin. Themolecular pathogenesis and clinical featuresof lipodystrophy have recently been reviewed (15, 97

    101), are not covered in detail here, but are summarized inTable 2. Within the past 2 yr, three novel subtypes ofcongenital lipodystrophy were identified by a candidategene approach. Biallelic nonsense mutations in CAV1(102) and PTRF (103106) were identified in patientswith generalized lipodystrophy and inCIDEC in a patientwith partial lipodystrophy (107). Caveolins are essentialfor the formation of caveolae, which are abundant in adi-pocytes and appear to play a role in fatty acid uptake,insulin receptor signaling (108), and lipid droplet forma-tion (109). PTRF (polymerase 1 and transcript release fac-tor) stabilizes caveolins 13 and is required for the for-mation of caveolae (110, 111). CAV3 mutations areknown to cause muscular dystrophy (112), which prob-ably explains why PTRF mutations were also associatedwith a muscular dystrophy phenotype.

    One female patient with partial lipodystrophy (affect-ing limb, femorogluteal, and sc abdominal fat), white adi-pocytes with multiloculated lipid droplets, and insulin-resistant diabetes was found to be homozygous for apremature truncation mutation in the lipid droplet pro-tein,CIDEC (cell death-inducingDNA fragmentation fac-

    TABLE 2. Classification and clinical features of lipodystrophies

    Inheritance Major clinical features Ref.

    CGL Common features: severe IR, T2DM, severe dyslipidemia,fatty liver, pseudoacromegaly, PCOS

    15

    AGPAT2a AR Adiponectin levels are particularly low in this form ofCGL, whereas they are slightly higher (although stilllower than the reference range) in BSCL2-associatedCGL

    67, 174

    BSCL2a AR See note above 174CAV1a AR (single case) Short stature 102PTRFa AR Muscular dystrophy, modest metabolic disturbance (?) 103106

    Familial partial lipodystrophy Common features: IR, T2DM, dyslipidemia, fatty liver,PCOS

    LMNAa AD Preserved/excess facial and neck fat 15, 175PPARGa AD Preserved abdominal fat, hypertension (?) 18, 176, 177179ZMPSTE24a AR Mandibuloacral dysplasiaAKT2a AD (single family) 19CIDECa AR (single case) Preserved facial and neck fat, multiloculated lipid

    droplets107

    Acquired generalizedlipodystrophy

    Common features: severe IR, T2DM, severe dyslipidemia,fatty liver, pseudoacromegaly, PCOS

    14, 15

    Associated with otherautoimmune diseases;commonly alsoassociated with low C4complement levels

    N/A May be associated with juvenile dermatomyositis, SLE,autoimmune hemolytic anemia, autoimmunehepatitis. The low C4 complement subgroup isparticularly associated with autoimmune hepatitis andautoimmune hemolytic anemia.

    11, 13, 14

    Acquired partial lipodystrophy 14HIV-associated

    lipodystrophyN/A Not typically associated with severe IR but is

    associated with IR, dyslipidemia, fatty liver180

    C3 nephritic factorassociated

    N/A Cephalocaudal pattern of fat loss, low C3 complementlevels, MPGN, not usually insulin resistant, althoughmay become so if overweight

    14

    AD, Autosomal dominant; CGL, congenital generalized lipodystrophy; C3/4, complement factor 3/4; T2DM, type 2 diabetes mellitus; PCOS, polycystic ovary syndrome;MPGN, mesangioproliferative glomerulonephritis; N/A, not applicable; SLE, systemic lupus erythematosus.a Genetic subtype.

    Endocrine Reviews, August 2011, 32(4):498514 edrv.endojournals.org 505

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • torA-like effector family-C) (107).Cidecknockdowncellsmanifest multiloculated lipid droplets with increased mi-tochondria (113), and in mice, Cidec deficiency also re-duces fat mass and induces the formation of white adi-pocytes with multilocular lipid droplets (114, 115).However, in contrast to the human phenotype associatedwith a homozygous loss of function mutation in CIDEC,Cidec null mice are protected against diet-induced obesityand IR (114116).BSCL2, an enigmatic geneof unknownfunction in which homozygous mutations were the firstidentified cause of congenital lipodystrophy (95), has alsorecently been implicated in lipid droplet biogenesis (117119) and adipocyte differentiation (120).

    XI. Digenic IR

    In 2002 (121),we described a family inwhich five severelyinsulin-resistant subjects and no unaffected relativeswere doubly heterozygous for frameshift/premature stopmutations in two unlinked genes, namely PPARG, a keyregulator of adipocyte biology, and PPP1R3A, a muscle-specific protein involved in regulating glycogen turnover(122125). This report was of particular interest becausethe observation that genetic defects inmolecules primarilyinvolved in either lipid or carbohydrate metabolism cancombine to result in an extreme phenotype of IR providesa model for the type of metabolic interaction that mayunderlie common forms of IR/type 2 diabetes.

    XII. Complex Syndromes

    Several genetic syndromes feature severe IR as part of awider constellation of abnormality (Table 3). In many of

    these, the IR is related to severe obesity. However, thisgroup may roughly be divided into those conditions inwhich IR does not seem to be disproportionate to the de-gree of excess adiposity, such as genetically severe hy-perphagia due to congenital leptin deficiency (126), andthose in which IR appears unusually severe, suggesting arole for the defective genes concerned in systemic insulinsensitivity as well as in hypothalamic appetite control.However, in most cases this issue has not been rigorouslystudied. One well-established example is Alstrom syn-drome, which is due to genetic defects in the large centro-somal ALMS1 protein, where predominantly centripetaladiposity is associated with severely disproportionate IRand dyslipidemia (127).

    Although the molecular pathogenesis of severe IR inAlstrom syndrome is not clear, it is notable that defects inthepericentrosomalproteinpericentrin, causingosteodys-plastic primordial dwarfism of Majewski type 2, are alsoassociated with highly penetrant severe IR (128), whichmay also be true in many cases of Bardet Biedl syndrome(129), caused by defects in a variety of proteins involvedin basal body/centrosomal function (130). Collectively,these observations hint at an important role for the cen-trosome or basal body, or the cellular functions they sub-serve, in maintaining metabolic homeostasis.

    Another notable group of disorders that feature dis-proportionate and often severe IR are associated withDNA repair defects and/or progeria, including Wernersyndrome (131, 132), Bloom syndrome (133), and man-dibuloacral dysplasia (97, 134). In further DNA repairdefects such as ataxia telangiectasia, severe IR has beenreported in several molecularly defined cases (135, 136).However, the precise mechanism of severe IR in these set-tings has yet to be established.

    TABLE 3. Selected complex genetic disorders associated with severe IR

    Syndrome Gene(s)Adipose tissuephenotype

    IR disproportionateto adiposity?

    Cellular component orfunction affected

    Alstrm ALMS1 Centripetal obesity Yes (127) Centrosome/ basal bodyMOPDII PCNT Centripetal fat

    distributionYes (128) Centrosome/basal body

    Bardet Biedl BBS1, BBS2, ARL6, BBS4, BBS5,MKKS, BBS7, BBS8, BBS9,BBS10, BBS11, BBS12,MKS1, CEP290

    Obesity Unclear (129) Centrosome/basal body (130)

    Bloom RECQ2 Lipodystrophy Yes (133) DNA repairWerner RECQL2 Lipodystrophy Yes (131132) DNA repair

    LMNAMandibuloacral

    dysplasiaLMNA Lipodystrophy Yes (97, 134) Involved in formation of nuclear

    laminaZMPSTE24

    Myotonicdystrophy

    DMPK None Yes (181) Transcriptional/splicing regulationon chromosome 19, includingthe INSR (182)

    MOPDII, Osteodysplastic primordial dwarfism of Majewski type II.

    506 Semple et al. Severe Insulin Resistance Syndromes Endocrine Reviews, August 2011, 32(4):498514

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • XIII. Therapy

    The rarity and underdiagnosis of severe IR means thatalmost all therapeutic decisions are based on rational tar-geting of underlying defects and anecdotal evidence ratherthan randomized controlled trials. Therapy aims to reducehyperglycemia, to ameliorate dyslipidemia, and to lessenthe sometimes debilitating reproductive and cosmetic con-sequences of hyperinsulinemia. This is achieved firstthrough mitigation of the underlying signaling defect,through minimizing secretory demands on the pancreatic-cells, and throughoptimal delivery of exogenous insulinwhen required. In the context of adipose tissue absence ordysfunction, offloading adipose tissue by reducing lipiddelivery to it is also critical, and, finally, countering hy-perandrogenism, ovulatory dysfunction, and acanthosisnigricans by measures targeted at the relevant end organsis also often of value. The treatment of such secondarymanifestations of the IR state has been reviewed elsewhere(137, 138).

    A. Dietary and lifestyle modificationIn severe IR, whether or not a single gene defect is iden-

    tified, weight gain inevitably exacerbates metabolic de-rangement and either worsens hyperglycemia in patientswith overt diabetes or increases -cell stress, so restrictingenergy intake and maximizing aerobic exercise are essen-tial elements of management. This is particularly impor-tant in lipodystrophy where the apparent leanness of pa-tients frequently results in a failure of caregivers to placesufficient emphasis on dietary modification. Indeed, fail-ure to restrict energy intake in patientswith lipodystrophymakes it almost impossible to obtain good glycemic andlipidemic control.

    B. Insulin sensitization and replacementInsulin-sensitizing agents also play a key role in man-

    agement. Metformin is often effective, and in some casesthiazolidinediones also exert markedly beneficial effects,but no comparative studies exist to guide the choice oftherapy in different subgroups of severe IR. When -celldecompensation occurs in severe IR to produce diabetes,this is only relative to the very high insulin requirements,and plasma insulin levels remain extremely elevated. Thismeans that insulin secretagogues such as sulfonylureas of-ten produce little benefit. When insulin is required, thismay need to be used in concentrated form to achieve met-abolic control (139), and limited evidence suggests thatdelivery by sc infusion may be efficacious (140). Use ofrecombinant human IGF-I has been reportedmostly in thesetting of severe insulin receptoropathies and appears toimprove glycemia and perhaps survival in some infantilecases (141151). It may exert activity by acting as an in-

    sulin mimetic, as a trophic factor for pancreatic -cells, orby enhancing insulin sensitivity through postreceptorcross talk between insulin and IGF-I signaling pathways.However, its dominant mode of action, optimal dosing,and clinical indications remain unclear.

    C. Adipose tissue offloading

    Minimizing caloric intake and hence strain on adiposestorage capacity is particularly difficult in lipodystrophybecause either absoluteor relative leptindeficiency, in gen-eralized and partial lipodystrophy, respectively, leads tohyperphagia (152). Recombinant leptin therapy substan-tially reduces food intake in this setting and dramaticallyimproves dyslipidemia, hepatic steatosis, and glycemiccontrol (153157). The response to leptin and dose titra-tion is largely based on clinical criteria because many pa-tients develop antibodies that interfere with leptin assays(158) but do not, on the whole, appear to reduce its long-term efficacy (159). Leptin has been used in all prevalentforms of generalized and familial partial lipodystrophy,with particularly dramatic results in the former (157). Ini-tial reports also suggest that it may be useful in at leastsome cases ofHIV-associated partial lipodystrophy (160),whereas a single study has also reported benefits in RMS(161). In principle, increasing oxidative catabolism of ex-cess calories may achieve the same benefits as limitingintake, as illustrated by the dramatically beneficial effectof suppressive doses of T4 in a patientwith an INSR defectwho also had a papillary thyroid carcinoma (162), but thisstrategy has yet to be developed safely for more wide-spread application.

    Given the importance of restricting energy intake, par-ticularly in patients with lipodystrophy, other weight losstherapies used in obese diabetic patients, including gluca-gon-like peptide-1 agonists and appetite suppressants,have the potential to produce clinical benefits (163), andscattered reports have even suggested that bariatric sur-gery can be helpful in severe cases (164).

    A complementary strategy to reducing the energy inputinto adipose tissue is to increase its storage capacity byusing insulin-sensitizing thiazolidinedione peroxisomeproliferator-activated receptor agonists. These were anobvious choice, particularly in lipodystrophywhere it washoped that they might restore fat mass. However, thiazo-lidinediones are not helpful in generalized lipodystrophyand exacerbate hepatic steatosis in animal models (165),and reports of their use in partial lipodystrophy conflict(166168).Our ownexperience is similar to that of Simhaet al. (167), who noted that fat tended to accumulate inresidual adipose depots, with modest metabolic benefits.

    Endocrine Reviews, August 2011, 32(4):498514 edrv.endojournals.org 507

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • XIV. Summary

    Genetic syndromes of severe IR, with or without lipodys-trophy, are underrecognized conditions that exact an im-mense toll of early morbidity and mortality for those af-fected. Considerable progress over the past 20 yr inidentifying the molecular basis of these disorders nowpresents the opportunity for trials or therapy targeted atspecific subgroups of these patients. It is anticipated thatthis will not only improve clinical outcomes for these rarepatients but will also give insights into the pathophysiol-ogy and therapy of more prevalent forms of IR.

    Acknowledgments

    Address all correspondence and requests for reprints to:Dr.R.K. Sempleor Dr. D. B. Savage, Metabolic Research Laboratories, Institute of Met-abolic Science, University of Cambridge, Addenbrookes Hospital, HillsRoad, Cambridge CB2 0QQ, United Kingdom. E-mail: [email protected] or [email protected].

    This work was supported by research grants from the WellcomeTrust (Intermediate Clinical Fellowship 080952/Z/06/Z, to R.K.S.; Pro-gramme Grant 078986/Z/06/Z, to S.O.), GlaxoSmithKline (to D.B.S.),the UK National Institute for Health Research Cambridge BiomedicalResearch Centre, and the UKMedical Research Council Centre forObe-sity and Related Metabolic Disease.

    Disclosure Summary: R.K.S., D.B.S., E.K.C., and P.G. have nothingto disclose. S.O. acts as a consultant in drug discovery for GlaxoSmith-Kline, Pfizer, and OSI Pharmaceuticals, Inc.

    References

    1. ResnickHE, JonesK,RuotoloG, JainAK,Henderson J, LuW,Howard BV 2003 Insulin resistance, themetabolic syn-drome, and risk of incident cardiovascular disease in non-diabetic American Indians: the Strong Heart Study. Dia-betes Care 26:861867

    2. HansonRL, ImperatoreG,Bennett PH,KnowlerWC2002Components of the metabolic syndrome and incidenceof type 2 diabetes. Diabetes 51:31203127

    3. Dunaif A, Segal KR, Futterweit W, Dobrjansky A 1989Profoundperipheral insulin resistance, independentofobe-sity, in polycystic ovary syndrome. Diabetes 38:11651174

    4. Reaven GM 1988 Banting lecture 1988. Role of insulinresistance in human disease. Diabetes 37:15951607

    5. Souren NY, Paulussen AD, Loos RJ, Gielen M, Beunen G,Fagard R, Derom C, Vlietinck R, Zeegers MP 2007 An-thropometry, carbohydrate and lipid metabolism in theEast Flanders Prospective Twin Survey: heritabilities. Dia-betologia 50:21072116

    6. Freeman MS, Mansfield MW, Barrett JH, Grant PJ 2002Heritability of features of the insulin resistance syndromein a community-based study of healthy families. DiabetMed 19:994999

    7. Mills GW, Avery PJ, McCarthy MI, Hattersley AT, LevyJC, Hitman GA, SampsonM,WalkerM 2004Heritability

    estimates for -cell function and features of the insulin re-sistance syndrome in UK families with an increased sus-ceptibility to type 2 diabetes. Diabetologia 47:732738

    8. FalchiM,Wilson SG, PaximadasD, SwaminathanR, Spec-tor TD 2008 Quantitative linkage analysis for pancreaticB-cell function and insulin resistance in a large twin cohort.Diabetes 57:11201124

    9. Kahn CR, Flier JS, Bar RS, Archer JA, Gorden P, MartinMM, Roth J 1976 The syndromes of insulin resistance andacanthosis nigricans. Insulin-receptor disorders in man.N Engl J Med 294:739745

    10. Sissons JG, West RJ, Fallows J, Williams DG, Boucher BJ,Amos N, Peters DK 1976 The complement abnormalitiesof lipodystrophy. N Engl J Med 294:461465

    11. Savage DB, Semple RK, ClatworthyMR, Lyons PA, Mor-gan BP, Cochran EK, Gorden P, Raymond-Barker P,Mur-gatroyd PR, Adams C, Scobie I, Mufti GJ, Alexander GJ,Thiru S, Murano I, Cinti S, Chaudhry AN, Smith KG,ORahilly S 2009 Complement abnormalities in acquiredlipodystrophy revisited. J Clin Endocrinol Metab 94:1016

    12. AriogluE,AndeweltA,DiaboC,BellM,TaylorSI,GordenP 2002 Clinical course of the syndrome of autoantibodiesto the insulin receptor (type B insulin resistance): a 28-yearperspective. Medicine (Baltimore) 81:87100

    13. Misra A, Garg A 2003 Clinical features and metabolic de-rangements in acquired generalized lipodystrophy: case re-ports and review of the literature. Medicine (Baltimore)82:129146

    14. Misra A, Peethambaram A, Garg A 2004 Clinical featuresand metabolic and autoimmune derangements in acquiredpartial lipodystrophy: report of 35 cases and review of theliterature. Medicine (Baltimore) 83:1834

    15. Garg A 2004 Acquired and inherited lipodystrophies.N Engl J Med 350:12201234

    16. Flier JS, Kahn CR, Roth J 1979 Receptors, antireceptorantibodies and mechanisms of insulin resistance. N EnglJ Med 300:413419

    17. Hjlund K, Hansen T, Lajer M, Henriksen JE, Levin K,Lindholm J, Pedersen O, Beck-Nielsen H 2004 A novelsyndrome of autosomal-dominant hyperinsulinemic hypo-glycemia linked to a mutation in the human insulin recep-tor gene. Diabetes 53:15921598

    18. Agostini M, Schoenmakers E, Mitchell C, Szatmari I, Sav-age D, Smith A, Rajanayagam O, Semple R, Luan J, BathL,ZalinA,LabibM,KumarS, SimpsonH,BlomD,MaraisD, Schwabe J, Barroso I, Trembath R, Wareham N, NagyL, Gurnell M, ORahilly S, Chatterjee K 2006 Non-DNAbinding, dominant-negative, human PPAR mutationscause lipodystrophic insulin resistance. CellMetab 4:303311

    19. George S, Rochford JJ, Wolfrum C, Gray SL, Schinner S,Wilson JC, Soos MA, Murgatroyd PR, Williams RM,Acerini CL, Dunger DB, Barford D, Umpleby AM, Ware-ham NJ, Davies HA, Schafer AJ, Stoffel M, ORahilly S,Barroso I 2004 A family with severe insulin resistance anddiabetes due to a mutation in AKT2. Science 304:13251328

    20. Kotronen A, Juurinen L, Tiikkainen M, Vehkavaara S,Yki-Jarvinen H 2008 Increased liver fat, impaired insulinclearance, and hepatic and adipose tissue insulin resistancein type 2 diabetes. Gastroenterology 135:122130

    508 Semple et al. Severe Insulin Resistance Syndromes Endocrine Reviews, August 2011, 32(4):498514

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • 21. Okada T, Liew CW, Hu J, Hinault C, Michael MD,Krtzfeldt J, Yin C, Holzenberger M, Stoffel M, KulkarniRN 2007 Insulin receptors in -cells are critical for isletcompensatory growth response to insulin resistance. ProcNatl Acad Sci USA 104:89778982

    22. Kaltsas GA, Isidori AM, Kola BP, Skelly RH, Chew SL,Jenkins PJ, Monson JP, Grossman AB, Besser GM 2003The value of the low-dose dexamethasone suppression testin the differential diagnosis of hyperandrogenism inwomen. J Clin Endocrinol Metab 88:26342643

    23. Poretsky L, Cataldo NA, Rosenwaks Z, Giudice LC 1999The insulin-relatedovarian regulatory system inhealth anddisease. Endocr Rev 20:535582

    24. Nestler JE, Jakubowicz DJ 1996 Decreases in ovarian cy-tochrome P450c17 activity and serum free testosteroneafter reduction of insulin secretion in polycystic ovary syn-drome. N Engl J Med 335:617623

    25. Musso C, Shawker T, Cochran E, Javor ED, Young J,Gorden P 2005 Clinical evidence that hyperinsulinaemiaindependent of gonadotropins stimulates ovarian growth.Clin Endocrinol (Oxf) 63:7378

    26. Dunaif A, Hoffman AR, Scully RE, Flier JS, Longcope C,Levy LJ, Crowley Jr WF 1985 Clinical, biochemical, andovarian morphologic features in women with acanthosisnigricans and masculinization. Obstet Gynecol 66:545552

    27. KaltsasGA,Mukherjee JJ, KolaB, IsidoriAM,Hanson JA,Dacie JE, Reznek R, Monson JP, Grossman AB 2003 Isovarian and adrenal venous catheterization and samplinghelpful in the investigation of hyperandrogenic women?Clin Endocrinol (Oxf) 59:3443

    28. Brisigotti M, Fabbretti G, Pesce F, Gatti R, Cohen A,ParentiG,CalleaF1993Congenital bilateral juvenile gran-ulosa cell tumor of the ovary in leprechaunism: a case re-port. Pediatr Pathol 13:549558

    29. Torley D, Bellus GA, Munro CS 2002 Genes, growth fac-tors and acanthosis nigricans. Br J Dermatol 147:10961101

    30. Ellis DL, Kafka SP, Chow JC, Nanney LB, Inman WH,McCadden ME, King Jr LE 1987 Melanoma, growth fac-tors, acanthosis nigricans, the sign of Leser-Trelat, andmultiple acrochordons. A possible role for -transforminggrowth factor in cutaneous paraneoplastic syndromes.N Engl J Med 317:15821587

    31. Hermanns-Le T, Scheen A, Pierard GE 2004 Acanthosisnigricans associated with insulin resistance: pathophysiol-ogy and management. Am J Clin Dermatol 5:199203

    32. Greenfield JR,Tuthill A, SoosMA, SempleRK,HalsallDJ,Chaudhry A, ORahilly S 2009 Severe insulin resistancedue to anti-insulin antibodies: response to plasma ex-change and immunosuppressive therapy. Diabet Med 26:7982

    33. Barth JH, Ng LL, Wojnarowska F, Dawber RP 1988 Ac-anthosis nigricans, insulin resistance and cutaneous viril-ism. Br J Dermatol 118:613619

    34. Utzschneider KM, Kahn SE 2006 Review: the role of in-sulin resistance in nonalcoholic fatty liver disease. J ClinEndocrinol Metab 91:47534761

    35. Bellentani S, Saccoccio G, Masutti F, Croce` LS, Brandi G,Sasso F, Cristanini G, Tiribelli C 2000 Prevalence of andrisk factors for hepatic steatosis in northern Italy. Ann In-tern Med 132:112117

    36. Bonora E, Kiechl S, Willeit J, Oberhollenzer F, Egger G,Meigs JB, Bonadonna RC, Muggeo M 2004 Population-based incidence rates and risk factors for type 2 diabetes inwhite individuals: the Bruneck study. Diabetes 53:17821789

    37. Brown CD, Higgins M, Donato KA, Rohde FC, GarrisonR, Obarzanek E, Ernst ND, Horan M 2000 Body massindex and the prevalence of hypertension and dyslipide-mia. Obes Res 8:605619

    38. Starley BQ, CalcagnoCJ,Harrison SA 2010Nonalcoholicfatty liver disease and hepatocellular carcinoma: a weightyconnection. Hepatology 51:18201832

    39. SempleRK, SleighA,MurgatroydPR,AdamsCA,BluckL,JacksonS,VotteroA,KanabarD,Charlton-MenysV,Dur-rington P, Soos MA, Carpenter TA, Lomas DJ, CochranEK, Gorden P, ORahilly S, Savage DB 2009 Postreceptorinsulin resistance contributes to human dyslipidemia andhepatic steatosis. J Clin Invest 119:315322

    40. Musso C, Cochran E, Moran SA, Skarulis MC, Oral EA,Taylor S,GordenP2004Clinical course of genetic diseasesof the insulin receptor (type A and Rabson-Mendenhallsyndromes): a 30-year prospective. Medicine (Baltimore)83:209222

    41. Shimomura I, Matsuda M, Hammer RE, Bashmakov Y,Brown MS, Goldstein JL 2000 Decreased IRS-2 and in-creased SREBP-1c lead tomixed insulin resistance and sen-sitivity in livers of lipodystrophic and ob/obmice.MolCell6:7786

    42. Biddinger SB, Hernandez-Ono A, Rask-Madsen C, HaasJT, Aleman JO, Suzuki R, Scapa EF, Agarwal C, CareyMC, Stephanopoulos G, Cohen DE, King GL, GinsbergHN, Kahn CR 2008Hepatic insulin resistance is sufficientto produce dyslipidemia and susceptibility to atheroscle-rosis. Cell Metab 7:125134

    43. BrownMS,Goldstein JL2008 Selective versus total insulinresistance: a pathogenic paradox. Cell Metab 7:9596

    44. Savage DB, Semple RK 2010 Recent insights into fattyliver, metabolic dyslipidaemia and their links to insulinresistance. Curr Opin Lipidol 21:329336

    45. Murphy R, Baptista J, Holly J, Umpleby AM, Ellard S,Harries LW,Crolla J,CundyT,HattersleyAT2008Severeintrauterine growth retardation and atypical diabetes as-sociated with a translocation breakpoint disrupting regu-lation of the insulin-like growth factor 2 gene. J Clin En-docrinol Metab 93:43734380

    46. Srinivasan S, Waters MJ, Rowland JE, Baxter RC, VergeCF 2003 Hyperinsulinism and overgrowth without obe-sity. Arch Dis Child 88:332334

    47. Flier JS, Moller DE, Moses AC, ORahilly S, Chaiken RL,Grigorescu F, Elahi D, Kahn BB, Weinreb JE, Eastman R1993 Insulin-mediated pseudoacromegaly: clinical andbiochemical characterization of a syndrome of selectiveinsulin resistance. J Clin EndocrinolMetab 76:15331541

    48. Kausch C, Bergemann C, Hamann A, Matthaei S 1999Insulin-mediated pseudoacromegaly in a patient with se-vere insulin resistance: association of defective insulin-stimulated glucose transport with impaired phosphatidyl-inositol 3-kinase activity in fibroblasts. Exp ClinEndocrinol Diabetes 107:148154

    49. Dib K, Whitehead JP, Humphreys PJ, Soos MA, BaynesKC, Kumar S, Harvey T, ORahilly S 1998 Impaired acti-vation of phosphoinositide 3-kinase by insulin in fibro-

    Endocrine Reviews, August 2011, 32(4):498514 edrv.endojournals.org 509

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • blasts frompatientswith severe insulin resistance andpseu-doacromegaly. A disorder characterized by selectivepostreceptor insulin resistance. J Clin Invest 101:11111120

    50. KumarS,DurringtonPN,ORahilly S,Laing I,HumphreysPJ, Olukoga AO, Bhatnagar D, Mackness MI, Davis JR,Boulton AJ 1996 Severe insulin resistance, diabetes melli-tus, hypertriglyceridemia, and pseudoacromegaly. J ClinEndocrinol Metab 81:34653468

    51. Frasca F, Pandini G, Scalia P, Sciacca L,Mineo R, Costan-tino A, Goldfine ID, Belfiore A, Vigneri R 1999 Insulinreceptor isoform A, a newly recognized, high-affinity in-sulin-like growth factor II receptor in fetal and cancer cells.Mol Cell Biol 19:32783288

    52. Pollak M 2008 Insulin and insulin-like growth factor sig-nalling in neoplasia. Nat Rev Cancer 8:915928

    53. Rajaram S, BaylinkDJ,Mohan S 1997 Insulin-like growthfactor-binding proteins in serum and other biological flu-ids: regulation and functions. Endocr Rev 18:801831

    54. Tsugane S, Inoue M 2010 Insulin resistance and cancer:epidemiological evidence. Cancer Sci 101:10731079

    55. Kang HW, Kim D, Kim HJ, Kim CH, Kim YS, Park MJ,KimJS,ChoSH,SungMW,JungHC,LeeHS,Song IS2010Visceral obesity and insulin resistance as risk factors forcolorectal adenoma: a cross-sectional, case-control study.Am J Gastroenterol 105:178187

    56. Garg A 2000 Gender differences in the prevalence of met-abolic complications in familial partial lipodystrophy(Dunnigan variety). J Clin Endocrinol Metab 85:17761782

    57. VantyghemMC,Vincent-DesplanquesD,Defrance-FaivreF, Capeau J, Fermon C, Valat AS, Lascols O, Hecart AC,Pigny P, Delemer B, Vigouroux C, Wemeau JL 2008 Fer-tility andobstetrical complications inwomenwithLMNA-related familial partial lipodystrophy. J Clin EndocrinolMetab 93:22232229

    58. Li S, Brown MS, Goldstein JL 2010 Bifurcation of insulinsignaling pathway in rat liver: mTORC1 required for stim-ulation of lipogenesis, but not inhibition of gluconeogen-esis. Proc Natl Acad Sci USA 107:34413446

    59. Laplante M, Sabatini DM 2010 mTORC1 activatesSREBP-1c and uncouples lipogenesis from gluconeogene-sis. Proc Natl Acad Sci USA 107:32813282

    60. DuvelK,Yecies JL,MenonS,RamanP,LipovskyAI, SouzaAL, Triantafellow E, Ma Q, Gorski R, Cleaver S, VanderHeidenMG,MacKeigan JP, Finan PM, Clish CB,MurphyLO, Manning BD 2010 Activation of a metabolic generegulatory network downstream of mTOR complex 1.Mol Cell 39:171183

    61. Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J,Boldt MD, Parks EJ 2005 Sources of fatty acids stored inliver and secreted via lipoproteins in patients with nonal-coholic fatty liver disease. J Clin Invest 115:13431351

    62. Haas JT, Biddinger SB 2009 Dissecting the role of insulinresistance in the metabolic syndrome. Curr Opin Lipidol20:206210

    63. Dann SG, Selvaraj A, Thomas G 2007 mTOR Complex1-S6K1 signaling: at the crossroads of obesity, diabetes andcancer. Trends Mol Med 13:252259

    64. Newgard CB, An J, Bain JR,MuehlbauerMJ, Stevens RD,Lien LF, Haqq AM, Shah SH, Arlotto M, Slentz CA,Rochon J,GallupD, IlkayevaO,WennerBR,Yancy JrWS,

    Eisenson H,Musante G, Surwit RS, Millington DS, ButlerMD, Svetkey LP 2009 A branched-chain amino acid-re-latedmetabolic signature that differentiates obese and leanhumans and contributes to insulin resistance. Cell Metab9:311326

    65. HuffmanKM, Shah SH, StevensRD,Bain JR,MuehlbauerM, Slentz CA, Tanner CJ, Kuchibhatla M, Houmard JA,Newgard CB, Kraus WE 2009 Relationships between cir-culating metabolic intermediates and insulin action inoverweight to obese, inactive men and women. DiabetesCare 32:16781683

    66. Semple RK, Halberg NH, Burling K, Soos MA, Schraw T,Luan J, Cochran EK, Dunger DB, Wareham NJ, SchererPE, Gorden P, ORahilly S 2007 Paradoxical elevation ofhigh-molecularweight adiponectin in acquired extreme in-sulin resistancedue to insulin receptor antibodies.Diabetes56:17121717

    67. Antuna-Puente B, Boutet E, Vigouroux C, Lascols O,Slama L, Caron-Debarle M, Khallouf E, Levy-Marchal C,Capeau J, Bastard JP, Magre J 2010 Higher adiponectinlevels in patients with Berardinelli-Seip congenital lipodys-trophy due to seipin as compared with 1-acylglycerol-3-phosphate-o-acyltransferase-2 deficiency. J Clin Endocri-nol Metab 95:14631468

    68. Hattori Y, Hirama N, Suzuki K, Hattori S, Kasai K 2007Elevated plasma adiponectin and leptin levels in sisterswith genetically defective insulin receptors. Diabetes Care30:e109

    69. Semple R, Savage DB, ORahilly S 2009 Sex hormone-binding globulin and risk of type 2 diabetes. N Engl J Med361:2677; author reply 26772678

    70. Semple RK, Cochran EK, Soos MA, Burling KA, SavageDB, Gorden P, ORahilly S 2008 Plasma adiponectin as amarker of insulin receptor dysfunction: clinical utility insevere insulin resistance. Diabetes Care 31:977979

    71. Maddux BA, Chan A, De Filippis EA, Mandarino LJ,Goldfine ID 2006 IGF-binding protein-1 levels are relatedto insulin-mediated glucose disposal and are a potentialserum marker of insulin resistance. Diabetes Care 29:15351537

    72. Tsai EC, Matsumoto AM, Fujimoto WY, Boyko EJ 2004Association of bioavailable, free, and total testosteronewith insulin resistance: influence of sex hormone-bindingglobulin and body fat. Diabetes Care 27:861868

    73. Weyer C, Funahashi T, Tanaka S, Hotta K,MatsuzawaY,Pratley RE, Tataranni PA 2001 Hypoadiponectinemia inobesity and type 2 diabetes: close association with insulinresistance and hyperinsulinemia. J Clin Endocrinol Metab86:19301935

    74. Semple RK, Soos MA, Luan J, Mitchell CS, Wilson JC,Gurnell M, Cochran EK, Gorden P, Chatterjee VK, Ware-hamNJ, ORahilly S 2006 Elevated plasma adiponectin inhumans with genetically defective insulin receptors. J ClinEndocrinol Metab 91:32193223

    75. Cook JR, Semple RK 2010 Hypoadiponectinemiacauseor consequence of human insulin resistance? J Clin En-docrinol Metab 95:15441554

    76. Barbieri RL, Ryan KJ 1983 Hyperandrogenism, insulinresistance, and acanthosis nigricans syndrome: a commonendocrinopathy with distinct pathophysiologic features.Am J Obstet Gynecol 147:90101

    77. Kadowaki T, Bevins CL, Cama A, Ojamaa K, Marcus-

    510 Semple et al. Severe Insulin Resistance Syndromes Endocrine Reviews, August 2011, 32(4):498514

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • Samuels B, Kadowaki H, Beitz L, McKeon C, Taylor SI1988 Two mutant alleles of the insulin receptor gene in apatient with extreme insulin resistance. Science 240:787790

    78. Yoshimasa Y, Seino S, Whittaker J, Kakehi T, Kosaki A,Kuzuya H, Imura H, Bell GI, Steiner DF 1988 Insulin-resistant diabetes due to a point mutation that preventsinsulin proreceptor processing. Science 240:784787

    79. Ullrich A, Bell JR, Chen EY, Herrera R, Petruzzelli LM,Dull TJ, Gray A, Coussens L, Liao YC, Tsubokawa M,Mason A, Seeburg PH, Grunfeld C, Rosen OM, Ram-achandran J 1985Human insulin receptor and its relation-ship to the tyrosine kinase family of oncogenes. Nature313:756761

    80. RabsonSM,Mendenhall EN1956Familial hypertrophyofpineal body, hyperplasia of adrenal cortex and diabetesmellitus; report of 3 cases. Am J Clin Pathol 26:283290

    81. Donohue WL, Uchida I 1954 Leprechaunism: a euphe-mism for a rare familial disorder. J Pediatr 45:505519

    82. Semple RK, Savage DB, Halsall DJ, ORahilly S 2010 Syn-dromes of severe insulin resistance and/or lipodystrophy.In: Weiss RE, Refetoff S, eds. Genetic diagnosis of endo-crine disorders. Philadelphia: Elsevier

    83. Ogilvy-Stuart AL, Soos MA, Hands SJ, Anthony MY,Dunger DB, ORahilly S 2001 Hypoglycemia and resis-tance toketoacidosis in a subjectwithout functional insulinreceptors. J Clin Endocrinol Metab 86:33193326

    84. Longo N, Langley SD, Griffin LD, Elsas 2nd LJ 1992 Re-duced mRNA and a nonsense mutation in the insulin-re-ceptor gene produce heritable severe insulin resistance.Am J Hum Genet 50:9981007

    85. Foti D, Chiefari E, Fedele M, Iuliano R, Brunetti L, Paon-essa F, Manfioletti G, Barbetti F, Brunetti A, Croce CM,Fusco A, Brunetti A 2005 Lack of the architectural factorHMGA1 causes insulin resistance and diabetes in humansand mice. Nat Med 11:765773

    86. Berger D, Barroso I, SoosM,YeoG, Schafer AJ, ORahillyS, Whitehead JP 2002 Genetic variants of insulin receptorsubstrate-1 (IRS-1) in syndromes of severe insulin resis-tance. Functional analysis of Ala513Pro and Gly1158GluIRS-1. Diabet Med 19:804809

    87. Whitehead JP, Humphreys P, Krook A, Jackson R, Hay-ward A, Lewis H, Siddle K, ORahilly S 1998 Molecularscanning of the insulin receptor substrate 1 gene in subjectswith severe insulin resistance: detection and functionalanalysis of a naturally occurring mutation in a YMXMmotif. Diabetes 47:837839

    88. Bottomley WE, Soos MA, Adams C, Guran T, HowlettTA, Mackie A, Miell J, Monson JP, Temple R, Tenen-baum-Rakover Y, Tymms J, Savage DB, Semple RK,ORahilly S, Barroso I 2009 IRS2 variants and syndromesof severe insulin resistance. Diabetologia 52:12081211

    89. Tan K, Kimber WA, Luan J, Soos MA, Semple RK, Ware-ham NJ, ORahilly S, Barroso I 2007 Analysis of geneticvariation in Akt2/PKB- in severe insulin resistance, lipo-dystrophy, type 2 diabetes, and related metabolic pheno-types. Diabetes 56:714719

    90. Baynes KC, Beeton CA, Panayotou G, Stein R, Soos M,Hansen T, Simpson H, ORahilly S, Shepherd PR, White-head JP 2000 Natural variants of human p85 phospho-inositide 3-kinase in severe insulin resistance: a novel vari-

    ant with impaired insulin-stimulated lipid kinase activity.Diabetologia 43:321331

    91. Dash S, Sano H, Rochford JJ, Semple RK, Yeo G, HydenCS, Soos MA, Clark J, Rodin A, Langenberg C, Druet C,Fawcett KA, Tung YC, WarehamNJ, Barroso I, LienhardGE, ORahilly S, Savage DB 2009 A truncation mutationin TBC1D4 in a family with acanthosis nigricans and post-prandial hyperinsulinemia. Proc Natl Acad Sci USA 106:93509355

    92. Peters JM, Barnes R, Bennett L, Gitomer WM, BowcockAM, Garg A 1998 Localization of the gene for familialpartial lipodystrophy (Dunnigan variety) to chromosome1q2122. Nat Genet 18:292295

    93. Cao H, Hegele RA 2000 Nuclear lamin A/C R482Q mu-tation in Canadian kindreds with Dunnigan-type familialpartial lipodystrophy. Hum Mol Genet 9:109112

    94. Shackleton S, Lloyd DJ, Jackson SN, Evans R, NiermeijerMF, Singh BM, Schmidt H, Brabant G, Kumar S, Dur-rington PN, Gregory S, ORahilly S, Trembath RC 2000LMNA, encoding lamin A/C, is mutated in partial lipodys-trophy. Nat Genet 24:153156

    95. Magre J, Delepine M, Khallouf E, Gedde-Dahl Jr T, VanMaldergem L, Sobel E, Papp J, Meier M, Megarbane A,Bachy A, Verloes A, dAbronzo FH, Seemanova E, AssanR, Baudic N, Bourut C, Czernichow P, Huet F, GrigorescuF, de KerdanetM, LacombeD, Labrune P, LanzaM, LoretH, Matsuda F, Navarro J, Nivelon-Chevalier A, Polak M,Robert JJ, Tric P, Tubiana-Rufi N, Vigouroux C, Weis-senbach J, Savasta S, Maassen JA, Trygstad O, Bogalho P,Freitas P,Medina JL, Bonnicci F, Joffe BI, Loyson G, PanzVR, Raal FJ, ORahilly S, Stephenson T, Kahn CR, Lath-ropM, Capeau J 2001 Identification of the gene altered inBerardinelli-Seip congenital lipodystrophy on chromo-some 11q13. Nat Genet 28:365370

    96. AgarwalAK,AriogluE,DeAlmeidaS,AkkocN,Taylor SI,Bowcock AM, Barnes RI, Garg A 2002 AGPAT2 is mu-tated in congenital generalized lipodystrophy linked tochromosome 9q34. Nat Genet 31:2123

    97. Hegele RA, Joy TR, Al-Attar SA, Rutt BK 2007 Thematicreview series: adipocyte biology. Lipodystrophies: win-dows on adipose biology and metabolism. J Lipid Res 48:14331444

    98. Monajemi H, Stroes E, Hegele RA, Fliers E 2007 Inheritedlipodystrophies and the metabolic syndrome. Clin Endo-crinol (Oxf) 67:479484

    99. Garg A, Agarwal AK 2009 Lipodystrophies: disorders ofadipose tissue biology. Biochim Biophys Acta 1791:507513

    100. JeningaEH,KalkhovenE2010Central players in inheritedlipodystrophies. Trends Endocrinol Metab 21:581588

    101. Capeau J,Magre J, Caron-DebarleM, LagathuC,AntoineB, Bereziat V, Lascols O, Bastard JP, Vigouroux C 2010Human lipodystrophies: genetic and acquired diseases ofadipose tissue. Endocr Dev 19:120

    102. Kim CA, Delepine M, Boutet E, El Mourabit H, Le Lay S,Meier M, Nemani M, Bridel E, Leite CC, Bertola DR,Semple RK, ORahilly S, Dugail I, Capeau J, Lathrop M,Magre J 2008 Association of a homozygous nonsensecaveolin-1 mutation with Berardinelli-Seip congenital lip-odystrophy. J Clin Endocrinol Metab 93:11291134

    103. HayashiYK,MatsudaC,OgawaM,GotoK,TominagaK,Mitsuhashi S, Park YE, Nonaka I, Hino-Fukuyo N, Hagi-

    Endocrine Reviews, August 2011, 32(4):498514 edrv.endojournals.org 511

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • noya K, Sugano H, Nishino I 2009 Human PTRF muta-tions cause secondary deficiency of caveolins resulting inmuscular dystrophywith generalized lipodystrophy. JClinInvest 119:26232633

    104. Rajab A, Straub V, McCann LJ, Seelow D, Varon R, Bar-resi R, Schulze A, Lucke B, Lutzkendorf S, KarbasiyanM,Bachmann S, Spuler S, Schuelke M 2010 Fatal cardiac ar-rhythmia and long-QT syndrome in a new form of con-genital generalized lipodystrophy with muscle rippling(CGL4) due to PTRF-CAVIN mutations. PLoS Genet6:e1000874

    105. Shastry S, Delgado MR, Dirik E, Turkmen M, AgarwalAK, Garg A 2010 Congenital generalized lipodystrophy,type 4 (CGL4) associated with myopathy due to novelPTRF mutations. Am J Med Genet A 152A:22452253

    106. Dwianingsih EK, Takeshima Y, Itoh K, Yamauchi Y,AwanoH,Malueka RG,Nishida A, OtaM, YagiM,Mat-suoM 2010A Japanese childwith asymptomatic elevationof serum creatine kinase shows PTRF-CAVIN mutationmatching with congenital generalized lipodystrophy type4. Mol Genet Metab 101:233237

    107. Rubio-Cabezas O, Puri V, Murano I, Saudek V, SempleRK, Dash S, Hyden CS, Bottomley W, Vigouroux C,Magre J, Raymond-Barker P, Murgatroyd PR, Chawla A,Skepper JN,ChatterjeeVK,SulimanS,PatchAM,AgarwalAK, Garg A, Barroso I, Cinti S, Czech MP, Argente J,ORahilly S, Savage DB 2009 Partial lipodystrophy andinsulin resistant diabetes in a patient with a homozygousnonsense mutation in CIDEC. EMBO Mol Med 1:280287

    108. CohenAW,HnaskoR, SchubertW, LisantiMP 2004Roleof caveolae andcaveolins inhealthanddisease. PhysiolRev84:13411379

    109. Blouin CM, Le Lay S, Eberl A, Kofeler HC, Guerrera IC,Klein C, Le Liepvre X, Lasnier F, Bourron O, Gautier JF,Ferre P, Hajduch E, Dugail I 2010 Lipid droplet analysis incaveolin-deficient adipocytes: Alterations in surface phos-pholipid composition and maturation defects. J Lipid Res51:945956

    110. Liu L, Brown D, McKee M, Lebrasseur NK, Yang D, Al-brecht KH, Ravid K, Pilch PF 2008 Deletion of Cavin/PTRF causes global loss of caveolae, dyslipidemia, andglucose intolerance. Cell Metab 8:310317

    111. de Haan W 2010 Lipodystrophy and muscular dystrophycaused by PTRF mutations. Clin Genet 77:436437

    112. Minetti C, Sotgia F, BrunoC, Scartezzini P, Broda P, BadoM,Masetti E, MazzoccoM, Egeo A, Donati MA, VolonteD, Galbiati F, Cordone G, Bricarelli FD, Lisanti MP, ZaraF 1998 Mutations in the caveolin-3 gene cause autosomaldominant limb-girdle muscular dystrophy. Nat Genet 18:365368

    113. Keller P, Petrie JT, De Rose P, Gerin I,WrightWS, ChiangSH, Nielsen AR, Fischer CP, Pedersen BK, MacDougaldOA 2008 Fat-specific protein 27 regulates storage of tri-acylglycerol. J Biol Chem 283:1435514365

    114. Toh SY, Gong J, Du G, Li JZ, Yang S, Ye J, Yao H, ZhangY, Xue B, Li Q, Yang H, Wen Z, Li P 2008 Up-regulationof mitochondrial activity and acquirement of brown adi-pose tissue-like property in the white adipose tissue offsp27 deficient mice. PLoS One 3:e2890

    115. Nishino N, Tamori Y, Tateya S, Kawaguchi T, ShibakusaT, Mizunoya W, Inoue K, Kitazawa R, Kitazawa S, Mat-

    suki Y, Hiramatsu R,Masubuchi S, Omachi A, Kimura K,Saito M, Amo T, Ohta S, Yamaguchi T, Osumi T, ChengJ, Fujimoto T, Nakao H, Nakao K, Aiba A, Okamura H,Fushiki T, Kasuga M 2008 FSP27 contributes to efficientenergy storage in murine white adipocytes by promotingthe formation of unilocular lipid droplets. J Clin Invest118:28082821

    116. Puri V, Czech MP 2008 Lipid droplets: FSP27 knockoutenhances their sizzle. J Clin Invest 118:26932696

    117. Szymanski KM, Binns D, Bartz R, Grishin NV, Li WP,Agarwal AK, Garg A, Anderson RG, Goodman JM 2007The lipodystrophy protein seipin is found at endoplasmicreticulum lipiddroplet junctions and is important fordrop-let morphology. Proc Natl Acad Sci USA 104:2089020895

    118. Fei W, Shui G, Gaeta B, Du X, Kuerschner L, Li P, BrownAJ, Wenk MR, Parton RG, Yang H 2008 Fld1p, a func-tional homologue of human seipin, regulates the size oflipid droplets in yeast. J Cell Biol 180:473482

    119. Boutet E, El Mourabit H, ProtM, NemaniM, Khallouf E,ColardO,MauriceM,Durand-SchneiderAM,ChretienY,Gre`s S, Wolf C, Saulnier-Blache JS, Capeau J, Magre J2009Seipindeficiency alters fatty acidDelta9desaturationand lipid droplet formation in Berardinelli-Seip congenitallipodystrophy. Biochimie 91:796803

    120. Payne VA, Grimsey N, Tuthill A, Virtue S, Gray SL, DallaNora E, Semple RK, ORahilly S, Rochford JJ 2008 Thehuman lipodystrophy gene BSCL2/seipin may be essentialfor normal adipocyte differentiation. Diabetes 57:20552060

    121. Savage DB, Agostini M, Barroso I, Gurnell M, Luan J,Meirhaeghe A, Harding AH, Ihrke G, Rajanayagam O,Soos MA, George S, Berger D, Thomas EL, Bell JD,MeeranK,RossRJ,Vidal-PuigA,WarehamNJ,ORahillyS, Chatterjee VK, Schafer AJ 2002 Digenic inheritance ofsevere insulin resistance in a human pedigree. Nat Genet31:379384

    122. Newgard CB, BradyMJ, ODoherty RM, Saltiel AR 2000Organizing glucose disposal: emerging roles of the glyco-gen targeting subunits of protein phosphatase-1. Diabetes49:19671977

    123. Lanner C, Suzuki Y, Bi C, Zhang H, Cooper LD, Bowker-Kinley MM, DePaoli-Roach AA 2001 Gene structure andexpression of the targeting subunit, RGL, of the muscle-specific glycogen-associated type 1 protein phosphatase,PP1G. Arch Biochem Biophys 388:135145

    124. Suzuki Y, Lanner C, Kim JH, Vilardo PG, Zhang H, YangJ, Cooper LD, SteeleM, Kennedy A, Bock CB, ScrimgeourA, Lawrence Jr JC, DePaoli-Roach AA 2001 Insulin con-trol of glycogen metabolism in knockout mice lacking themuscle-specific protein phosphatase PP1G/RGL.Mol CellBiol 21:26832694

    125. Savage DB, Zhai L, Ravikumar B, Choi CS, Snaar JE,McGuire AC, Wou SE, Medina-Gomez G, Kim S, BockCB, Segvich DM, Solanky B, Deelchand D, Vidal-Puig A,Wareham NJ, Shulman GI, Karpe F, Taylor R, PedersonBA, Roach PJ, ORahilly S, DePaoli-Roach AA 2008 Aprevalent variant in PPP1R3A impairs glycogen synthesisand reduces muscle glycogen content in humans and mice.PLoS Med 5:e27

    126. Montague CT, Farooqi IS, Whitehead JP, Soos MA, RauH, Wareham NJ, Sewter CP, Digby JE, Mohammed SN,

    512 Semple et al. Severe Insulin Resistance Syndromes Endocrine Reviews, August 2011, 32(4):498514

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • Hurst JA,CheethamCH,EarleyAR,BarnettAH, Prins JB,ORahilly S1997Congenital leptin deficiency is associatedwith severe early-onset obesity in humans. Nature 387:903908

    127. Minton JA, Owen KR, Ricketts CJ, Crabtree N, Shaikh G,Ehtisham S, Porter JR, Carey C, Hodge D, Paisey R,Walker M, Barrett TG 2006 Syndromic obesity and dia-betes: changes in body compositionwith age andmutationanalysis of ALMS1 in 12 United Kingdom kindreds withAlstrom syndrome. J Clin Endocrinol Metab 91:31103116

    128. Huang-Doran I, Bicknell LS, Finucane FM, RochaN, Por-ter KM, Tung YC, Szekeres F, Krook A, Nolan JJ,ODriscoll M, Bober M, ORahilly S, Jackson AP, SempleRK 2011 Genetic defects in human pericentrin are associ-ated with severe insulin resistance and diabetes mellitus.Diabetes 60:925935

    129. Green JS, Parfrey PS, Harnett JD, Farid NR, Cramer BC,Johnson G, Heath O, McManamon PJ, OLeary E, Pryse-Phillips W 1989 The cardinal manifestations of Bardet-Biedl syndrome, a form of Laurence-Moon-Biedl syn-drome. N Engl J Med 321:10021009

    130. Zaghloul NA, Katsanis N 2009 Mechanistic insights intoBardet-Biedl syndrome, a model ciliopathy. J Clin Invest119:428437

    131. Yamada K, Ikegami H, Yoneda H, Miki T, Ogihara T1999 All patients with Werners syndrome are insulin re-sistant, but only those who also have impaired insulin se-cretion develop overt diabetes. Diabetes Care 22:20942095

    132. ImuraH,NakaoY, KuzuyaH,OkamotoM,OkamotoM,YamadaK 1985Clinical, endocrine andmetabolic aspectsof the Werner syndrome compared with those of normalaging. Adv Exp Med Biol 190:171185

    133. Diaz A, Vogiatzi MG, Sanz MM, German J 2006 Evalu-ation of short stature, carbohydrate metabolism and otherendocrinopathies in Blooms syndrome. Horm Res 66:111117

    134. Jacob KN, Garg A 2006 Laminopathies: multisystem dys-trophy syndromes. Mol Genet Metab 87:289302

    135. Schalch DS, McFarlin DE, Barlow MH 1970 An unusualform of diabetes mellitus in ataxia telangiectasia. N EnglJ Med 282:13961402

    136. Blevins Jr LS, Gebhart SS 1996 Insulin-resistant diabetesmellitus in a black woman with ataxia-telangiectasia.South Med J 89:619621

    137. Koulouri O, Conway GS 2009Management of hirsutism.BMJ 338:b847

    138. Higgins SP, Freemark M, Prose NS 2008 Acanthosis nig-ricans: a practical approach to evaluation and manage-ment. Dermatol Online J 14:2

    139. Cochran E, Musso C, Gorden P 2005 The use of U-500 inpatients with extreme insulin resistance. Diabetes Care 28:12401244

    140. LaneWS 2006 Use of U-500 regular insulin by continuoussubcutaneous insulin infusion in patients with type 2 dia-betes and severe insulin resistance. Endocr Pract 12:251256

    141. Takahashi Y, Kadowaki H,Momomura K, Fukushima Y,Orban T, Okai T, Taketani Y, Akanuma Y, Yazaki Y,Kadowaki T 1997 A homozygous kinase-defective muta-

    tion in the insulin receptor gene in a patient with lepre-chaunism. Diabetologia 40:412420

    142. Nakae J, Kato M, Murashita M, Shinohara N, Tajima T,Fujieda K 1998 Long-term effect of recombinant humaninsulin-like growth factor I on metabolic and growth con-trol in a patient with leprechaunism. J Clin EndocrinolMetab 83:542549

    143. LongoN, Singh R, Griffin LD, Langley SD, Parks JS, ElsasLJ 1994 Impaired growth in Rabson-Mendenhall syn-drome: lack of effect of growth hormone and insulin-likegrowth factor-I. J Clin Endocrinol Metab 79:799805

    144. Vestergaard H, Rossen M, Urhammer SA, Muller J, Ped-ersen O 1997 Short- and long-term metabolic effects ofrecombinant human IGF-I treatment in patients with se-vere insulin resistance and diabetes mellitus. Eur J Endo-crinol 136:475482

    145. Kuzuya H, Matsuura N, Sakamoto M, Makino H, Saka-moto Y, Kadowaki T, Suzuki Y, Kobayashi M, AkazawaY, Nomura M 1993 Trial of insulin-like growth factor Itherapy for patients with extreme insulin resistance syn-dromes. Diabetes 42:696705

    146. Zenobi PD,Glatz Y, Keller A,Graf S, Jaeggi-Groisman SE,Riesen WF, Schoenle EJ, Froesch ER 1994 Beneficial met-abolic effects of insulin-like growth factor I in patientswithsevere insulin-resistant diabetes type A. Eur J Endocrinol131:251257

    147. Dozio N, Scavini M, Beretta A, Sartori S, Meschi F, Saru-geri E, Pozza G 1995 In vivo metabolic effects of insulin-like growth factor-I not mediated through the insulin re-ceptor. J Clin Endocrinol Metab 80:13251328

    148. Quin JD,CheckleyA,GallagherA, Jones J,MacCuishAC,Miell JP 1994 Response of insulin-like growth factor(IGF)-binding protein-1 (IGFBP-1) and IGFBP-3 to IGF-Itreatment in severe insulin resistance. J Endocrinol 141:177182

    149. George S, Johansen A, SoosMA,Mortensen H, Gammelt-oft S, Saudek V, Siddle K, Hansen L, ORahilly S 2003Deletion of V335 from the L2 domain of the insulin re-ceptor results in a conformationally abnormal receptorthat is unable to bind insulin and causes Donohues syn-drome in a human subject. Endocrinology 144:631637

    150. McDonald A, Williams RM, Regan FM, Semple RK,DungerDB 2007 IGF-I treatment of insulin resistance. EurJ Endocrinol 157(Suppl 1):S51S56

    151. Regan FM, Williams RM, McDonald A, Umpleby AM,Acerini CL, ORahilly S, Hovorka R, Semple RK, DungerDB 2010 Treatment with recombinant human insulin-likegrowth factor (rhIGF)-I/rhIGF binding protein-3 compleximproves metabolic control in subjects with severe insulinresistance. J Clin Endocrinol Metab 95:21132122

    152. Haque WA, Shimomura I, Matsuzawa Y, Garg A 2002Serum adiponectin and leptin levels in patients with lipo-dystrophies. J Clin Endocrinol Metab 87:2395

    153. Oral EA, Simha V, Ruiz E, Andewelt A, Premkumar A,Snell P, Wagner AJ, DePaoli AM, ReitmanML, Taylor SI,Gorden P, Garg A 2002 Leptin-replacement therapy forlipodystrophy. N Engl J Med 346:570578

    154. Petersen KF, Oral EA, Dufour S, Befroy D, Ariyan C, YuC, Cline GW, DePaoli AM, Taylor SI, Gorden P, ShulmanGI 2002 Leptin reverses insulin resistance and hepatic ste-atosis in patients with severe lipodystrophy. J Clin Invest109:13451350

    Endocrine Reviews, August 2011, 32(4):498514 edrv.endojournals.org 513

    The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 25 September 2014. at 12:56 For personal use only. No other uses without permission. . All rights reserved.

  • 155. JavorED,GhanyMG,CochranEK,OralEA,DePaoliAM,Premkumar A, Kleiner DE, Gorden P 2005 Leptin reversesnonalcoholic steatohepatitis in patients with severe lipo-dystrophy. Hepatology 41:753760

    156. Beltrand J, Beregszaszi M, Chevenne D, Sebag G, De Ker-danet M, Huet F, Polak M, Tubiana-Rufi N, Lacombe D,De Paoli AM, Levy-Marchal C 2007Metabolic correctioninduced by leptin replacement treatment in young childrenwith Berardinelli-Seip congenital lipoatrophy. Pediatrics120:e291e296

    157. Chong AY, Lupsa BC, Cochran EK, Gorden P 2010 Effi-cacy of leptin therapy in the different forms of human li-podystrophy. Diabetologia 53:2735

    158. Beltrand J, Lahlou N, Le Charpentier T, Sebag G, Leka S,Polak M, Tubiana-Rufi N, Lacombe D, de Kerdanet M,Huet F, Robert JJ, Chevenne D, Gressens P, Levy-MarchalC 2010 Resistance to leptin-replacement therapy in Berar-dinelli-Seip congenital lipodystrophy: an immunologicalorigin. Eur J Endocrinol 162:10831091

    159. Savage DB, ORahilly S 2010 Leptin therapy in lipodys-trophy. Diabetologia 53:79

    160. Mantzoros CS 2009 Whither recombinant human leptintreatment for HIV-associated lipoatrophy and the meta-bolic syndrome? J Clin Endocrinol Metab 94:10891091

    161. Cochran E, Young JR, Sebring N, DePaoli A, Oral EA,Gorden P 2004 Efficacy of recombinant methionyl humanleptin therapy for the extreme insulin resistance of theRab-son-Mendenhall syndrome. J Clin Endocrinol Metab 89:15481554

    162. Skarulis MC, Celi FS, Mueller E, Zemskova M, Malek R,Hugendubler L, Cochran C, Solomon J, Chen C, GordenP 2010 Thyroid hormone induced brown adipose tissueand amelioration of diabetes in a patient with