throbber
Disease Markers 20 (2004) 161–165
`IOS Press
`
`161
`
`Update on diabetes mellitus
`
`Murray Korc
`Division of Endocrinology, Departments of Medicine, and Pharmacology and Toxicology, Dartmouth-Hitchcock
`Medical Center and Dartmouth Medical School, Hanover, NH, USA
`
`Abstract. Diabetes mellitus is a complex multi-system disorder that may be classified as autoimmune mediated type 1 diabetes,
`or as insulin resistance associated type 2 diabetes. In type 1 diabetes, there is selective loss of the beta cells within the endocrine
`islets, as a consequence of T-cell and cytokine mediated destruction of these cells, perhaps in conjunction with destruction of
`the peri-islet Schwann cells. In type 2 diabetes, the etiology of the resistance ranges from post-receptor defects in the insulin
`signaling pathway to excessive production of adipocyte derived cytokines that antagonize insulin action to mitochondrial defects
`that interfere with glucose disposal. Proteome based technologies are providing new insights into these defects.
`
`Keywords: Beta cell, insulin resistance, mitochondrial defects, pancreatic cancer
`
`1. Introduction
`
`Diabetes mellitus afflicts 6 to 8% of the population in
`the US. Type 1 diabetes mellitus (T1DM) patients have
`lost their beta cells as a result of aberrant activation
`of cellular immune mechanisms, and they no longer
`produce insulin. They are dependent, therefore, on
`insulin injections for survival. Most individuals with
`diabetes have type 2 diabetes mellitus (T2DM) and are
`resistant to insulin action. T2DM is also associated
`with beta cell dysfunction, and with production of fat-
`derived cytokines that antagonize insulin actions. This
`review will focus on several recent developments in
`T1DM and T2DM with an emphasis on the application
`of proteomic technologies.
`
`2. The beta cell and insulin secretion
`
`The pancreas is a complex tissue consisting of acinar
`cells that produce digestive enzymes, duct cells that
`produce bicarbonate rich fluid, and approximately one
`million endocrine islets which are distributed through-
`
`∗
`Correspondence to: Murray Korc, M.D., Department of
`Medicine, One Medical Center Drive, Lebanon, NH 03756, USA.
`Tel.: +1 603 650 7936; Fax: +1 603 650 6122; E-mail: murray.korc@
`dartmouth.edu.
`
`out the exocrine pancreas, with the greatest density oc-
`curring in the tail of the pancreas. The islets are rich
`in beta cells which secrete insulin, a hormone that is
`packaged into heterogeneous granules with electron-
`dense cores [1]. In addition, there are adjoining cells
`in the islets which secrete glucagon (alpha cells) and
`somatostatin (delta cells), and these hormones coun-
`teract insulin’s hypoglycemic effects and insulin se-
`cretion, respectively. There is also a small population
`of endocrine cells that secrete pancreatic polypeptide,
`whose function is not yet fully elucidated.
`Small amounts of insulin are secreted from the beta
`cells in the fasting state, acting to inhibit hepatic glu-
`coneogenesis [2]. Following a meal, glucose enters the
`beta cell through a process of facilitated diffusion that
`is mediated by the GLUT-2 glucose transporter. The
`same glucose transporter is also found in the liver, renal
`tubules, and the small intestine, but not in other types of
`islet cells [3]. GLUT-2 exhibits a high Km for glucose
`uptake, allowing the beta cell to transport glucose in
`proportion to the extracellular glucose concentration,
`thereby leading to a large increase in insulin secretion.
`Glucose stimulated release of insulin is biphasic,
`with a rapid first phase and a more gradual sec-
`ond phase that lasts for two hours and that is partly
`dependent on the release of newly synthesized in-
`sulin [4]. Glucose acts by generating ATP which in-
`hibits a K+ channel, thereby depolarizing the beta cell
`
`ISSN 0278-0240/04/$17.00  2004 – IOS Press and the authors. All rights reserved
`
`MPI EXHIBIT 1074 PAGE 1
`
`MPI EXHIBIT 1074 PAGE 1
`
`

`

`162
`
`M. Korc / Update on diabetes mellitus
`
`plasma membrane, activating voltage-dependent cal-
`cium channels, and inducing a rise in intracellular free
`calcium [5–7]. Calcium-calmodulin protein kinases,
`cyclic AMP (cAMP)-dependent protein kinases, phos-
`phatidic acid, lysophospholipids, and arachidonic acid
`and its metabolites also have important roles in modu-
`lating insulin secretion [8–10]. Inter- and intracellular
`signaling by the cells of the pancreatic islet in response
`to glucose, as well as other metabolites, also contribute
`to the integrated response of the beta cell and to the
`controlled release of insulin in response to a meal.
`
`3. Type 1 diabetes mellitus
`
`T1DM is characterized by progressive destruction of
`the beta cells due to the aberrant activation of cellular
`immune mechanisms, as manifested by the presence of
`T-cell infiltrates around and within the islets. There are
`approximately one million T1DM patients in the US,
`all of whom are dependent on insulin therapy for their
`survival. The absence of insulin also makes them prone
`to develop ketoacidosis, a potentially deadly metabolic
`complication.
`The mechanisms that lead to beta cell destruction in
`T1DM are still not completely understood. It is widely
`accepted, however, that the selective assault on the beta
`cell is mediated by cytotoxic T-cells and by certain cy-
`tokines [11]. For example, interleukin-1β (IL-1β) can
`suppress beta cell function and survival [12]. Proteome
`based studies of islets and islet derived cell lines that
`were exposed to IL-1β has revealed a complex pat-
`tern of protein alterations, including increased and de-
`creased protein expression and de novo protein induc-
`tion, underscoring the wide range of responses that can
`be elicited in the these cells and that can modulate their
`responsiveness and susceptibility to cytokine mediated
`cell death [13–15].
`In addition to in vitro studies, investigators have ad-
`dressed the issue of beta cell destruction in numer-
`ous in vivo studies. For example, in a recent study
`with the non-obese diabetic (NOD) mouse, which is
`an excellent model of T1DM, the Schwann cells that
`surround the islets were noted to be targeted at an
`early, pre-diabetic stage by the aberrantly activated T
`cells [16]. These peri-islet Schwann cells (pSC) ex-
`press glial fibrillary acidic protein (GFAP), and the
`authors noted that the diabetes-prone NOD mice de-
`velop pSC-autoreactive T and B cell responses that are
`associated with progressive pSC death that precedes
`beta cell death. They used surface-enhanced laser des-
`
`orption/ionization (SELDI) with time-of-flight (TOF)
`mass spectrometry to detect auto-antibodies against
`GFAP in the sera of NOD mice and newly diagnosed
`diabetic children with T1DM [16]. Thus, an important
`component of the immune mediated destruction of the
`beta cell may be mediated by aberrant targeting of pSC.
`In another study, peptide epitopes from naturally pro-
`cessed proinsulin were delineated by TOF mass spec-
`trometry, and used to establish very sensitive enzyme-
`linked immunosorbent assays to assess the nature of
`auto-reactive T-cells in T1DM [17]. These T-cells ex-
`hibited a pro-inflammatory Th1 response in T1DM pa-
`tients, but a T-regulatory cell response leading to the
`preferential production of the anti-inflammatory cy-
`tokine interleukin-10 (IL-10) in non-diabetic individu-
`als [17]. These observations suggest that the immune
`system actively protects against potential beta cell de-
`struction, and that this protective mechanism is lost in
`T1DM.
`
`4. Type 2 diabetes mellitus
`
`T2DM is characterized by insulin resistance, a fail-
`ure of the beta cell to produce enough insulin to over-
`come the resistance, inappropriate hepatic glucose re-
`lease, and production of cytokines by adipose tissue
`that interfere with insulin action. There are approxi-
`mately 16 million T2DM patients in the US, some of
`whom may require insulin therapy to achieve adequate
`blood glucose control. The presence of normal, in-
`creased, or slightly decreased circulating insulin levels
`assures that they only rarely develop ketoacidosis.
`Insulin resistance in T2DM may be the consequence
`of abnormal production of anti-insulin receptor an-
`tibodies [18], but is generally due to post-receptor
`defects [19]. Some examples of post-receptor de-
`fects include silent polymorphisms and naturally oc-
`curring amino acid substitutions in the insulin recep-
`tor substrate-1 (IRS-1) signaling protein which may
`contribute to insulin resistance [20,21].
`In support
`of this hypothesis, IRS-1 knockout mice exhibit in-
`sulin resistance and glucose intolerance [22], and IRS-
`2 knockout mice exhibit decreased insulin-stimulated
`glucose transport in conjunction with decreased beta
`cell mass and overt diabetes [23]. Similarly, there
`is homology between the ataxia-telangiectasia (AT)
`gene product and phosphatidylinositol 3-kinase (PI 3-
`kinase). The relatively high frequency of T2DM in pa-
`tients with AT raises the possibility that perturbations
`in PI 3-kinase function may represent one of the post-
`
`MPI EXHIBIT 1074 PAGE 2
`
`MPI EXHIBIT 1074 PAGE 2
`
`

`

`M. Korc / Update on diabetes mellitus
`
`163
`
`receptor defects that contributes to insulin resistance
`in some T2DM [24]. Mutations in the peroxisome
`proliferator-activated receptor (PPARγ) may lead to al-
`tered adipocyte differentiation and energy storage and
`may also contribute to insulin resistance in T2DM [25].
`Furthermore, excessive secretion of certain hormones
`such as cortisol, as seen in Cushing’s disease, or growth
`hormone, as seen in acromegaly, may antagonize in-
`sulin action sufficiently to induce a T2DM-like state.
`T2DM is often associated with obesity, which is a
`major cause for insulin resistance. Increased visceral
`obesity is especially deleterious in this regard because
`visceral fat is prone to release free fatty acids (FFA)
`which directly interfere with efficient insulin signaling,
`partly as a result of enhanced protein kinase C isoforms
`activity and increased hepatic glucose release [26,27].
`Furthermore, visceral fat expresses the β3-adrenergic
`receptor, which participates in the regulation of ther-
`mogenesis and lipolysis [28], and mutations in this re-
`ceptor are associated with insulin resistance, an earlier
`onset of T2DM and attenuated insulin secretion [29,
`30]. The adipocyte is also the source of several cy-
`tokines such as tumor necrosis factor alpha (TNF-α),
`which induces insulin resistance by down-regulating
`GLUT4 and increasing free fatty acid release [31],
`and interleukin-6 (IL-6), which induces insulin resis-
`tance in fat cells and hepatocytes [32,33]. By con-
`trast, adiponectin and IL-10 exert anti-diabetogenic ef-
`fects [34,35], and reduced levels of IL-10 increase the
`risk for the development of the dysmetabolic syndrome
`in women [36]. In addition to obesity and diabetes, pa-
`tients with the dysmetabolic syndrome exhibit hyper-
`tension, hyperlipidemia, heart disease and peripheral
`vascular disease, and a tendency towards elevated uric
`acid levels.
`The role of adipose tissue in the pathogenesis of in-
`sulin resistance has been investigated in several model
`cell lines, including the mouse 3T3-L1 fibroblastic cell
`line which differentiates rapidly into an adipocyte phe-
`notype in response to insulin and is useful,therefore, for
`studying insulin action and adipogenic differentiation.
`Thus, two-dimensional gel electrophoresis of 3T3-L1
`cell lysates revealed that insulin does not stimulate
`calmodulin phosphorylation under conditions in which
`it stimulates the phosphorylation of other proteins [37],
`and indicated that altered expression of several cellu-
`lar proteins contributes to the differentiation process in
`these cells [38]. More recently, a proteome based ap-
`proach using velocity gradient centrifugation to achieve
`the initial separation of proteins revealed that there was
`a marked increase in mitochondrial proteins in these
`
`cells during the differentiation process [39]. The au-
`thors also examined the effects of rosiglitazone, an ag-
`onist that activates the gamma isoform of the peroxi-
`some proliferator-activated receptor (PPARγ), which is
`a nuclear receptor that modulates adipocyte differentia-
`tion. In addition to altering mitochondrial morphology,
`rosiglitazone increased the levels of proteins involved
`in fatty acid oxidation, such as acyl-CoA synthetase and
`dehydrogenase [40]. Rosiglitazone also increases car-
`boxypeptidase B expression in mouse islets [41], and
`modulates components of the peroxisomal fatty acid
`metabolism pathway in adipose tissue [42], raising the
`possibility that it may also ameliorate glucose home-
`ostasis by improving insulin processing and fatty acid
`metabolism.
`Another site of insulin resistance is the skeletal mus-
`cle. A proteome based analysis of human skeletal
`muscle identified 8 potential markers of T2DM [43].
`The levels of two proteins that have a crucial role in
`ATP synthesis, creatine kinase B and ATP synthase β-
`subunit, were decreased in the muscle tissue of T2DM
`patients. Genetic muscular disorders may also be as-
`sociated with a high incidence of T2DM. For example,
`patients with mitochondrial myopathies may present
`with muscle weakness, symmetric paralysis of the ex-
`traocular muscles, drooping eyelids (ptosis), T2DM
`and cardiomyopathy [44]. Other patients with T2DM
`exhibit a maternal pattern of inheritance in conjunc-
`tion with inherited deafness and mitochondrial gene de-
`fects [45]. The importance of mitochondrial defects in
`T2DM was highlighted in a recent study which revealed
`that insulin-resistant children of T2DM parents exhib-
`ited increased muscle cell lipid content in conjunction
`with decreased mitochondrial phosphorylation, as de-
`termined by magnetic resonance spectroscopy [46]. In-
`terestingly, rates of lipolysis and plasma levels of IL-6,
`TNF-a, and adiponectin were not different in the two
`groups, further underscoring the potential importance
`of the mitochondrial defect in the pathogenesis of in-
`sulin resistance.
`Disorders of the pancreas that are neither T1DM nor
`T2DM may also be associated with glucose intolerance
`and diabetes. These include chronic pancreatitis, cystic
`fibrosis, pancreatic cancer, sequelae of partial pancrea-
`tectomy, hemochromatosis, and transfusion associated
`iron overload. It has been suggested that increased iron
`stores in general may lead to an increased propensity
`to T2DM [47], and that both chronic pancreatitis and
`pancreatic cancer may be associated with a significant
`component of insulin resistance that contributes to the
`hyperglycemia that may occur in these conditions [48,
`
`MPI EXHIBIT 1074 PAGE 3
`
`MPI EXHIBIT 1074 PAGE 3
`
`

`

`164
`
`M. Korc / Update on diabetes mellitus
`
`49]. Future proteome based studies in these conditions
`may therefore shed new light on novel mechanisms for
`insulin resistance.
`
`5. Conclusion
`
`T1DM is caused by immune mediated beta cell de-
`struction. T2DM is caused by insulin resistance in con-
`junction with variable degrees of a defective beta cell
`response to hyperglycemia [50]. Proteome based stud-
`ies are likely to yield additional insight into the mech-
`anisms involved in pathophysiology of both disorders.
`Already, proteome based studies are contributing infor-
`mation about beta cell responses to insulin treatment
`and to inflammatory cytokines, as well as potential
`biomarkers of T2DM in skeletal muscle.
`
`References
`
`[4]
`
`[1] S. Falkmer and Y. Ostberg, Comparative morphology of pan-
`creatic islets in animals, in: The Diabetic Pancreas, B.M.
`Volk and K.F. Wellman eds., Plenum Press, New York, 1977,
`pp. 15–60.
`[2] D.A. Lang, D.R. Matthews, J. Peto and R.C. Turner, Cyclic
`oscillations of basal plasma glucose and insulin concentrations
`in human beings, New. Engl. J. Med. 301 (1979), 1023–1027.
`[3] A.L. Olson and J.E. Pessin, Structure, function, and regulation
`of the mammalian facilitative glucose transporter gene family,
`Annu. Rev. Nutr. 16 (1996), 235–256.
`J.C. Henquin, N. Ishiyama, M. Nenquin, M.A. Ravier and J.C.
`Jonas, Signals and pools underlying biphasic insulin secretion,
`Diabetes 51 (2002), S60–67.
`[5] D.L. Cook and C.N. Hales, Intracellular ATP directly blocks
`K+ channels in pancreatic -cells, Nature 311 (1984), 271–273.
`[6] S.W. Chouinard, F. Lu, B. Ganetzky and M.J. MacDonald,
`Evidence for voltage-gated potassium channel β-subunits with
`oxidoreductase motifs in human and rodent pancreatic beta-
`cells, Receptor Channels 7 (2000), 237–243.
`[7] H. Kindmark, M. K¨ohler, G. Brown, R. Br¨anstr¨am, O. Lars-
`son and P.O. Berggren, Glucose-induced oscillations in cyto-
`plasmic free Ca2+ concentration precede oscillations in mito-
`chondrial membrane potential in the pancreatic -cel1, J. Biol.
`Chem. 276 (2001), 30–36.
`[8] M. Prentki, New insights into pancreatic beta-cell metabolic
`signaling in insulin secretion, Eur. J. Endocrinol. 134 (1996),
`272–286.
`[9] R.A. Easom, CaM kinase II: a protein kinase with extraordi-
`nary talents germane to insulin exocytosis, Diabetes 48 (1999),
`675–684.
`J.T. Lawrence and M.J. Birnbaum, ADP-ribosylation factor 6
`regulates insulin secretion through plasma membrane phos-
`phatidylinositol 4,5-bisphosphate, Proc. Natl. Acad. Sci. USA
`100 (2003), 13320–13325.
`[11] A. Rabinovitch and W.L. Suarez-Pinzon, Role of cytokines
`in the pathogenesis of autoimmune diabetes mellitus, Rev.
`Endocr. Metab. Disord. 4 (2003), 291–299.
`
`[10]
`
`[12] T. Mandrup-Poulsen, Beta cell death and protection, Ann. N.Y.
`Acad. Sci. 1005 (2003), 32–42.
`[13] N.E. John, H.U. Andersen, S.J. Fey, P.M. Larsen, P. Roep-
`storff, M.R. Larsen, F. Pociot, A.E. Karlsen, J. Nerup, I.C.
`Green and T. Mandrup-Poulsen, Cytokine- or chemically de-
`rived nitric oxide alters the expression of proteins detected by
`two-dimensional gel electrophoresis in neonatal rat islets of
`Langerhan, Diabetes 49 (2000), 1819–1829.
`[14] P.M. Larsen, S.J. Fey, M.R. Larsen, A. Nawrocki, H.U. An-
`dersen, H. Kahler, C. Heilmann, M.C. Voss, P. Roepstorff,
`F. Pociot, A.E. Karlsen and J. Nerup, Proteome analysis of
`interleukin-1beta–induced changes in protein expression in rat
`islets of Langerhans, Diabetes 50 (2001), 1056–1063.
`[15] K. Nielsen, T. Sparre, M.R. Larsen, M. Nielsen, S.J. Fey, P.
`Mose Larsen, P. Roepstorff, J. Nerup and A.E. Karlsen, Protein
`expression changes in a cell system of beta-cell maturation
`reflect an acquired sensitivity to IL-1beta, Diabetologia 47
`(2004), 62–74.
`[16] S. Winer, H. Tsui, A. Lau, A. Song, X. Li, R.K. Cheung, A.
`Sampson, F. Afifiyan, A. Elford, G. Jackowski, D.J. Becker,
`P. Santamaria, P. Ohashi and H.M. Dosch, Autoimmune islet
`destruction in spontaneous type 1 diabetes is not beta-cell
`exclusive, Nat. Med. 9 (2003), 198–205.
`[17] S. Arif, T.I. Tree, T.P. Astill, J.M. Tremble, A.J. Bishop, C.M.
`Dayan, B.O. Roep and M. Peakman, Autoreactive T cell re-
`sponses show proinflammatory polarization in diabetes but a
`regulatory phenotype in health, J. Clin. Invest. 113 (2004),
`451–463.
`[18] E. Arioglu, A. Andewelt, C. Diabo, M. Bell, S.I. Taylor and
`P. Gorden, Clinical course of the syndrome of autoantibodies
`to the insulin receptor (type B insulin resistance): a 28-year
`perspective, Medicine (Baltimore) 81 (2002), 87–100.
`[19] S.C. Elbein, Perspective:
`the search for genes for type 2 di-
`abetes in the post-genome era, Endocrinology 143 (2002),
`2012–2018.
`[20] Y. Imai, N. Philippe, G. Sesti, D. Accili and S.I. Taylor, Ex-
`pression of variant forms of insulin receptor substrate-1 iden-
`tified in patients with noninsulin-dependent diabetes mellitus,
`J. Clin. Endocrinol. Metab. 82 (1997), 4201–4207.
`[21] D.L. Esposito, Y. Li, C. Vanni, S. Mammarella, S. Veschi, F.
`Della Loggia, R. Mariani-Costantini, P. Battista, M.J. Quon
`and A. Cama, A novel T608R missense mutation in insulin
`receptor substrate-1 identified in a subject with type 2 dia-
`betes impairs metabolic insulin signaling, J. Clin. Endocrinol.
`Metab. 88 (2003), 1468–1475.
`[22] E. Araki, M.A. Lipes, M.E. Patti, J.C. Bruning, B. Haag 3rd,
`R.S. Johnson and C.R. Kahn, Alternative pathway of insulin
`signalling in mice with targeted disruption of the IRS-1 gene,
`Nature 372 (1994), 186–190.
`[23] D.J. Withers, J.S. Gutierrez, H. Towery, D.J. Burks, J.M. Ren,
`S. Previs, Y. Zhang, D. Bernal, S. Pons, G.I. Shulman, S.
`Bonner-Weir and M.F. White, Disruption of IRS-2 causes type
`2 diabetes in mice, Nature 391 (1998), 900–904.
`[24] K. Savitsky, A. Bar-Shira, S. Gilad, G. Rotman, Y. Ziv, L.
`Vanagaite, D.A. Tagle, S. Smith, T. Uziel and S. Sfez, A single
`ataxia telangiectasia gene with a product similar to PI-3 kinase,
`Science 268 (1995), 1749–1753.
`[25] R. Rosmond, M. Chagnon and C. Bouchard, The Pro12Ala
`PPARgamma2 gene missense mutation is associated with obe-
`sity and insulin resistance in Swedish middle-aged men, Dia-
`betes Metab. Res. Rev. 19 (2003), 159–163.
`[26] G. Boden, Interaction between free fatty acids and glucose
`metabolism, Curr. Opin. Clin. Nutr. Metab. Care 5 (2002),
`545–549.
`
`MPI EXHIBIT 1074 PAGE 4
`
`MPI EXHIBIT 1074 PAGE 4
`
`

`

`M. Korc / Update on diabetes mellitus
`
`165
`
`[30]
`
`[29]
`
`[27] T.K. Lam, A. Carpentier, G.F. Lewis, G. Van De Werve, I.G.
`Fantus and A. Giacca, Mechanisms of the free fatty acid-
`induced increase in hepatic glucose production, Am. J. Physiol.
`Endocrinol. Metab. 284 (2003), E863–873.
`[28] M. Griffiths, P.R. Payne, A.J. Stunkard, J.P. Rivers and M.
`Cox, Metabolic rate and physical development in children at
`risk of obesity, Lancet 336 (1990), 76–78.
`J. Walston, K. Silver, C. Bogardus, W.C. Knowler, F.S. Celi,
`S. Austin, B. Manning, A.D. Strosberg, M.P. Stern, N. Raben,
`J.D. Sorkin, J. Roth and A.R. Shuldiner, Time of onset of
`non-insulin-dependent diabetes mellitus and genetic variation
`in the beta 3-adrenergic-receptor gene, N. Engl. J. Med. 333
`(1995), 343–347.
`J. Walston, K. Silver, H. Hilfiker, R.E. Andersen, M. Seibert,
`B. Beamer, J. Roth, E. Poehlman and A.R. Shuldiner, Insulin
`response to glucose is lower in individuals homozygous for
`the Arg 64 variant of the beta-3-adrenergic receptor, J. Clin.
`Endocrinol. Metab. 85 (2000), 4019–4022.
`[31] P. Peraldi and B. Spiegelman, TNF-alpha and insulin resis-
`tance: summary and future prospects, Mol. Cell. Biochem.
`182 (1998), 169–175.
`[32] V. Rotter, I. Nagaev and U. Smith, Interleukin-6 (IL-6) induces
`insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and
`tumor necrosis factor-alpha, overexpressed in human fat cells
`from insulin-resistant subjects, J. Biol. Chem. 278 (2003),
`45777–45784.
`[33] P.J. Klover, T.A. Zimmers, L.G. Koniaris and R.A. Mooney,
`Chronic exposure to interleukin-6 causes hepatic insulin re-
`sistance in mice, Diabetes 52 (2003), 2784–2789.
`[34] C. Weyer, T. Funahashi, S. Tanaka, K. Hotta, Y. Matsuzawa,
`R.E. Pratley and P.A. Tataranni, Hypoadiponectinemia in obe-
`sity and type 2 diabetes: close association with insulin resis-
`tance and hyperinsulinemia, J. Clin. Endocrinol. Metab. 86
`(2001), 1930–1935.
`[35] E. van Exel, J. Gussekloo, A.J. de Craen, M. Frolich, A.
`Bootsma-Van Der Wiel and R.G. Westendorp, Leiden 85 Plus
`Study. Low production capacity of interleukin-10 associates
`with the metabolic syndrome and type 2 diabetes: the Leiden
`85-Plus Study, Diabetes 51 (2002), 1088–1092.
`[36] K. Esposito, A. Pontillo, F. Giugliano, G. Giugliano, R.
`Marfella, G. Nicoletti and D. Giugliano, Association of low
`interleukin-10 levels with the metabolic syndrome in obese
`women, J. Clin. Endocrinol. Metab. 88 (2003), 1055–1058.
`[37] P.J. Blackshear and D.M. Haupt, Evidence against insulin-
`stimulated phosphorylation
`of
`calmodulin in 3T3-L1
`adipocytes, J. Biol. Chem. 264 (1989), 3854–3858.
`[38] H.B. Sadowski, T.T. Wheeler and D.A. Young, Characteriza-
`tion of initial responses to the inducing agents and changes dur-
`ing commitment to differentiation, J. Biol. Chem. 267 (1992),
`4722–4731.
`
`[41]
`
`[39] L. Wilson-Fritch, A. Burkart, G. Bell, K. Mendelson, J.
`Leszyk, S. Nicoloro, M. Czech and S. Corvera, Mitochon-
`drial biogenesis and remodeling during adipogenesis and in
`response to the insulin sensitizer rosiglitazone, Mol. Cell Biol.
`23 (2003), 1085–1094.
`[40] U. Edvardsson, H. Brockenhuus Von Lowenhielm, O. Panfilov,
`A.C. Nystrom, F. Nilsson and B. Dahllof, Hepatic protein ex-
`pression of lean mice and obese diabetic mice treated with per-
`oxisome proliferator-activated receptor activators, Proteomics
`3 (2003), 468–478.
`J.C. Sanchez, V. Converset, A. Nolan, G. Schmid, S. Wang, M.
`Heller, M.V. Sennitt, D.F. Hochstrasser and M.A. Cawthorne,
`Effect of rosiglitazone on the differential expression of
`diabetes-associated proteins in pancreatic islets of C57Bl/6
`lep/lep mice, Mol. Cell Proteomics. 1 (2002), 509–516.
`[42] U. Edvardsson, M. Bergstrom, M. Alexandersson, K.
`Bamberg, B. Ljung and B. Dahllof, Rosiglitazone
`(BRL49653), a PPARgamma-selective agonist, causes peroxi-
`some proliferator-like liver effects in obese mice, J. Lipid Res.
`40 (1999), 1177–1184.
`[43] K. Hojlund, K. Wrzesinski, P.M. Larsen, S.J. Fey, P. Roep-
`storff, A. Handberg, F. Dela, J. Vinten, J.G. McCormack, C.
`Reynet and H. Beck-Nielsen, Proteome analysis reveals phos-
`phorylation of ATP synthase beta -subunit in human skeletal
`muscle and proteins with potential roles in type 2 diabetes, J.
`Biol. Chem. 278 (2003), 10436–10442.
`[44] D.K. Simon and D.R. Johns, Mitochondrial disorders: clinical
`and genetic features, Annu. Rev. Med. 50 (1999), 111–127.
`J.A. Maassen, Mitochondrial diabetes: pathophysiology, clin-
`ical presentation, and genetic analysis, Am. J. Med. Genet. 115
`(2002), 66–70.
`[46] K.F. Petersen, S. Dufour, D. Befroy, R. Garcia and G.I. Shul-
`man, Impaired mitochondrial activity in the insulin-resistant
`offspring of patients with type 2 diabetes, N. Engl. J. Med. 350
`(2004), 664–671.
`J.G. Wilson, J.H. Lindquist, S.C. Grambow, E.D. Crook and
`J.F. Maher, Potential role of increased iron stores in diabetes,
`Am. J. Med. Sci. 325 (2003), 332–339.
`[48] G. Cavallini, B. Vaona, P. Bovo, M. Cigolini, L. Rigo, F.
`Rossi, E. Tasini, M.P. Brunori, V. Di Francesco and L. Frulloni,
`Diabetes in chronic alcoholic pancreatitis. Role of residual
`beta cell function and insulin resistance, Dig. Dis. Sci. 38
`(1993), 497–501.
`[49] F. Wang, M. Herrington, J. Larsson and J. Permert, The rela-
`tionship between diabetes and pancreatic cancer, Mol. Cancer
`2 (2003), 1–5.
`[50] S.E. Kahn, The relative contributions of insulin resistance
`and beta-cell dysfunction to the pathophysiology of Type 2
`diabetes, Diabetologia 46 (2003), 3–19.
`
`[45]
`
`[47]
`
`MPI EXHIBIT 1074 PAGE 5
`
`MPI EXHIBIT 1074 PAGE 5
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket