<<

REVIEW

The Role of Mitochondria in the Pathogenesis of Type 2 Diabetes

Mary-Elizabeth Patti and Silvia Corvera

Research Division, Joslin Diabetes Center (M.-E.P.), and Harvard Medical School (M.-E.P.), Boston, Massachusetts 02215; and Program in Molecular Medicine (S.C.), University of Massachusetts Medical School, Worcester, Massachusetts 01605 Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021

The pathophysiology of type 2 diabetes mellitus (DM) is varied and complex. However, the association of DM with obesity and inactivity indicates an important, and potentially pathogenic, link between fuel and energy homeostasis and the emergence of metabolic disease. Given the central role for mitochondria in fuel utilization and energy production, disordered mitochondrial function at the cellular level can impact whole-body meta- bolic homeostasis. Thus, the hypothesis that defective or insufficient mitochondrial function might play a potentially pathogenic role in mediating risk of type 2 DM has emerged in recent years. Here, we summarize current literature on risk factors for diabetes pathogenesis, on the specific role(s) of mitochondria in tissues involved in its pathophysiology, and on evidence pointing to alterations in mitochondrial function in these tissues that could contribute to the development of DM. We also review literature on metabolic phenotypes of existing animal models of impaired mitochondrial function. We conclude that, whereas the association between impaired mitochondrial function and DM is strong, a causal pathogenic relationship remains uncertain. How- ever, we hypothesize that genetically determined and/or inactivity-mediated alterations in mitochondrial ox- idative activity may directly impact adaptive responses to overnutrition, causing an imbalance between oxida- tive activity and nutrient load. This imbalance may lead in turn to chronic accumulation of lipid oxidative metabolites that can mediate insulin resistance and secretory dysfunction. More refined experimental strategies that accurately mimic potential reductions in mitochondrial functional capacity in humans at risk for diabetes will be required to determine the potential pathogenic role in human insulin resistance and type 2 DM. (Endocrine Reviews 31: 364–395, 2010)

I. Type 2 Diabetes Pathogenesis I. Type 2 Diabetes Pathogenesis A. Risk factors associated with type 2 diabetes II. General Overview of Mitochondrial Biology ype 2 diabetes mellitus (DM) in the United States and A. The dynamic morphology of mitochondria B. Mechanisms that control mitochondrial density and T around the world has reached epidemic proportions. capacity At present, 17.9 million people in the United States have III. Role of Mitochondria in Tissue-Specific Contexts been diagnosed with diabetes, with an additional 5.7 mil- A. Muscle lion undiagnosed (1). Together, this encompasses 8% of B. Adipose tissue C. Liver the population, and thus, diabetes is a major public health D. Pancreatic ␤-cells issue. In addition, current data indicate that 57 million IV. Experimental Strategies to Explore the Relationship be- Americans suffer from prediabetes (defined as fasting tween Mitochondrial Function and DM blood glucose between 100 and 125 mg/dl) (1). Diabetes A. PGC-1 ␣ and ␤ overexpression disproportionately affects specific ethnic populations, B. PGC-1 knockout models C. Other mitochondrial function defects with risk increased 1.8-fold in African-Americans, 1.7- V. Conclusions fold in Mexican-Americans, and 2.2-fold in Native Amer-

ISSN Print 0021-972X ISSN Online 1945-7197 Abbreviations: BMI, Body mass index; CoA, coenzyme A; COX, cytochrome oxidase; CPT1, Printed in U.S.A. carnitine palmitoylotransferase 1; DM, diabetes mellitus; ERR, estrogen-related receptor;

Copyright © 2010 by The Endocrine Society ETC, electron transport chain; FADH2, reduced flavin adenine dinucleotide; MIDD, mater- doi: 10.1210/er.2009-0027 Received July 2, 2009. Accepted December 24, 2009. nally inherited diabetes and deafness; mtDNA, mitochondrial DNA; NADH, reduced nic- First Published Online February 15, 2010 otinamide adenine dinucleotide; NASH, nonalcoholic steatohepatitis; NMR, nuclear mag- netic resonance; NRF, nuclear respiratory factor; OXPHOS, oxidative phosphorylation; PGC, PPAR␥ coactivator; PPAR, peroxisome proliferator-activated receptor; ROS, reactive oxygen species; RQ, respiratory quotient; TCA, tricarboxylic acid; UCP, uncoupling .

364 edrv.endojournals.org Endocrine Reviews, June 2010, 31(3):364–395 Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 365 icans. In addition to the major health consequences to glucose disposal during the hyperinsulinemic euglycemic individuals, including higher risk of death, heart disease, clamp or by iv glucose tolerance testing, is common in stroke, kidney disease, blindness, amputations, neuropa- high-risk individuals years before the onset of type 2 DM thy, and pregnancy-related complications, diabetes and its (27–29). However, insulin resistance is not predictive of complications result in a total cost of $174 billion in the diabetes in individuals without a family history of diabe- United States (2). By far, the largest proportion is derived tes, indicating that additional unidentified factors are nec- from type 2 DM, which accounts for more than 90% of essary for disease progression (30). diabetes. Unfortunately, the incidence of diabetes has Multiple mechanisms have recently emerged as poten- more than doubled in the past 25 yr, with 1.6 million new tial causes of insulin resistance and/or diabetes progres- cases diagnosed in adults in 2007 (2) and a projected in- sion, among them impaired mitochondrial capacity crease of 165% from 2000 to 2050 (4). and/or function; altered insulin signaling due to cellular Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 Intimately linked with the rise in diabetes prevalence is lipid accumulation, proinflammatory signals, and endo- the burgeoning epidemic of obesity around the world, par- plasmic reticulum stress; and reduced incretin-dependent ticularly in developed societies (5). In 2004, 17% of chil- and -independent ␤-cell insulin secretion. In this review, dren in the United States between ages 2 and 19 yr were we will focus on a critical assessment of the evidence link- overweight, and 32% of adults over age 20 were obese (6). ing mitochondrial function to diabetes pathogenesis, at Both obesity and related inactivity are likely to contribute both a cellular and whole-body level. to the pathogenesis of diabetes because the incidence of diabetes can be reduced by modest weight loss and exercise (7–9). In light of these findings, an important public health II. General Overview of Mitochondrial Biology goal should be to understand the complex pathophysiol- ogy of diabetes and to identify and target specific mech- Mitochondria are double-membrane organelles that serve anisms to prevent DM in at-risk individuals. multiple essential cellular functions (Fig. 1) mediated by thousands of mitochondrial-specific encoded by A. Risk factors associated with type 2 diabetes both the nuclear and mitochondrial genomes (31, 32). Multiple physiological abnormalities can be found in Although mitochondria are most often recognized for individuals with established type 2 DM, defined on the their role in generating the majority of cellular ATP via basis of elevations in fasting and/or postprandial glucose oxidative phosphorylation (OXPHOS), other essential (2). These include insulin resistance in muscle and adipose metabolic functions include the generation by the tricar- tissue, ␤-cell dysfunction leading to impaired insulin se- boxylic acid (TCA) cycle of numerous metabolites that cretion, increased hepatic glucose production, abnormal function in cytosolic pathways, oxidative catabolism of secretion and regulation of incretin hormones, and altered amino acids, ketogenesis, ornithine cycle activity (“urea balance of central nervous system pathways controlling cycle”), the generation of reactive oxygen species (ROS) food intake and energy expenditure. Given this diverse with important signaling functions (33, 34), the control of constellation of abnormalities in multiple tissues and the cytoplasmic calcium (35, 36), and the synthesis of all cel- secondary consequences of established hyperglycemia and lular Fe/S clusters, protein cofactors essential for cellular hyperlipidemia, it is difficult to identify the primary events functions such as protein translation and DNA repair (37). that lead to the development of diabetes. To address this The rate-limiting first step in steroidogenesis also occurs in key clinical and scientific question, it is important not only mitochondria, thus linking mitochondrial function to to determine abnormalities associated with established endocrine homeostasis (38–41). This multiplicity of disease, but also to identify underlying metabolic char- organelle functions explains the variability in patho- acteristics preceding the onset of disease in at-risk physiology, severity, and age of onset of the increasing individuals. number of diseases recognized to arise from primary or Risk factors for the development of and/or progression secondary alterations in specific mitochondrial path- of type 2 DM include: 1) genetics (10–16), exemplified by ways (37, 42–44). the high risk of type 2 DM in particular ethnic groups (17) and the high concordance rates in monozygotic twin pairs A. The dynamic morphology of mitochondria (18); and 2) both prenatal and postnatal environmental In the thin sections observed by electron microscopy factors, including suboptimal intrauterine environment and shown in most textbooks, mitochondria appear (19, 20), low birth weight (19, 21), obesity (22, 23), in- as discrete, small, bean-shaped, double-membrane or- activity (24), gestational diabetes (25), and advancing age ganelles. However, more recent studies based on light mi- (26). Several longitudinal studies have indicated that in- croscopy in live cells have revealed that mitochondria exist sulin resistance, measured as reduced insulin-stimulated as a reticulum that is in continuous communication 366 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395 Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021

FIG. 1. Basic structural and functional features of the mitochondrial reticulum (illustrated from left to right). The mitochondrial reticulum is composed of an inner and outer membrane, between which lies the intermembrane space, and a matrix contained within the inner membrane. The surface of the inner membrane is folded into cristae. The organization and distribution of the mitochondrial reticulum is controlled by interactions with cytoskeletal elements such as microtubules. The matrix contains the enzymatic machinery for fatty acid ␤ oxidation, which generates acetyl-CoA from acyl chains, and reducing equivalents in the form of NADH and FADH2 in the process. Acetyl-CoA fuels the TCA cycle, which also produces NADH and FADH2. These donate electrons to the ETC, leading to the generation of a proton gradient across the inner mitochondrial membrane. Dissipation of this gradient through the mitochondrial ATPase generates ATP. Delay of electron transport by the ETC results in the production of ROS, which can activate UCPs that dissipate the proton gradient without producing ATP. The electrochemical gradient also causes cytoplasmic Caϩϩ to enter the matrix, buffering cytoplasmic Caϩϩ levels and promoting TCA cycle flux. Mitochondria are also crucial in the generation of iron-sulfur clusters that form the prosthetic group of numerous proteins involved in multiple cellular pathways. The mitochondrial reticulum undergoes continuous fusion and fission reactions that involve both the inner and outer mitochondrial membranes, allowing redistribution of matrix content, such as mtDNA, within the reticulum. The proteins that compose all mitochondrial machineries are encoded both by mtDNA and by nuclear DNA. The master transcription factor operating on mtDNA is TFAM, which is encoded in the nuclear genome. The expression of mitochondrial in the nucleus is driven by numerous transcription factors, which are in turn controlled by specific coactivators and corepressors that respond to cellular energy demands. through dynamic fusion and fission events, moving ac- products. The ETC transports electrons from donors tively to different regions of the cell through interactions (NADH at complex I, FADH2 at complex II) to a final with the cytoskeleton (Fig. 2). The mitochondrial reticu- acceptor, molecular oxygen, forming H2O at complex IV. lum is composed of an outer and an inner membrane, The transport of electrons is accompanied by release of between which is the intermembrane space, and a matrix large amounts of free energy, most of which is harnessed limited by the inner membrane (Fig. 1). The area of the for the translocation of protons from the matrix to the inner membrane can be greater than that of the outer mem- intermembrane space; the remainder is dissipated as heat brane due to the presence of cristae, inner membrane in- (Fig. 3). The energy contained in the proton electrochem- vaginations that contain all the transmembrane proteins ical gradient generated by the ETC is then coupled to ATP of the electron transport chain (ETC) as well as the mito- production as protons flow back into the matrix through chondrial ATPase (45–47). The mitochondrial matrix the mitochondrial ATPase. Thus, OXPHOS results from contains the components of the TCA cycle and of the ␤-ox- electron transport, the generation of a proton gradient, idation pathway, which provide reduced nicotinamide ad- and subsequent proton flux coupled to the mitochondrial enine dinucleotide (NADH) and reduced flavin adenine ATPase. Each of these steps can vary in efficiency; for dinucleotide (FADH2) to the ETC. example, the exact stoichiometry between electron flow The ETC is composed of four large multisubunit com- and proton pumping, or between proton pumping and plexes (complexes I to IV) with more than 85 individual ATP synthesis varies depending on the probability of loss Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 367 Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021

FIG. 2. Visualization of the dynamic nature of the mitochondrial reticulum in a cultured muscle cell. C2C12 mouse myoblasts were FIG. 3. Coupled and uncoupled respiration. Electrons derived from stained with MitoTracker green and imaged in culture at 37 C. Images reduced donors NADH and FADH2 are transported within the ETC to of a segment of the cell captured at 30-sec intervals are shown on the molecular oxygen, producing water. The flow of electrons within the left. The comparison between successive frames reveals scission events ETC is coupled to translocation of protons due to the large amount of (arrowheads), branching events (V), and fusion events (brackets) free energy released during electron transport. The remainder of this occurring at 30-sec intervals. free energy is released as heat. The proton gradient thus produced is dissipated through the mitochondrial ATPase, and the consequent decrease in free energy drives ATP synthesis. This process is known as of electrons from the ETC before reaching complex IV OXPHOS, or coupled respiration. Under circumstances where NADH and on non-ATPase-coupled proton leak through the and FADH2 are available, but movement of electrons down the respiratory chain is slow, some of those electrons will be released from inner mitochondrial membrane [e.g., via uncoupling the respiratory chain and reduce molecular oxygen, forming the Ϫ proteins (UCPs)]. superoxide anion O2 , hydrogen peroxide, and the hydroxyl radical The high electronegative potential generated by the OHϪ. These are the main ROS formed at steady state. Accumulation of proton gradient also drives the rapid entry of Caϩϩ into ROS activates UCPs, which dissipate the proton gradient without producing ATP, resulting in uncoupled respiration. the mitochondrial matrix, buffering its concentration in ϩϩ the cytoplasm. In the mitochondrial matrix, Ca can generated in the mitochondrial matrix, to be shared within stimulate flux through the Krebs cycle by stimulating the entire mitochondrial reticulum. Although the molec- ϩϩ dehydrogenase activities (36). The exit of Ca from ular machinery of mitochondrial fusion and fission has the matrix is driven by electroneutral exchange with been elucidated (48), it has only recently been established ϩ ϩ Na or H . that mitochondrial fusion and fission also contribute mul- The ETC is also a potent source of ROS. Loss of elec- tiple other mitochondrial functions, including the control trons from the ETC can result in reduction of oxygen to of cellular calcium handling, ROS production, and ener- Ϫ form O2 , which can be dismutated to H2O2 and subse- getic output (49–51). Moreover, human diseases arising Ϫ quently converted to the hydroxyl radical, OH . These from in conserved elements of the mitochon- three products constitute the major ROS formed during drial fusion machinery have been identified, such as respiration. As the name implies, these species are highly Charcot-Marie-Tooth type 2A caused by mutations in reactive, and acute, very high elevations, or more chronic mitofusin 2, and autosomal dominant optic atrophy, elevations can be extremely damaging to the cell. ROS caused by mutations in optic atrophy 1 (49). A role for generation is more likely to occur when the proton gra- mitochondrial fusion machinery in metabolic control has dient is large and electron carriers are highly reduced, e.g., also been suggested by the findings that mitofusin 2 levels when ADP is rate-limiting for ATP production or when are controlled during muscle development and are reduced availability of O2 is limiting. Uncoupling proteins are con- in both obesity and type 2 DM in parallel with insulin sidered to be natural regulators of this process, responding resistance (52). to and controlling ROS production by mitigating the for- mation of a large proton gradient. B. Mechanisms that control mitochondrial density The mitochondrial matrix also contains the circular and capacity mitochondrial DNA (mtDNA) molecule, which encodes The term mitochondrial biogenesis is often used to de- for 37 genes (13 of which are subunits of the ETC). Trans- scribe the generation of more mitochondria in response to lation of these proteins occurs within the mitochondrial increased energy demands, or the multiplication of mito- matrix, utilizing mtDNA-encoded rRNA and tRNA. chondria necessary for cell growth and division. However, Mitochondrial fission and fusion allow the transcrip- the copy number of specific mitochondrial proteins and tional products of mtDNA, as well as multiple metabolites the functional capacity of each distinct mitochondrial 368 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395 pathway may be very variable between different tissues required for mitochondrial biogenesis during develop- and between different physiological conditions. Thus, the ment (59), they are necessary for the expression of the full term mitochondrial biogenesis can be ambiguous because complement of proteins of mitochondrial OXPHOS and multiple parameters, including mtDNA copy number, fatty acid ␤-oxidation pathways in muscle and brown ad- mitochondrial density, levels of specific mitochondrial ipose tissue (59–69). Moreover, PGC-1␣ and PGC-1␤ are proteins, and mitochondrial functional output may vary crucial for the rapid bursts in mitochondrial proliferation independently of each other. For example, the prolifera- that accompany perinatal heart and brown adipose tissue tion of mitochondria occurring to sustain hyperplastic development (59). These data support the concept that growth is probably very different from that occurring to mitochondrial adaptation to specific energy needs is support hypertrophic growth in any given tissue, and the mediated by PGC-1␣ and PGC-1␤; by contrast, mito- regulatory mechanisms controlling these adaptive changes chondrial expansion during cell proliferation is more Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 are likely to be distinct. likely to depend on serum-responsive coactivators such as PPRC (70). 1. Transcriptional control mechanisms The role of corepressors in the transcriptional control Although we know very little about specific mecha- of energy metabolism genes is less extensively studied. nisms that control different modalities of mitochondrial However, evidence in cultured cells and in mouse models biogenesis, it is clear that these mechanisms require co- points to a critical role of the corepressor RIP140 in con- ordination between the nuclear and mitochondrial ge- trolling important aspects of mitochondrial energy me- nomes. Transcription of the mitochondrial genome is tabolism in both adipose tissue and muscle (71–75). under the control of a single transcription factor, Tfam, RIP140 suppresses UCP1 through interaction with spe- which is encoded by the nuclear genome. In turn, Tfam cific enhancer elements and also suppresses expression of expression is regulated by the transcription factors NRF genes involved in ␤-oxidation and respiratory chain as- (nuclear respiratory factor)-1 and NRF-2, which spe- sembly. RIP140 also interacts directly with many of the cifically activate numerous nuclear-encoded genes in- transcription factors coactivated by PGC-1␣ (76). The volved in mitochondrial respiration (53, 54). Thus, mechanisms that control the balance between PGC-1 co- through NRF-stimulated expression of Tfam, the tran- activators and RIP140 and other corepressors are not clear scription of the mitochondrial genome is stimulated in but are likely to represent key regulatory mechanisms of coordination with that of nuclear-encoded mitochondrial energetic adaptation. genes. The expression of many other mitochondrial genes is controlled by additional nuclear transcription factors, including peroxisome proliferator-activated 2. Posttranscriptional control mechanisms receptor (PPAR) ␣, PPAR␦, estrogen-related receptor The expansion of the mitochondrial reticulum requires (ERR) ␣/␥, and Sp1, which can induce expression of not only the expression of genes encoding mitochondrial mitochondrial genes in a tissue-dependent and physio- proteins but also the import of these into the mitochon- logical context-dependent manner (55). drial space (77–80) and the coordinated expansion of mi- A high level of transcriptional coordination is required tochondrial membranes. Mitochondrial inner and outer to ensure coupling of mitochondrial activity to other met- membranes have distinct lipid compositions that differ abolic activities within the cell and to mediate appropriate from that of other membrane-bound organelles and from parallel changes in all components of multiprotein com- the plasma membrane. Specific features of mitochondrial plexes. This coordination is accomplished through the ac- membranes are their relative lack of cholesterol and the tion of transcriptional coactivators and corepressors. The high content of cardiolipin, which is unique to mitochon- best studied coactivators of mitochondrial gene transcrip- drial membranes and essential for the proper assembly and tion are members of the PPAR␥ coactivator (PGC) family, function of the respiratory chain (81–83). Mitochondrial including PGC-1␣, PGC-1␤ (56, 57), and PPRC, a related lipids are most likely synthesized in the endoplasmic serum-responsive coactivator (58). These respond to cel- reticulum (the primary site of lipid biosynthesis in eu- lular energy-requiring conditions such as cell growth, hyp- karyotic cells) and transferred to mitochondria via as- oxia, glucose deprivation, and exercise (55) to activate yet unidentified mechanisms. However, recent work transcription factors promoting mitochondrial remodel- has identified mechanisms regulating the synthesis of ing and/or biogenesis, thus restoring cellular energetics. cardiolipin and phosphatidylethanolamine in mito- For example, PCG-1␣ is highly expressed in muscle, liver, chondria inner membranes via the action of mitochon- and brown fat, and expression is further increased in these drial prohibitins (84). In addition, cardiolipin synthesis tissues in response to exercise, fasting, and cold exposure, requires the mitochondrial translocator assembly and respectively. Although PGC-1␣ and -␤ do not appear to be maintenance protein Tam41, revealing a mechanism for Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 369 the coordination of protein import and mitochondrial sition to carbohydrate oxidation (increased RQ) during membrane lipid assembly (85). the fed state. Availability of fuels, particularly lipids, and The area and composition of the mitochondrial inner capacity to oxidize them within mitochondria are also and outer membranes must be tailored to accommodate critical for sustained exercise. Thus, mitochondrial func- the specific components of mitochondria from different tional capacity is likely to directly affect muscle metabolic cells and tissues, which are each likely to have optimal lipid function and, because of its large contribution to total composition and density. This essential requirement for body mass, to have a significant impact on whole-body specific lipid composition is underscored by the morpho- metabolism. This possibility is supported by the findings logical and functional alterations in mitochondria seen in of increased mitochondrial content in skeletal muscle in an Barth syndrome, a disorder arising from mutations in a individual with hypermetabolism and resistance to weight lipid acyltransferase, tafazzin (41, 86). The resulting al- gain (Luft syndrome) (90). Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 terations in cardiolipin structure cause profound changes in the assembly and distribution of respiratory chain com- 2. Potential mechanisms by which impaired muscle ponents within mitochondrial cristae (84, 87, 88). Inter- mitochondrial oxidative function could result in estingly, lymphoblasts from patients with Barth syndrome insulin resistance can produce ATP at normal levels but display an expanded Skeletal muscle is the largest insulin-sensitive organ mitochondrial reticulum (89). These observations under- in humans, accounting for more than 80% of insulin- score the existence of mechanisms that can compensate in stimulated glucose disposal. Thus, insulin resistance in part for specific mitochondrial deficiencies. this tissue has a major impact on whole-body glucose Given the complex and dynamic structure of mitochon- homeostasis. Indeed, multiple metabolic defects have dria and the diversity and physiological importance of been observed in muscle from insulin-resistant but nor- their multiple functions, assessing the role of mitochon- moglycemic subjects at high risk for diabetes develop- dria in human pathology requires a comprehensive char- ment, including: 1) reduced insulin-stimulated glycogen acterization not only of mitochondrial structure and synthesis (27, 91, 92); 2) alterations in insulin signal abundance, but also of the pathways that compensate transduction (93); and 3) increased muscle lipid accu- for suboptimal mitochondrial capacity and functional mulation (94). Although it remains unclear whether any output—which may then modify disease severity and of these defects play a causal role in insulin resistance, progression. In the following sections, we will critically intramyocellular lipid excess strongly correlates with analyze the findings that have suggested a role for mi- the severity of insulin resistance, even after correction tochondrial function in the establishment of diabetes for the degree of obesity (94), and has been observed in risk and the gaps in our knowledge that must be filled muscles of multiple fiber types (95). Moreover, lipid to determine the merits of this hypothesis. excess has been linked experimentally to induction of insulin resistance (96) and alterations in insulin signal transduction (97–99). III. Role of Mitochondria in Thus, one possible mechanism by which impaired mi- Tissue-Specific Contexts tochondrial function might contribute to insulin resis- tance is via altered metabolism of fatty acids. Increased A. Muscle tissue lipid load, as with obesity, and/or sustained inac- 1. Role of mitochondria in muscle tivity, may lead to the accumulation of fatty acyl coenzyme Mitochondria are particularly important for skeletal A (CoA), diacylglycerols, ceramides, products of incom- muscle function, given the high oxidative demands im- plete oxidation, and ROS, all of which have been linked posed on this tissue by intermittent contraction. Mito- experimentally to reduced insulin signaling and action chondria play a critical role in ensuring adequate levels of (96–102). Additional mechanisms potentially linking im- ATP needed for contraction by the muscle sarcomere. This paired mitochondrial oxidative function to insulin re- high-level requirement for ATP by sarcomeres has likely sistance include: 1) reduced ATP synthesis for energy- contributed to the distinct subsarcolemmal and sarco- requiring functions such as insulin-stimulated glucose mere-associated populations of mitochondria in muscle. uptake; 2) abnormalities in calcium homeostasis (neces- Moreover, muscle cells must maintain metabolic flexibil- sary for exercise-induced glucose uptake) (103–105); and ity, defined as the ability to rapidly modulate substrate 3) reduced ATP production during exercise (106), poten- oxidation as a function of ambient hormonal and ener- tially contributing to reduced aerobic capacity, muscle getic conditions. For example, healthy muscle tissue pre- fatigue, and decreased voluntary exercise over time— dominantly oxidizes lipid in the fasting state, as evidenced further feeding a vicious cycle of inactivity-fueled insulin by low respiratory quotient (RQ), with subsequent tran- resistance. 370 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395

3. Evidence for reduced muscle mitochondrial oxidative particularly in subsarcolemmal fractions (114). Interest- function in DM ingly, this fraction is also characterized by even greater An important early clue suggesting that muscle mito- reductions in OXPHOS activity (114). chondrial oxidative dysfunction may be associated with Nuclear magnetic resonance (NMR) spectroscopy has insulin resistance in humans was the series of observations also been used to assess mitochondrial function in vivo, by Simoneau and Kelley that obesity is associated with with studies finding similar reductions in oxidative func- reductions in citrate synthase, malate dehydrogenase, car- tion in both insulin resistance and type 2 DM. For exam- nitine palmitoylotransferase 1 (CPT1), and cytochrome ple, rates of mitochondrial OXPHOS in offspring of type oxidase (COX) activity in the fasting state (107, 108) and 2 diabetic subjects, as assessed by 31P spectroscopy, are with parallel increases in activity of the glycolytic reduced by 30% in the fasting state (117), and TCA cycle 13 and (109). Moreover, flux, modeled using rates of 4- C-glutamate enrichment Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 oxidative activity (e.g., citrate synthase, acyl CoA dehy- during infusion of 13C-acetate, is reduced by 30% (118). drogenase) is a robust correlate of insulin sensitivity, even The magnitude of these changes is strikingly similar to the better than either im triglycerides or long-chain fatty acyl 38% lower muscle mitochondrial density, assessed by CoA (110). Furthermore, leg balance studies demon- electron microscopy, in this same population—again sug- strated that obesity-linked insulin resistance and diabetes gesting that decreased mitochondrial density might be an are both associated with reduced fasting lipid oxidation, important factor in reduced oxidative capacity in individ- as indicated by higher RQ, as well as inability to suppress uals with a family history of diabetes. lipid oxidation and switch to carbohydrate oxidation in Alterations in intrinsic function of mitochondria have response to meals/insulin stimulation (111), a state termed also been identified in isolated mitochondria from humans “metabolic inflexibility” (112). Impaired flexibility also with insulin resistance and DM. Mogensen et al. (119) correlates with intramyocellular accumulation of lipids observed decreases in maximal ADP-stimulated respira- (107), and 24-h RQ can predict subsequent weight gain tion (state 3, malate and pyruvate as substrates) in (110, 113). Together, these data suggest that an intrinsic mitochondria isolated from obese subjects with DM as defect in multiple components of oxidative metabolism, or compared with obesity alone; these differences persisted altered regulation, may contribute to the development of even after normalization to citrate synthase activity. Thus, both obesity and insulin resistance. these data suggest that in addition to decreased mitochon- The diminished capacity for appropriate regulation of drial density, there is an additional intrinsic defect(s) in oxidative metabolism observed in the above studies could TCA, OXPHOS, membrane potential, or adenine nucle- be linked to reduced mitochondrial function due to: 1) otide transporters in mitochondria of individuals with es- abnormal mitochondrial density and/or in vivo function; tablished diabetes. and/or 2) intrinsic defects in oxidative metabolism of lip- Such underlying functional defects may be subtle at ids or other substrates. Multiple studies suggest that hu- baseline but may be unmasked during acute energetic man insulin resistance is indeed accompanied by impaired stress. For example, short-term exercise normally in- in vivo mitochondrial oxidative function—in turn linked, creases ATP synthesis rates. However, this adaptive re- at least in part, to reduced mitochondrial density. Ritov et sponse is completely mitigated/abolished in nonobese al. (114) demonstrated that the enzymatic activity of first-degree relatives of type 2 diabetics—despite normal OXPHOS complex I, as assessed by the activity of rote- basal ATP synthesis rates (106). Similarly, insulin-stimu- none-sensitive NADH:O2 oxidoreductase, was reduced lated ATP synthesis is reduced by more than 90% in nono- by about 40% in skeletal muscle biopsy samples from bese first-degree relatives of type 2 diabetics (120), more individuals with type 2 DM and by 20% in obese individ- than would be expected from the 30% decrease in mito- uals. Similarly, Boushel et al. (115) found modest reduc- chondrial density and oxidative function observed in the tions in ADP and succinate-stimulated oxygen consump- same population. Because these short-term experimental tion in permeabilized muscle fibers from obese individuals protocols (several hours in duration at most) would not be with type 2 DM. In each of these studies, differences in expected to alter mitochondrial density, DNA content, or oxidative capacity did not remain after normalization for number, these data strongly suggest that inability to ap- mitochondrial mass by citrate synthase activity or mtDNA propriately modulate oxidative function in response to the content, respectively, suggesting that reduced mitochon- prevailing energetic environment is a signature of insulin drial mass might be a major contributor. This possibility resistance and diabetes risk. is consistent with electron microscopy demonstrating di- Analysis of global patterns has also minished mitochondrial size in obesity and diabetes (116), demonstrated a 20–30% reduction in mRNA expression Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 371 levels for multiple nuclear-encoded genes of the OXPHOS DNA content, potentially mediated by chronic fatty acid pathway in humans with type 2 DM (121–123). Impor- activation of PPAR nuclear receptors (130–132). Simi- tantly, similar reductions in OXPHOS gene expression larly, relatively short-term reductions in serum fatty acids have been observed in some, but not all, populations of and intracellular fatty acyl CoA levels mediated by acipi- insulin-resistant, but completely normoglycemic, individ- mox treatment in healthy humans are associated with uals (122, 124).1 These differences may reflect popula- reduced expression of nuclear-encoded mitochondrial ox- tion-specific differences in obesity, physical fitness, or idative genes—in parallel with enhanced insulin sensitivity ethnicity. Interestingly, a recent study of Asian Indian (294). Together, these seemingly disparate data suggest subjects found no correlation between changes in that genetic background (127), age at dietary intervention, OXPHOS gene expression and insulin resistance (125). In specific dietary lipid composition, and duration of diet these individuals, expression of OXPHOS and TCA cycle may be important variables to consider when analyzing Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 genes, mtDNA content, and ATP production rates were the interaction between OXPHOS gene expression and actually higher in both nondiabetic and diabetic individ- diet. Moreover, alterations in OXPHOS gene expression uals compared with Northern European controls, despite may be a secondary response to an underlying primary lower overall insulin sensitivity. However, circulating trig- defect in oxidative metabolism, reflecting attempts to lycerides were significantly elevated in both nondiabetic compensate for reductions in mitochondrial capacity (in- and diabetic individuals of Asian Indian origin (125). creased OXPHOS expression), or the deleterious effects of These results also raise the question of whether levels of lipid overload and accumulation on transcription of OXPHOS gene expression and function must be consid- OXPHOS genes (decreased OXPHOS expression), or a ered relative to the oxidative fuel load in an individual. For mixture of both. Additionally, because OXPHOS gene example, high OXPHOS expression in the population expression is coordinately regulated, patterns of differen- mentioned above may still be inadequate for appropriate tial OXPHOS expression may be more readily detectable and complete oxidation of a chronic high load of circu- in disease states, yet not necessarily mirror other aspects of lating lipids, whereas lower OXPHOS levels may be mitochondrial oxidative capacity. sufficient under conditions of a low circulating lipid Reduced physical fitness is associated with reduced load (see Fig. 5). muscle OXPHOS gene expression. In humans, maximal Such data also highlight the importance of considering oxygen uptake is robustly correlated with OXPHOS gene additional aspects of oxidative mitochondrial function be- expression (133). Similarly, in rats bred for low aerobic yond OXPHOS expression or capacity. For example, pri- capacity over multiple generations, expression of several mary myotubes isolated from obese humans with type 2 OXPHOS genes is markedly reduced, even in the absence DM display reduced basal lipid oxidation and insulin- stimulated glucose oxidation with no differences in of obesity (134). Conversely, OXPHOS expression can be OXPHOS gene expression (126). Thus, defects in lipid increased with exercise training (133, 135), a potent in- oxidation in DM can be significant contributors to disor- sulin sensitizer. dered oxidative metabolism even in the absence of detect- Genetic and epigenetic modifications may also con- able alterations in OXPHOS gene expression or function. tribute to reduced expression of OXPHOS genes in type 2 DM. For example, expression of COX7A1, a complex 4. Factors affecting OXPHOS gene expression in muscle IV gene down-regulated in type 2 DM, is heritable (50– Several conditions associated with susceptibility to in- 72% heritability, as assessed by analysis in monozy- sulin resistance, including obesity, lipid accumulation, gotic and dizygotic twins), indicating a strong genetic and aging, have all been associated with reduced nuclear- or shared familial environmental contribution (136). encoded OXPHOS gene expression. Reduced OXPHOS Similar patterns are observed for the complex I gene gene expression has been observed in response to genetic NDUFB6 (137) and the ATP synthase component and nutritional obesity (127), short-term high-fat feeding ATP5O (138). Indeed, expression of nuclear-encoded (even in humans) (128), lipid infusion (129), and lipid OXPHOS genes is significantly more concordant be- loading of myotubes (127). However, these responses are tween monozygotic twins than expected and is the top- not observed in all studies of high-fat feeding; in fact, some ranking gene set for concordance in pathway analysis of studies demonstrate that high-fat feeding is associated global gene expression. Mediators of mitochondrial with increased numbers of mitochondrial protein and biogenesis, including ERR␣, may contribute to the 1 Patti ME, Liu M, Zin W, Lerin C, Dreyfuss J, Vokes M, Schroeder J, Tatro E, Park P, Kohane I, Kasif S, Goldfine AB, submitted. Transcriptome analysis reveals parallel dysregulation of oxidative metabolism and inflammation in muscle and adipose tissue with progression of insulin resistance in humans. 372 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395 strong heritability of OXPHOS components.2 Interest- 124, 137). In turn, PGC-1␣ expression may also be re- ingly, epigenetic mechanisms may also contribute to duced as a consequence of promoter methylation (146) or these patterns because reduced expression parallels in- caused by insulin itself (145), obesity (126), and sustained creased DNA methylation of both the COX7A1 pro- lipid exposure (126). For example, saturated fatty acids moter (136) and NDUFB6 (137, 139). reduce PGC-1␣ promoter transcriptional activity and ex- Aging is also linked to impaired oxidative function pression in cultured myotubes, in parallel with reduced

(140) in parallel with reductions in OXPHOS gene ex- OXPHOS expression and O2 consumption (127). PGC-1 pression, including COX7A1, NDUFB6, and ATP50 activity can also be modulated at the level of translation (136–138). It is unclear at this time whether this is a direct and by posttranscriptional changes, including inhibitory effect of aging per se or related to reduced physical fitness, GCN5-mediated acetylation (147) and stimulatory sirtuin 1 mediated deacetylation (148). These multiple modes of increased tissue lipid accumulation, or other factors ac- Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 companying typical patterns of aging. Genetic polymor- PGC-1␣ regulation are likely to have evolved from the phisms may also influence age-dependent reductions in need to adapt mitochondrial energy metabolism in re- expression (137). sponse to increasingly diverse inputs. A key question is whether the changes in OXPHOS In summary, insulin resistance has been associated with gene expression observed in type 2 DM are secondary alterations in skeletal muscle mitochondrial oxidative features of the diabetes metabolic environment such as function and its transcriptional regulatory pathways. hyperglycemia or insulin resistance. Reductions in However, several lines of evidence suggest that this may OXPHOS gene expression in patients with established not be a causal relationship in all situations. First, oxida- type 2 DM can be partially normalized by insulin treat- tive dysfunction is not observed in all insulin resistant in- ment (123). Expression of multiple OXPHOS genes is also dividuals (125). Second, oxidative activity is determined markedly reduced in mice made insulin deficient by treat- by the need to generate energy to meet cellular demands, ment with the ␤-cell toxin streptozotocin, and can be e.g., contraction and ion transport; thus oxidative capac- normalized by insulin (141). Similarly, withdrawal of ity is not likely to be limiting in the resting state in muscle insulin in individuals with type 1 diabetes reduces muscle (3). Rather, alterations in relative utilization of substrates, OXPHOS gene expression and ATP production rates an imbalance between fuel load and cellular energy re- (142). Short-term experimental induction of acute hyper- quirements, and/or differential thresholds for generation glycemia in humans does not fully mirror this pattern of of or resolution of oxidative stress in this setting may con- tribute to differential susceptibility to insulin resistance in gene expression (143), suggesting that the response to in- muscle. These concepts are examined more fully in the sulin deficiency is not completely due to resultant hyper- conclusion (Section V). glycemia. Moreover, experimental insulin therapy does not modulate mitochondrial respiration (144), so mech- anisms linking insulin action with OXPHOS gene expres- B. Adipose tissue sion remain unclear. 1. Roles of mitochondria in adipose tissue Changes in the levels of OXPHOS and other oxidative The role of adipose tissue mitochondria is most appar- genes must occur in response to cellular energetic and met- ent in brown adipose tissue, where flux through the ETC abolic needs, and in a coordinated manner that ensures the generates heat in the process of thermogenesis, a poten- stoichiometric assembly of the products of distinct genes tially important mechanism regulating systemic metab- into functional complexes. As in other tissues, the coor- olism even in adult humans (149–152). In this tissue, dination of OXPHOS gene expression in muscle is medi- electron transport is greatly accelerated due to tissue-spe- ated in part by the action of coactivators and corepressors. cific expression of the mitochondrial UCP1. UCP1 hinders PGC-1␣ has been recognized as an important coactivator the establishment of, or dissipates, a proton gradient of in skeletal muscle, contributing to fiber type determina- sufficient magnitude to sustain the synthetic activity of the tion, glucose uptake, and oxidative capacity (see Section mitochondrial ATPase (150, 153–155), thus driving con- IV. A). Moreover, alterations in muscle PGC-1␣ and -␤ tinuous accelerated electron transport. UCP1-mediated mRNA expression are observed in humans with insulin uncoupling alone, however, cannot fully account for the resistance—being reduced by nearly 50% in muscle from large thermogenic capacity of brown adipocytes in the individuals with diabetes (122, 145) and in some popula- absence of mechanisms that ensure continuous substrate tions of normoglycemic insulin-resistant humans (121, delivery to the ETC. Thus, brown adipocyte mitochondria 2 Stender-Petersen KL, Poulsen P, Butte A, Jensen CB, Yee J, Leykin I, Vaag A, Pedersen O, Patti ME, manuscript under review. Gene expression analysis in monozygotic twins reveals heritable contributions to PGC-1/ERR pathways. Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 373 also contain high levels of CPT1b, which is critical for the (168), and regional differences in metabolic activity can be entry of fatty acids into the mitochondria for ␤-oxidation. linked to varying mitochondria densities (169). Higher ␤-Oxidation, in turn, generates large amounts of reducing mitochondrial density and even UCP1 can be induced in equivalents for the ETC. response to pharmacological or genetic alterations of White adipocytes have been described to contain low white adipocytes (170–177), suggesting that white adi- levels of mitochondria, which is indeed the case when pose tissue could potentially be induced to acquire more compared with brown adipocytes or muscle. However, oxidative metabolic phenotypes, promoting increased fuel mitochondrial density increases dramatically, and mi- consumption and thus energy expenditure. Whether re- tochondrial remodeling occurs during white adipocyte spiratory chain uncoupling mediated through the induc- differentiation (156–158), suggesting that mitochondrial tion of UCP1 in white adipocytes alone could reduce free functions are required to support the multiple biological fatty acid release, or whether an additional increase in Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 roles of mature white adipocytes. Interestingly, a recent mitochondrial oxidative capacity would be required, is compendium of mitochondrial proteins from 14 different debated (178–182). mouse tissues indicates that white adipocyte mitochondria Gain-of-function studies in mice where ectopic expres- contain a more diverse protein repertoire than mitochon- sion of UCPs mitigate diet-induced obesity support the dria from heart, skeletal muscle, or brain (31). Thus, white notion that uncoupling could be sufficient (183, 184). adipocyte mitochondria appear to be equipped for a However, UCP1 expression in adipocytes driven by the broader array of functions compared with mitochondria aP2 promoter failed to significantly raise resting metabolic in tissues that must sustain rapid bursts of energy-requir- rate (185). Moreover, in cultured adipocytes, ectopic ex- ing processes. Among the mitochondrial functions that pression of UCP1 impairs fatty acid synthesis (186, 187). may be relevant for white adipose tissue function are the These results suggest that, in the absence of mechanisms to anaplerotic generation of metabolic intermediates for ensure continuously elevated fuel oxidation, such as those fatty acid synthesis and esterification (159), the mainte- present in brown fat, uncoupling of white adipose tissue nance of a robust pathway for the folding and secretion of mitochondria may decrease ATP levels and impair ana- high abundance circulating proteins such as adiponectin bolic flux (183). (160), and interactions between mitochondrial function In addition to effects on fuel utilization, decreased mi- and components of the insulin signaling pathway (161). tochondrial capacity in adipocytes may also alter adipo- cyte insulin sensitivity and/or function due to the high 2. Potential mechanisms by which impaired adipose tissue energetic requirements for fatty acid storage, adipokine mitochondrial oxidative capacity could result in secretion (160), insulin signaling (161), and glucose up- insulin resistance take. Interestingly, in cultured adipocytes, impairment of The large capacity of brown adipose tissue mitochon- respiratory chain function through depletion of Tfam dur- dria to oxidize fatty acids results in a measurable impact ing adipocyte differentiation results in impaired insulin- on whole-body metabolism; increased brown adipose tis- stimulated glucose transport (161); data in animal models sue abundance correlates negatively with fuel storage and are necessary to determine the physiological relevance of weight gain in rodents, and vice versa (162). The role of this finding. brown adipose tissue in human metabolism has typically been thought to be minor. However, recent work has led 3. Evidence for reduced adipose tissue mitochondrial to reconsideration of this notion, noting that humans pos- capacity in DM sess adipose tissue depots that are cold-sensitive and hy- White adipocyte mitochondrial content is decreased in permetabolic, as assessed by their very high uptake of both rodent and human obesity (177, 188–191) and cor- labeled glucose (152, 163). Such depots appear to be less relates with insulin resistance that accompanies obesity. In active as a function of aging and/or obesity (151, 164– humans, white adipocyte mtDNA copy number is in- 167). Thus, impaired mitochondrial capacity in brown versely correlated with age and BMI and directly corre- adipose tissue might be functionally linked to impaired lated with basal and insulin-induced lipogenesis (192). thermogenesis and energy expenditure, and thus increased Thus, reduced mtDNA content could reduce adipocyte susceptibility to obesity-linked insulin resistance. capacity for lipid storage, promoting ectopic lipid accu- The relevance of white adipocyte mitochondria to mulation in peripheral tissues such as muscle and liver. In whole-body metabolism and metabolic disease may de- parallel, expression of nuclear-encoded OXPHOS genes is pend on the extent to which mitochondrial respiratory down-regulated in visceral adipose tissue of humans with capacity and/or the total mass of white adipose tissue type 2 DM (193). Administration of thiazolidinediones would be sufficient to impact circulating free fatty acid induces changes in mitochondrial content and remodeling levels. White adipocytes display a high degree of plasticity in white adipocytes concomitantly with an improvement 374 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395 in insulin sensitivity (170, 173, 177, 190, 194–198). Mi- mitochondrial density are not known but could be medi- tochondrial levels in white adipocytes are also increased in ated by decreased expression of PGC-1␣, as observed in response to adrenergic stimulation, ␤-3 agonists, and CB1 obese humans (206). blockade in mice (195, 199, 200), again in parallel with enhanced insulin sensitivity. C. Liver Whether changes in mitochondrial density are a cause The liver plays a central, unique role in carbohydrate, or consequence of changes in insulin sensitivity is unclear. protein, and fat metabolism. It is critical for maintaining However, some evidence suggests that lack of insulin sig- glucose homeostasis (1) during fuel availability, via stor- naling does not reduce mitochondrial capacity in adipose age of glucose as glycogen or conversion to lipid for export tissue. For example, mice with adipose tissue-specific ab- and storage in adipose tissue, and (2) in the fasting state, lation of the insulin receptor (FIRKO mice) display high via catabolism of glycogen, synthesis of glucose from Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 levels of mitochondrial genes involved in fatty acid oxi- noncarbohydrate sources such as amino acids (gluconeo- dation and OXPHOS over the lifespan of the animals genesis), and ketogenesis. In turn, these responses are reg- (201). Thus, mechanisms that induce and maintain active ulated by the key hormones insulin and glucagon, which mitochondria in adipocytes can bypass defects in insulin modulate signaling pathways and gene expression, lead- signaling, and indeed, insulin signaling may repress mito- ing to inhibition or stimulation of glucose production, chondrial gene expression and/or function. respectively. 4. Factors affecting mitochondrial OXPHOS expression and Recent human data have highlighted the importance of function in adipose tissue disordered hepatic metabolism, including inappropriately The genetic program leading to brown adipose tissue increased hepatic glucose production, hyperlipidemia, development, and potentially to the high abundance of and lipid accumulation, in both obesity and type 2 DM mitochondria, is initiated by the zinc-finger protein (207). Similarly, rodent data also support an important PRDM16 (202–204). Current reports support the hypoth- role for the liver in diabetes pathogenesis. For example, esis that brown adipocytes and myocytes share a common liver-specific insulin receptor knockout (LIRKO) mice de- cellular lineage, potentially explaining their similarity velop insulin resistance, glucose intolerance, impaired in- with regard to containing mitochondria specialized in fuel sulin suppression of hepatic glucose production, and oxidation. In addition, the transcriptional coactivators altered patterns of hepatic gene expression (208). Inter- PGC-1␣ and -1␤ (56) play a critical role in the expansion estingly, these mice are also dyslipidemic and susceptible of the mitochondrial reticulum and in the induction of to atherosclerosis (209). UCP1 and the brown adipose tissue thermogenic program during the perinatal period (59). 1. Role of mitochondria in liver Adipocyte mitochondrial density and OXPHOS activ- Given the diverse array of unique metabolic functions ity can be regulated in response to factors that affect lipid centered in the liver, it is not surprising that ultrastructure metabolism. For example, Toh et al. (176) and Nishino, et and function of hepatic mitochondria are distinct from al. (205) find that mice deficient in Fsp27, a lipid droplet that of muscle. Electron microscopy demonstrates that protein that promotes lipid storage in white and brown mitochondrial area is 44% lower in liver than in heart adipocytes, have increased whole-body energy expendi- (210) with smaller size, fewer cristae, and lower matrix ture, resistance to diet-induced obesity, and enhanced in- density. Protein expression of multiple OXPHOS compo- sulin sensitivity. This apparent paradoxical result (high nents and Tfam (expressed per milligram of protein) and insulin sensitivity despite deficiency in lipid storage), ap- citrate synthase activity are also lower in liver (e.g., 7% pears to be due to the increased mitochondrial density and that of cardiac muscle) (211). Similarly, patterns of gene activity in white adipocytes, which are brown-like in their expression are distinct in liver (32). Functionally, isolated increased capacity to oxidize large quantities of fatty ac- hepatic mitochondria have relative reductions in ids. Nitric oxide production by the endothelial nitric oxide OXPHOS proteins, respiratory chain cytochromes, and synthase has also been linked to enhanced adipose tissue maximal activity of complexes III and IV (211). Despite mitochondrial biogenesis and prevention of high-fat diet- lower OXPHOS capacity, state 3 respiration and respira- induced obesity (200). Conversely, both genetic and diet- tory control ratio are equivalent in liver and muscle, in- induced obesity result in decreased mitochondrial density dicating differences in relative substrate concentrations and OXPHOS activity in adipose tissue (127, 177, and lower “excess capacity” in liver. Recent application of 189–191), potentially contributing to adipose tissue dys- 31P NMR to the liver in humans demonstrates that rates of function and exacerbation of insulin resistance. The mech- ATP synthesis are 3-fold higher in liver than in muscle anisms whereby obesity results in a reduction in adipose (212). By contrast, the content of mtDNA, expressed ei- Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 375 ther per gram of tissue or per mitochondrion, is actually hepatic insulin resistance. Transgenic mice expressing li- higher in liver than in other tissues. Together, these data poprotein lipase in the liver have a 2-fold increase in he- again emphasize differences in protocols assessing mi- patic triglyceride content and are insulin resistant (219). tochondrial abundance, capacity, and function and At a cellular level, incubation of hepatocytes with satu- highlight tissue diversity of mitochondrial structure and rated long-chain fatty acids induces insulin resistance by function, which may contribute to tissue-specific disease reducing insulin-stimulated tyrosine phosphorylation of susceptibility. the insulin receptor and its downstream substrates (220, 221). These effects in the liver appear to be mediated via 2. Potential mechanisms by which impaired hepatic reduced expression of the insulin receptor (221). Although mitochondrial function could influence hepatic insulin these effects could be mediated by accumulation of fatty sensitivity acyl CoA, diacylglycerols, and ceramides (as in muscle; Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 Impairments in mitochondrial number and/or oxida- Section III.A), it is intriguing that effects of fatty acids in tive function could potentially affect multiple cellular liver cells can be prevented by inhibition of CPT1, indi- functions within hepatocytes, both directly (e.g., reduced cating a critical role for mitochondrial oxidation in induc- ATP generation, alterations in oxidative stress, reduced ing lipid-mediated insulin resistance, perhaps via products capacity for fatty acid oxidation) and indirectly, via effects of incomplete oxidation and/or generation of ROS (220). on energy-requiring processes, including gluconeogenesis, Fatty acids can also alter expression and/or function of key synthesis of urea, bile acids, cholesterol, and proteins, and regulatory transcription factors in the liver (e.g., PGC-1␤, detoxification. Because accumulation of lipid within PPAR␣, hepatic nuclear factor 4␣) (127, 222–224) or hepatocytes is a key marker of insulin resistance in humans posttranscriptional regulation of mRNA stability (225). (207) and a major contributor to nonalcoholic fatty liver Fatty acid-induced reductions in insulin receptor number disease, nonalcoholic steatohepatitis (NASH), and cirrho- and function in the liver (211) may also reduce hepatic sis, we will first consider relationships between hepatic insulin clearance (226), causing systemic hyperinsulin- lipid metabolism and insulin resistance, and in Section emia, itself a contributor to both insulin resistance and III.C.3 will review evidence linking DM and hepatic ste- reduced mitochondrial function (214, 227, 228). atosis to alterations in fatty acid metabolism or more A second possibility is that hepatic insulin resistance global mitochondrial dysfunction. itself contributes to alterations in mitochondrial oxidative Hepatic lipid accumulation may result when adipose capacity. Indeed, a recent paper demonstrated that mice lipid storage capacity is exceeded, as in obesity or adi- with hepatic insulin resistance due to deletions of the pocyte dysfunction (e.g., lipodystrophy) (213). Alter- major insulin receptor substrates (IRS-1 and IRS-2) have natively, lipid accumulation may reflect an additional impaired mitochondrial function and biogenesis, as dem- imbalance between de novo hepatic lipogenesis and mi- onstrated by reduced NADH oxidation, reduced ATP tochondrial oxidative metabolism. Although the relative production rates, reduced numbers of mitochondria per roles of each of these possibilities is incompletely understood, cell, reduced fatty acid oxidation, and increased hepatic hepatic lipid accumulation is associated with obesity in hu- triglyceride accumulation (229). Mitochondrial dysfunc- mans, particularly central (abdominal) in location (214, tion was reversed by deletion of Foxo1. These data indi- 215), and in parallel with low adiponectin levels (216). cate that normal insulin signaling, which inhibits Foxo1, Interestingly, hepatic lipid accumulation is also a robust is required for maintenance of normal mitochondrial func- predictor of not only hepatic, but also muscle and adipose tion in this model. It remains unclear whether additional insulin sensitivity [better than intraabdominal fat, body components of the in vivo environment, such as glucose mass index (BMI), or other obesity measures] (217, 218). intolerance and hyperinsulinemia, contribute to mito- Conversely, modest weight loss (about 8 kg) normalizes chondrial dysfunction in these mice. However, more intrahepatic lipid in subjects with type 2 DM, in parallel broadly, these data indicate that hepatic insulin resistance with normalization of hepatic insulin sensitivity, even in can cause mitochondrial dysfunction, at least in mice. the absence of changes in intramyocellular lipid accumu- lation or circulating adipocytokines (215). 3. Evidence for impaired liver mitochondrial function Although these data highlight an intimate relationship in diabetes and NASH between obesity, intrahepatic lipid metabolism, and insu- Although human liver studies have been limited due to lin sensitivity in humans, mechanisms responsible for lack of tissue biopsy samples from otherwise healthy in- these links remain unclear. One possibility is that excessive dividuals, two groups have examined hepatic gene expres- hepatic lipid accumulation may play a central, pathogenic sion related to mitochondrial function in both obesity and role in insulin resistance. Support for this hypothesis type 2 DM (230–232). In the first (232), severe obesity comes from experimental lipid loading, which can induce (mean BMI 52 kg/m2) was associated with reduced ex- 376 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395 pression of seven of 25 genes encoding OXPHOS genes; of ␤-oxidation genes (230, 236). Similarly, circulating expression of these genes was inversely correlated with ␤-hydroxybutyrate levels are increased in NASH (235). hepatic lipid accumulation and paralleled by reduced ex- Together, these data suggest excessive, but incomplete, pression of PGC-1␣ and genes known to be regulated by fatty acid oxidation, potentially limited by reduced thyroid hormone. Similar patterns were observed in obese availability of NADϩ and FAD. Byproducts of incom- subjects with established type 2 DM. Interestingly, re- plete fatty acid oxidation could act in concert with ad- duced expression of OXPHOS genes (e.g., COX7C, ipose tissue-derived inflammatory signals (e.g., TNF␣), ATP5C1) was also observed in mice fed a high-fat diet and and altered expression and activation of proinflamma- normalized by acute therapy with thyroid hormone T3— tory (e.g., IL-1R family) and profibrotic genes (e.g., suggesting that functional hepatic thyroid hormone resis- TGFB1, FGFR2), to increase production of ROS and ultimately contribute to the development of NASH and tance could contribute to reduced expression of mitochon- Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 drial oxidative genes in this context (232). cirrhosis (235). In contrast, studies in Japanese individuals with estab- In summary, available data indicate that hepatic lipid lished DM and modest obesity (BMI 27 kg/m2) observed accumulation and insulin resistance are intimately linked a modestly increased expression of multiple genes within with mitochondrial oxidative dysfunction. We hypothe- all complexes of OXPHOS complexes, in parallel with size that modest obesity may be associated with compen- BMI and insulin resistance (measured by homeostasis satory up-regulation of OXPHOS gene expression in model assessment of insulin resistance, HOMA-IR) (231). response to sustained lipid load and/or functional defects Up-regulation of these OXPHOS genes was also positively in complete fatty acid oxidation. Up-regulation of ␤ associated with expression of several genes linked to mi- PGC-1 in this context may contribute to increased glu- tochondrial biogenesis (e.g., PGC-1␤, ERR␣, NRF, thy- coneogenesis and hyperlipidemia, in part via coactivation roid hormone receptor) and both ROS generation (e.g., of sterol regulatory element binding transcription factor 1, NADPH oxidase) and attenuation (e.g., glutathione per- as observed in high-fat diet-fed mice (223). With aging, oxidase). Thus, increased ROS related to increased fatty chronic ROS exposure, and/or the development of insulin acid oxidation and/or hyperglycemia might contribute to resistance related to obesity or sustained lipid accumula- up-regulation of OXPHOS gene expression in coexisting tion, OXPHOS expression may fall. Although this may be obesity and type 2 DM. Although these two data sets ap- an appropriate response, limiting oxidative stress, it may pear to be discordant (i.e., obesity-linked down-regulation also contribute to a vicious cycle of further impairments in of mitochondrial oxidative gene expression in the first, oxidative capacity, increased lipid accumulation, and and up-regulation in the second), several differences in the progressive insulin resistance. To test this hypothesis, lon- study population may account for these findings: 1) much gitudinal measurements of gene expression, oxidative greater degree of adiposity and hepatic steatosis in the function, and lipid accumulation in humans with progres- sive obesity and evolution of insulin resistance would be first; 2) differences in ethnicity (Caucasian-Americans vs. required—but are unlikely to be performed due to the in- Japanese); and 3) differences in insulin sensitivity and gly- vasive nature of serial liver biopsies in humans. cemia (insulin sensitive vs. resistant comparison in the first study, coexisting DM in the second). Studies of individuals with NASH provide additional D. Pancreatic ␤-cells opportunities to identify potential interactions between 1. Roles of mitochondria in ␤-cells hepatic lipid accumulation, insulin resistance, and mito- Mitochondrial capacity is central to the key function of chondrial function in humans. Indeed, enzymatic activity the pancreatic ␤-cell—regulated insulin secretion. Both of complexes I-V is reduced in liver extracts from patients rapid (first phase) and more prolonged (second phase) with NASH and is inversely correlated with BMI and insulin secretion (237) are dependent on glucose metab- HOMA-IR (233, 234). Moreover, NASH is characterized olism and mitochondrial oxidative capacity; glucose ox- by prominent abnormalities in mitochondrial ultrastruc- idation increases the ATP/ADP ratio, inhibiting plasma ture, with increased size, loss of cristae, and paracrystal- membrane K-ATP channels and allowing voltage-gated line inclusion bodies similar to those observed in some calcium channels to open. Increased cytoplasmic calcium mitochondrial myopathies (235). Although these data then triggers exocytosis of plasma-membrane docked in- cannot address whether such changes are indeed patho- sulin granules (first phase). Subsequent recruitment of genic, it is interesting that reduced OXPHOS activity in granules to the plasma membrane (second phase) appears this setting is accompanied by increased tissue long-chain to depend on mitochondrial metabolites produced by acylcarnitines and reduced short-chain acylcarnitines, de- anaplerosis (238). Mitochondrial metabolism is also re- spite normal CPT1 activity and increased expression quired for the transient, controlled production of ROS, Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 377 which is required for the mitochondrial signaling path- produced by the activity of the ETC. Low levels of ROS are ways that trigger granule exocytosis (239, 240). necessary for insulin secretion, but chronic, high mito- chondrial ROS production can have a deleterious effect on ␤ 2. Evidence for reduced ␤-cell mitochondrial capacity in DM -cell function (254–256). Thus, the activation of UCP2 ␤ Given the crucial role of mitochondrial ATP genera- protects the -cell from the deleterious effects of excess tion, anaplerosis, and ROS production in insulin secre- ROS (257) by dissipating the proton gradient and decreas- tion, mitochondrial dysfunction in ␤-cells would be ing ROS production in a controlled negative feedback expected to reduce insulin secretion and thus promote the manner (Fig. 4). However, it also leads to decreased ATP development of DM. Consistent with this possibility, production, which impairs insulin secretion. Thus, UCPs ␤-cell specific deletion of Tfam reduces insulin secretory must uncouple respiration sufficiently to mitigate toxic levels of ROS, but not enough to decrease ATP and ROS capacity and ␤-cell mass, yielding so-called mitochondrial Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 DM (241). Moreover, Tfam has recently been shown to be below the levels necessary for insulin secretion. This del- ␤ directly downstream of PDX1, a key transcription factor icate balance in which UCP2 is desirable for -cell pro- for ␤-cell development (242). tection, but undesirable for glycemic control, probably In humans, the key role of ␤-cell mitochondria is ex- underlies the discrepancy in results between two reports emplified by the development of diabetes in families on the phenotype of UCP2 knockout mice. In a mixed harboring mutations in mtDNA. Of these, the best stud- background, UCP2 knockout improves glycemic control ied is the 3243AϾG in the mtDNA-encoded in ob/ob mice (258), whereas in a pure C57BL6/J back- ␤ tRNALeu, UUR gene, which is associated with maternally ground, UCP2 knockout accelerates -cell failure and di- inherited diabetes and deafness (MIDD) (243, 244). An- abetes (259). other example is mutation 14577 TϾC, a missense sub- The levels and activity of UCP2 and the rate of ROS stitution in the NADH dehydrogenase 6 gene (245). In this production are both increased by high-fat diet and hy- case, mitochondrial respiratory chain complex I activity perphagia, possibly through the actions of nonesterified fatty acids and their ceramide derivatives (260). It is likely and O2 consumption rates are decreased by 65 and 62%, respectively, in hybrid cell lines derived from probands. that decreased ATP production due to unbalanced acti- Interestingly, mitochondrial diabetes only develops vation of UCPs by direct actions of fatty acids and their upon aging, with an average age of onset between 35 and derivatives, in addition to excessive ROS, could underlie ␤ 40 yr for MIDD and 48 yr for 14577 TϾC. This contrasts the accumulation of -cell damage that precedes type 2 with the early childhood onset of diabetes in syndromes DM (Fig. 4). such as maturity-onset diabetes of the young 2 (MODY2), in which a mutation in , the first step of gly- colysis, results in attenuated glucose-stimulated ATP gen- eration and insulin secretion. These data suggest that mitochondrial diabetes is more likely to result from a gradual deterioration of ␤-cell function, rather than from an acute functional impairment due to insufficient ATP production (246). One of the mechanisms by which mtDNA mutations might lead to a gradual deterioration in ␤-cell function, and not to an acute failure of insulin secretion due to de- creased ATP levels, could be the stress imposed by an in- crease in metabolic flux to compensate for inefficiencies in FIG. 4. Hypothesized mechanism by which free fatty acid (FFA) excess the ETC. Consistent with this view, clonal cytosolic hybrid impairs insulin secretion. A, As described above, the activity of the ETC cells harboring mitochondria derived from MIDD pa- leads to the synthesis of ATP and the generation of a small amount of ␤ tients exhibit impaired calcium handling and elevated ROS. In the -cell, both ATP and ROS are signals that trigger insulin secretion. Excessive accumulation of ROS is mitigated normally by the ROS under metabolic stress (247, 248). Chronically in- activation of UCP2, which dissipates the proton gradient, decreasing creased ROS production could also induce ␤-cell death both ATP and ROS production. The presence of this normal negative and result in gradual onset of diabetes (249–253). feedback loop suggests that the control of excessive ROS generation is imperative in the ␤-cell, even if it occurs at the expense of decreasing ATP synthesis. B, In the presence of excess FFA, this normal feedback 3. Factors affecting mitochondrial function in ␤-cells loop is compromised by a direct activation of UCP2 by FFA, as well as ␤ an effect of FFA to increase the amount of UCP2. Thus, uncoupling Mitochondrial function in -cells is highly regulated by occurs to an excessive degree, compromising ATP synthesis enough to the levels and activities of UCPs, in turn regulated by ROS impair insulin secretion and ␤-cell fitness. 378 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395

Although the tissues reviewed above are considered and inflammatory signaling. However, these same ani- central to the pathophysiology of DM, other tissues such mals displayed increased liver gluconeogenic lev- as gut, brain, kidney, neuronal tissues, and endothelium els and impaired insulin suppression of hepatic glucose are also likely to be implicated in a primary or secondary production, thus nullifying the potentially beneficial effect manner in the pathophysiology of DM and /or its com- of modest muscle PGC-1␣ overexpression on whole-body plications. The aspects of mitochondrial function unique glucose homeostasis. to each of these tissues and the consequences of their po- Higher levels of overexpression of PGC-1␣ achieved tential dysfunction in relation to DM pathophysiology are through actin promoter-driven expression in transgenic relatively less explored areas and are thus outside the scope animals display a strikingly different phenotype (263, of this review. 264). In these animals, mitochondrial density and

OXPHOS gene expression are more than double basal Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 levels, and large increases in UCP2 gene expression are IV. Experimental Strategies to Explore the also observed. Mitochondrial energetics are impaired, Relationship between Mitochondrial Function with 60% decreases in ATP levels in muscle homogenates and DM and concomitant increases in AMP activation, probably as a compensatory response to decreased mito- Although available data demonstrate links between mito- chondrial functionality. Muscle function appears com- chondrial oxidative function and phenotypes linked to in- promised, as evidenced by decreased voluntary exercise, sulin resistance and diabetes, it remains unclear whether muscle atrophy, and decreased insulin sensitivity. these are simply associations or whether oxidative dys- Inducible overexpression of PGC-1␣ in skeletal muscle function can contribute to insulin resistance and diabetes (265) results in increased mitochondrial density and a ro- risk. To address this question, we will examine available bust increase in expression of OXPHOS genes and genes data from experimental models in which OXPHOS necessary for fatty acid oxidation. In these animals, both function has been altered. Such studies have shed light on the basic mechanisms underlying mitochondrial bio- muscle glucose uptake and glycogen deposition were in- genesis and on the consequences of disruption of normal creased. Although low-intensity exercise performance did mitochondrial homeostatic mechanisms on cell and not differ in this model, high-performance exercise was whole-body oxidative metabolism. A summary of these impaired, in parallel with failure to mobilize stored gly- studies is presented in Tables 1 and 2 and is discussed in cogen. Similarly, inducible expression in cardiac muscle Section IV.A and B. can have deleterious effects. When higher levels of PGC-1␣ overexpression are restricted to heart during A. PGC-1 ␣ and ␤ overexpression early life (266), increased neonatal mitochondrial prolif- PGC-1␣ and related coactivators are critical for the eration is observed, but it is accompanied by myofibrillar ␣ regulation of mitochondrial oxidative capacity, as dem- displacement. In adults, PGC-1 induction led to a more onstrated by the approximately 2-fold increases in modest mitochondrial proliferation, which was neverthe- mtDNA and oxygen consumption and a 50% increase less surprisingly accompanied by cardiomyopathy. Thus, in mitochondrial density in myotubes overexpressing whole-body and inducible skeletal muscle- or cardiac-spe- PGC-1␣ (56). To address whether this family plays the cific overexpression of PGC-1␣ can produce deleterious same functional role in vivo, several different models of effects on muscle structure and function. PGC-1␣ transgenic expression have been generated, each When PGC-1␣ overexpression is restricted to skeletal of which differs in tissue selectivity, levels of overexpres- muscle but overexpressed throughout development by the sion achieved, and resulting metabolic phenotype (Ta- use of the gene promoter, very large in- ble 1). The lowest level of PGC-1␣ overexpression was creases in mitochondrial mass, OXPHOS, and fatty acid achieved in rat muscle by means of electroporation (261). oxidation genes are observed (267–269). In this model, This resulted in modest up-regulation of mitochondrial ATP synthesis rate and exercise performance are in- proteins, increased palmitate oxidation, and increased in- creased, and fatty acid oxidation is also enhanced. De- sulin-stimulated glucose uptake. Similarly, transgenic spite these effects, insulin sensitivity is normal in mice expressing human PGC-1␣ driven by its own pro- PGC-1␣ transgenic mice fed a chow diet and, surpris- moter displayed a modest (30% higher than basal) in- ingly, is reduced during high-fat feeding. Insulin resis- crease in mRNA expression of several OXPHOS genes, tance was paralleled by accumulation of triglycerides fiber type switching, and enhanced muscle insulin sensi- and long-chain acyl CoA (270). tivity (262). Importantly, this modest PGC-1␣ overex- Although PGC-1␤-mediated gene expression and func- pression also was accompanied by decreased levels of ROS tion appear to overlap considerably with that of PGC-1␣, norn eiw,Jn 00 13:6–9 edrv.endojournals.org 31(3):364–395 2010, June Reviews, Endocrine

TABLE 1. Studies of PGC-1␣ transgenic expression models

First author, year Mito density OXPHOS Non-OXPHOS Insulin (Ref.) Model Mito DNA (EM) mRNA/protein mRNA/protein Mito energetics Exercise capacity sensitivity Other Benton, 2008 (261) PGC-1␣ 1 113% 180% 1 COXIV 1 muscle 1 AMP kinase electroporation protein activity Ward, 2009 (262) PCG-1␣ whole-body 130% 1 mRNA 1 muscle Hepatic insulin own promoter resistance Miura, 2003, 2006 PGC-1␣ whole-body 1 200–300% 1 number 150–200% 300% 1 in UCP2 60% 2 ATP levels in 2 voluntary 2 whole-body 1 AMP kinase (263, 264) ␣-actin promoter 1 mRNA mRNA homogenates activity Russell, 2004 (266) PGC-1␣ inducible 1 350% Myofibrillar disorganization heart cardiac failure Wende, 2007 (265) PGC-1␣ inducible 1 150–250% 150% 1 FAO gene No change in low intensity, 1 glucose uptake, skeletal muscle 1 mRNA mRNA 2 performance at high glycogen intensity deposition, decreased glycolysis Lin, 2002 (268); PGC-1␣ MCK 1 166–250% 1 250% in EDL 170–300% 200–400% 1 in 50–60% 1 ATP 1 exercise performance; 2 muscle and No change in AMP Sandri, 2006 promoter 1 mRNA FAO gene mRNA synthesis by NMR 2 fatigue in vitro, whole-body kinase activity (269); Calvo, protection from only in high- 2008 (267); denervation-induced fat diet Choi, 2008 (270) atrophy Arany, 2007 (271) PGC-1␤ MCK 1 200–500% 200–500% 1 FAO 120–130% 1 endurance promoter 1 mRNA and gene mRNA protein Kamei, 2003 (272) PGC1-␤ whole-body 1 whole-body ␤-actin promoter

Mito, Mitochondrial; EM, electron microscopy; MCK, muscle creatine kinase; 2, decrease; 1, increase; EDL, extensor digitorum longus; FAO, fatty acid oxidation.

379 Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 September 23 on guest by https://academic.oup.com/edrv/article/31/3/364/2354793 from Downloaded 380 at n ovr iohnraadTp MEdcieRves ue21,31(3):364–395 2010, June Reviews, Endocrine DM 2 Type and Mitochondria Corvera and Patti

TABLE 2. Studies of PGC-1␣ or PGC-1␤ knockout models

First author, Mito Mito density OXPHOS mRNA/ Non-OXPHOS Insulin year (Ref.) Model DNA (EM) protein mRNA/protein Mito energetics Exercise capacity sensitivity Other Lin, 2004 (69); PGC-1␣ whole-body KO Normal 30–60% 2 mRNA; ATP levels 2 20% in 10–50% 2 cardiac 1 whole-body 2 body weight, Arany, 50% 2 CytC heart contractile performance increased 2005 (68) protein AMP kinase activity Leone, 2005 PGC-1␣ whole-body KO 30% 2 40–60% 2 mRNA 10% 2 in state 3 50% 2 fatigue resistance, 1 whole-body Hepatic steatosis (67) respiration abnormal cardiac response to stress Lehman, 2008 PGC-1␣ whole-body KO 2 cristae density Slight 2 mRNA 60% 2 in metabolic 2 cardiac power (61) in heart efficiency Handschin, PGC-1␣ muscle-specific Normal 30–40% 2 mRNA 50% 2 ALAS1 60% 2 grip strength, 1 muscle 2 food 2007 KO endurance, muscle consumption, (63, 64) damage basal and 2 body exercise weight, muscle inflammation, ␤-cell dysfunction Lai, 2008 (62) PGC-1␤ whole-body KO Not changed Modest 2 in state 3 2 running duration (heart) respiration Lelliot, 2006 PGC-1␤ whole-body KO 2 20% 20–40% 2 mRNA 2 state 3 and 4 2 chronotropic response Normal 2 body weight, (66) respiration, to dobutamine hepatic 2 ATP synthesis steatosis on high-fat diet Vianna, 2006 PGC-1␤ whole-body 2 30% 20–30% 2 mRNA Normal 1 hepatic lipid (65) hypomorph levels Sonoda, 2007 PGC-1␤ whole-body Normal 30–40% 2 mRNA Normal Normal Hepatic steatosis (59) hypomorph on high-fat diet Lai, 2008 (62) PGC-1␣ and PGC-1␤ 2 60%, heart Perinatal lethality due to Perinatal muscle-specific KO cardiac failure lethality due to cardiac failure

2, Decrease; 1, increase; KO, knockout; CytC, cytochrome C; ALAS1, 5-aminolevulinate synthase 1. Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 September 23 on guest by https://academic.oup.com/edrv/article/31/3/364/2354793 from Downloaded Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 381

PGC-1␤ has distinct expression profiles in skeletal muscle, mice have been generated (61, 67–69). These mice con- being expressed in parallel with myosin IIx rather than sistently display 30–60% reductions in OXPHOS gene type I fibers (271). In addition, PGC-1␣ and PGC-1␤ co- expression in muscle, normal to decreased mitochondrial activate different nuclear receptors (272). Muscle-specific density in skeletal muscle, and decreased cristae density in transgenic overexpression of PGC-1␤ results in increased heart. Functionally, decreases in ATP levels, state 3 res- oxidative fiber content and increased expression of piration, and metabolic efficiency are seen, and cardiac OXPHOS and multiple other mitochondrial genes. and skeletal muscle performance is reduced in response to Mice displayed enhanced endurance and oxidative stress. Many of these phenotypes are recapitulated in a work (271). Consistent with these findings, whole-body muscle-specific PGC-1␣ knockout line (63, 64) (Table 2). ␤ transgenic overexpression of PGC-1 resulted in in- In addition, both lines of whole-body PGC-1␣ knockout creased oxidative metabolism, protection from obesity animals display cold sensitivity, likely related to impaired Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 induced by high-fat diet or hyperphagia, and enhanced mitochondrial function in brown adipose tissue (67, 69) insulin sensitivity (272). (Table 2). Taken together, these studies suggest that modest Interestingly, muscle is significantly more insulin sen- overexpression of PGC-1␣ (261, 262) and transgenic sitive in both whole-body and muscle-specific PGC-1␣ overexpression of PGC-1␤ (271, 272) increase oxidative null mice. This apparently paradoxical finding—where metabolism and insulin sensitivity. By contrast, high- decreased oxidative function is not paralleled by insulin level overexpression of PGC-1␣ may actually reduce resistance—may be explained by the leanness and hyper- insulin sensitivity via reduced glycolysis and/or glucose activity noted in one of the whole-body knockout lines oxidation (265) or myofibrillar disruption/myopathy, po- (68, 69). In the muscle-specific knockout lines, reduction tentially limiting exercise tolerance. Moreover, high-level expression of PGC-1␣ can induce not only increases in in food intake, body weight, and adiposity (despite de- lipid oxidation genes but also parallel increases in expres- creased physical activity) may also contribute to insulin ␣ sion of genes promoting lipid uptake and synthesis (270). sensitivity (63, 64). Thus, lack of PGC-1 per se does not The net balance of these effects, and thus the net accumu- lead to insulin resistance, despite clear alterations in mus- lation of pathogenic lipid species, is likely to be determined cle and metabolic phenotypes. It is possible that the com- by the metabolic and hormonal milieu. For example, in pensatory mechanisms elicited by the complete lack of cultured myotubes overexpressing PGC-1␣ (273), lipid PGC-1␣, which result in a leaner phenotype, have a par- content is decreased in the serum-starved condition, re- adoxical insulin-sensitizing effect. For example, decreased flecting increased oxidation of lipids; by contrast, in the metabolic efficiency (61), defined as decreased ATP pro- serum-replete state, intracellular fatty acid levels are in- duced per unit of oxidized fuel and/or oxygen, may lead to creased, likely due to both increased de novo fatty acid enhanced whole-body fuel utilization and thus a leaner, synthesis from glucose and fatty acid synthesis by chain more insulin-sensitive phenotype. Activation of AMP ki- elongation (273). Thus, high-level expression of PGC-1 nase in this setting of reduced ATP production may also may actually cause accumulation of lipids and/or incom- contribute to enhanced insulin sensitivity (274). plete oxidation products, leading to reduced insulin Despite pronounced muscle and brown adipose tissue sensitivity, as observed in PGC-1␣ transgenic mice (267– functional impairments seen in models of PGC-1␣ defi- 269). These results also suggest that inappropriate, non- ciency, mitochondrial density in muscle and brown adi- physiological regulation of PGC-1␣ expression may pre- pose tissue is not notably decreased, indicating the vent appropriate modulation of oxidative metabolism possible compensatory role of PGC-1␤ in mitochondrial with feeding/fasting, or exercise/recovery—the hallmarks biogenesis. PGC-1␤-null mice (59, 62, 65, 66) display phe- of impaired metabolic flexibility observed in human insu- notypes similar to, but not identical to, those of PGC-1␣ lin resistance. knockouts, including reduced expression of OXPHOS B. PGC-1 knockout models genes, reduced ATP synthesis, reduced exercise capacity, Because enhanced insulin sensitivity is seen in some and impaired thermogenesis (59, 62). Insulin sensitivity in models of PGC-1 transgenic overexpression and human these animals is reported to be normal. PGC-1␤-null mice diabetes is associated with reduced PGC-1␣ and PGC-1␤ have more striking hepatic phenotypes, with hepatic in- expression, an important question is whether PGC-1␣ or sulin resistance, even on normal chow, and steatosis with PBC-1␤ deficiencies per se could result in insulin resis- high-fat feeding (59, 65, 66). These data raise the possi- tance. The results from studies of PGC-1␣ or PGC-1␤ bility that reduced expression of PGC1 family genes and knockout models shed some light on this question (Table impaired oxidative function may play a more important, 2). Two independent lines of whole-body PGC-1␣-null and potentially pathogenic role in liver than in muscle. 382 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395

The existence of compensation for PGC-1␤ deficiency may enhance insulin action, potentially via ATP deficiency by PGC-1␣ and vice versa is evidenced by the phenotype and/or secondary activation of AMP kinase (280). of double PGC-1␣ and PGC-1␤ muscle-specific knockout To address whether reductions in OXPHOS content or mice (62). These animals display normal mitochondrial function may contribute to diabetes-related metabolic biogenesis during development but die early after birth phenotypes, we can also consider phenotypes related to due to failure of perinatal mitochondrial proliferation and genes regulating transcription of mtDNA. The nuclear- consequent cardiac failure (62). encoded mitochondrial transcription factor Tfam is Together, data from knockout mice have provided im- essential for transcription of mtDNA and thus mitochon- portant insights into the complex and multifaceted roles of drial development, as indicated by the embryonic lethality PGC-1␣ and PGC-1␤ as mediators of basal and adaptive of Tfam-null mice (281). Similarly, skeletal and cardiac energy homeostasis. PGC-1␣ and PGC-1␤ are both re- muscle-specific deletion of Tfam causes severe OXPHOS Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 quired for the full complement of OXPHOS gene expres- deficits, dilated cardiomyopathy, and death during early sion, the establishment of correct fuel partitioning, and postnatal life (282). Skeletal muscle-specific Tfam defi- optimal metabolic efficiency because whole-body and ciency is accompanied by increased AMP kinase activation tissue-specific knockout of either of these genes leads to and increased glycolysis, indicating that compensatory deranged multiorgan metabolic phenotypes that are more mechanisms to maintain ATP levels are activated in re- evident during energetic stress. In light of these pheno- sponse to impaired mitochondrial energetics (282). In types, it is interesting to consider whether the 40–50% these mice, insulin sensitivity is normal and glucose tol- reduction in both PGC-1␣ and PGC-1␤ expression ob- erance is enhanced, consistent with a need for enhanced served in humans with obesity and type 2 DM in both liver fuel utilization in response to inefficient mitochondrial (232) and muscle (122), potentially mediated by lipid ex- ATP production. Thus, in these experimental models in cess, may contribute to further impairment in oxidative which mitochondrial oxidative capacity is severely re- capacity and a vicious cycle of maladaptive responses dur- stricted, insulin action is enhanced. ing energetic stress. DNA ␥ is a mtDNA polymerase critical for maintenance of mtDNA. POLG-null mice die during em- C. Other mitochondrial function defects bryogenesis with severe mtDNA depletion (283), whereas PGC-1 family members interact with the family of es- knock-in mutant mice have accelerated aging potentially trogen-related nuclear receptors, or ERRs, to transacti- linked to impaired repair of mtDNA mutations (284). vate transcription of oxidative genes (275) and mitofusins Mice null for the mitochondrial Twinkle have (276). Interestingly, ERR␣-null mice have reduced body weight and are normoglycemic, in parallel with up-regu- impaired respiratory chain activity and muscle atrophy, lation of medium-chain acyl-CoA dehydrogenase and re- but no defect in exercise capacity (285). Thus, these data duced lipogenesis, both of which may contribute to resis- indicate that primary defects in mtDNA, even when pro- tance to diet-induced obesity (277). Although the duced experimentally by ablation of nuclear-encoded physiological effects of the related gene ERR␥ remain genes, do not produce overt metabolic phenotypes. These unclear, it interacts with PGC-1 to regulate expression observations are consistent with those in humans harbor- of ERR␣ (278), and polymorphisms at this locus were ing mutations in mtDNA, where mitochondrial diabetes found to be associated with increased glucose area un- typically manifests as a gradual deterioration of pancreatic ␤ der the curve in a genetic analysis of Old Order Amish -cell function (see Section III.D). subjects (279). Pospisilik et al. (274) used an alternative strategy to test whether experimentally induced respiratory chain V. Conclusions complex deficiency would induce metabolic phenotypes. Tissue-specific ablation of the nuclear-encoded gene, mi- Mitochondrial function in tissues involved in the patho- tochondrial apoptosis-inducing factor (AIF), produced genesis of DM (liver, muscle, adipose tissue, and pancre- modest defects in respiratory chain gene expression and atic ␤-cells) is critical for multiple aspects of cellular function, as expected; however, both muscle- and liver- metabolism. In each of these tissues, mitochondrial oxi- specific null mice had increased insulin sensitivity, reduced dative activity must be appropriate to fully oxidize nutri- fat mass, and improved glucose tolerance. Although de- ent loads, particularly fatty acids. Failure of complete fects in AIF may not precisely replicate respiratory chain oxidation can lead to accumulation of lipid intermediates, defects mediated by other genes, these data mirror to a incomplete fatty acid oxidation products, and ROS, in- large extent those in PGC-1-␣ null mice and again suggest ducing both insulin resistance (muscle, liver, adipose) and that modest defects in mitochondrial oxidative capacity altered secretion (␤-cells). Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 383

The sufficiency to fully oxidize fatty acids resides in the mal adaptive responses would be intolerant to moderate- balance between: 1) net mitochondrial oxidative activity, high fuel loads, leading to lipid accumulation, incomplete in turn determined by the need to generate energy to meet oxidation, production of ROS, and acute insulin resis- cellular demands, e.g., contraction and ion transport; tance (Fig. 5B). In turn, such alterations in mitochondrial and 2) fuel availability (determined by food intake, ad- activity could be mediated by genetic factors (family his- iposity, and adipose storage capacity) (Fig. 5A). Balance tory, ethnicity), epigenetic mechanisms, developmental is achieved when oxidative activity equals or exceeds exposures, and aging. With time, insufficient oxidative fuel loads. capacity could be resolved by compensatory mechanisms Under normal homeostatic conditions, both oxidative that increase oxidative capacity (e.g., exercise, mitochon- activity and cellular fuel availability could in principle be drial biogenesis) (Fig. 5C, top right) or decrease fuel load altered to ensure that mitochondrial function is appropri- (weight loss) (Fig. 5C, middle right). Insufficient compen- Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 ate for the ambient metabolic environment. For example, sation could result in chronic insulin resistance (Fig. 5C, cellular demand for energy can be increased through ex- bottom right). ercise, and fuel availability can be reduced through weight Two key questions arise from this model: 1) does in- loss and/or reduced food intake. terindividual variation in either baseline or adaptive In this context, interindividual variation in oxidative mitochondrial oxidative responses alter risk for insulin capacity and/or activity, fuel load, or ability to modulate resistance; and 2) does chronic imbalance itself resulting mitochondrial activity (acute response), increase mito- from chronic overnutrition (e.g., ROS damage to mtDNA) chondrial capacity (chronic response), or resolve oxida- impair mitochondrial capacity, leading to further meta- tive stress could determine the set point of metabolic bolic imbalance? With regard to the first question, mild balance. Such differences could become prominent par- deficiencies in mitochondrial activity, and/or an inability ticularly in an obesogenic environment. Thus, individuals to increase activity and capacity in response to cellular with a high oxidative capacity or adaptive responses energy demand, could explain the reduced exercise ability would have high tolerance to large fuel loads. Conversely, seen in individuals with a family history of DM (106). individuals with low oxidative capacity and/or subopti- Over time, this phenotype could contribute to reduced voluntary exercise and increase the likelihood of an im- balance between mitochondrial activity and fatty acid load. Secondly, chronic imbalance in energy metabolism due to overnutrition, obesity, and inactivity could directly contribute to increased cellular and mitochondrial ROS production. In turn, excessive ROS can induce both insu- lin resistance and mitochondrial dysfunction. For exam- ple, a high-fat, high-sucrose diet in the diabetes-prone C57BL6 mouse causes mitochondrial alterations in par- allel with enhanced ROS production and impaired insulin sensitivity. Similarly, exposure of muscle cells in vitro to saturated fatty acids or high-fat feeding in mice results in

FIG. 5. Dynamic relationship between oxidative activity and fuel load alterations in mitochondrial structure and insulin resis- leading to development of disease. In this diagram, oxidative capacity tance, both of which are reversed by antioxidants refers to the ability to generate energy in response to varying energy (286–288). Thus, oxidative stress can induce mitochon- requirements, and balance is indicated by oxidative capacity equaling or exceeding fuel loads. A, High oxidative activity can ensue from high- drial dysfunction in parallel with insulin resistance—per- energy requirements (chronic exercise, high metabolic rate). Individuals haps an adaptive response aimed at limiting further oxi- with a high oxidative capacity will have high tolerance to large fuel dative damage. More importantly, resolution of oxidative loads. B, Low oxidative activity can ensue from a lack of energy stress can reverse insulin resistance. These data also sug- requirement (sedentary lifestyle) or inability to generate energy (mitochondrial myopathies, intrauterine exposures, genetics) as seen in gest that defects in resolution of oxidative stress may be the failure of ATP synthesis in DM patients in response to exercise. another mechanism conferring increased risk for both mi- Individuals with low oxidative capacity are intolerant to moderate-high tochondrial dysfunction and insulin resistance. fuel loads that lead to ROS generation, lipid accumulation, incomplete oxidation, and acute insulin resistance. C, Insufficient oxidative The effects of these variables on disease progression can capacity can be resolved by compensatory mechanisms that increase be hypothesized to occur in three stages (Table 3). In the oxidative activity (e.g., exercise, top right) decrease fuel load (weight first stage, mitochondrial activity is adequate relative to loss, middle right), or resolve maladaptive patterns of oxidative stress. Insufficient compensation results in chronic insulin resistance (bottom fuel intake, and normal insulin sensitivity and secretion right). are observed. In a second stage, mitochondrial activity is 384 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395

TABLE 3. Stages of disease progression

Stage I: healthy Stage II: physiological stress Stage III: failure of insulin sensitive with appropriate compensation compensation Oxidative capacity Normal exercise capacity Inappropriately low relative to load, Further + and activity and activity due to: ● Inactivity ● Reduced fitness ● Genetic susceptibility ● Aging Oxidative load Normal Increased, due to: Sustained _ ● Cellular overnutrition ● Obesity Cellular response Normal ● Initial 1 mitochondrial ● + Nuclear-encoded OXPHOS Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 patterns biogenesis, inadequate to gene expression meet metabolic demand ● 1 Incomplete oxidation ● Vicious cycle of suboptimal ● 1 ROS generation oxidative capacity ● 1 Tissue lipid accumulation Physiological Normal insulin sensitivity Insulin resistance ● Progressive insulin resistance consequences and secretion ● Insulin secretory dysfunction ● Type 2 DM

2, Decrease; 1, increase. inadequate relative to oxidative load, leading to the de- are insufficient to answer this question with certainty or to velopment of oxidative stress, insulin resistance, impaired determine whether impaired oxidative capacity can cause insulin secretion, or both. Insulin resistance per se could insulin resistance, or result from insulin resistance. Animal result in further reductions in mitochondrial capacity data are inherently limited by interspecies comparisons. (229, 289), which in the absence of compensation (e.g., Moreover, it is also difficult to fully recapitulate in an decreased food intake) would lead to a third stage, char- animal model the subtle changes in mitochondrial activ- acterized by further imbalance, ␤-cell failure, and progres- ities and capacity and the interactions between mitochon- sion to DM. drial function and relative fuel load that, in humans, can We return to the key question—do variations in oxi- enhance diabetes risk. Nevertheless, results from multiple dative activity underlie the risk for and development of mouse models now indicate that absolute deficiency of the DM in humans? Unfortunately, the current human data PGC-1 family of coactivators or mitochondrial OXPHOS per se does not directly produce insulin resis- tance. Thus, we interpret the current data to indicate that an individual’s intrinsic mito- chondrial oxidative activity may determine the magnitude of chronic cellular dysfunction and influence adaptive responses to chronic fuel ex- cess, as with obesity or inactivity. Moreover, experimental induction of insulin resistance and/or components of the insulin-resistant/di- abetes milieu may contribute to reductions in mitochondrial oxidative capacity, further fuel- ing a vicious cycle of diabetes risk (Fig. 6). To fully dissect these possibilities, better an- imal models, together with prospective longi- tudinal studies in humans analyzing OXPHOS FIG. 6. Vicious cycle leading to progressively increased risk of DM. An individual’s capacity in different tissues, their variation intrinsic mitochondrial oxidative capacity is determined by numerous factors with aging, weight gain, and fuel load, and their including genetic and ethnic background, intrauterine exposures, and age. Chronic fuel excess, in the setting of suboptimal oxidative activity, results in fatty acid correlation with the development of metabolic accumulation, incomplete oxidation, increased oxidative stress, and ROS generation disease are necessary. The recent emergence of leading to impairment in oxidative capacity, compounding the deleterious effects of metabolomic approaches will likely facilitate fuel excess. With time, mitochondrial damage ensues, further exacerbating the process. Together, these factors contribute to progressively impaired insulin further assessment of mitochondrial function sensitivity, insulin secretion, and heightened risk of DM. in humans, potentially allowing use of clini- Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 385 cally accessible blood samples for such longitudinal stud- 2. Centers for Disease Control and Prevention, US Department ies (290–293). A still-unanswered but intriguing question of Health and Human Services 2007 National diabetes fact is whether altered energetics during developmentally sheet. http://www.cdc.gov/diabetes/pubs/factsheet07.htm 3. Holloszy JO 2009 Skeletal muscle “mitochondrial defi- sensitive periods (e.g., intrauterine or early postnatal ciency” does not mediate insulin resistance. Am J Clin Nutr life) could ultimately increase susceptibility to insulin 89:463S–466S resistance. 4. Boyle JP, Honeycutt AA, Narayan KM, Hoerger TJ, Geiss In summary, we hypothesize that mitochondrial oxi- LS, Chen H, Thompson TJ 2001 Projection of diabetes dative activity can be considered as a key determinant burden through 2050: impact of changing demography and disease prevalence in the U.S. Diabetes Care 24: underlying diabetes risk. In isolation, reductions in mito- 1936–1940 chondrial activity mediated by genetic factors (family his- 5. Roglic G, Unwin N, Bennett PH, Mathers C, Tuomilehto tory, ethnicity), epigenetic mechanisms, developmental J, Nag S, Connolly V, King H 2005 The burden of mortality Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 exposures, and aging may not be sufficient to induce in- attributable to diabetes: realistic estimates for the year sulin resistance. However, when sustained fuel excess 2000. Diabetes Care 28:2130–2135 (e.g., resulting from overnutrition or impaired fat storage) 6. Ogden CL, Carroll MD, Curtin LR, McDowell MA, Tabak CJ, Flegal KM 2006 Prevalence of overweight and obesity exceeds energetic demands and/or oxidative capacity, in the United States, 1999–2004. JAMA 295:1549–1555 and/or appropriate compensatory mechanisms are insuf- 7. Hu FB, Manson JE, Stampfer MJ, Colditz G, Liu S, ficient (e.g., due to inactivity or failure of mitochondria to Solomon CG, Willett WC 2001 Diet, lifestyle, and the adapt to higher cellular oxidative demands), a vicious cy- risk of type 2 diabetes mellitus in women. N Engl J Med cle of insulin resistance and impaired insulin secretion can 345:790–797 8. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, be initiated (Fig. 6). Resulting lipid accumulation and ox- Lachin JM, Walker EA, Nathan DM 2002 Reduction in the idative stress can alter transcriptional responses and dam- incidence of type 2 diabetes with lifestyle intervention or age mitochondria, further reducing OXPHOS capacity, metformin. N Engl J Med 346:393–403 compounding the deleterious effects of fuel excess and 9. Tuomilehto J, Lindstro¨ m J, Eriksson JG, Valle TT, increasing diabetes risk. Although the importance of the Ha¨ma¨la¨inen H, Ilanne-Parikka P, Keina¨nen-Kiukaanniemi PGC-1 family of coactivators has been stressed thus far, it S, Laakso M, Louheranta A, Rastas M, Salminen V, Uusitupa M 2001 Prevention of type 2 diabetes mellitus by is likely there are many additional transcriptional and met- changes in lifestyle among subjects with impaired glucose abolic mechanisms by which mitochondrial function tolerance. N Engl J Med 344:1343–1350 adapts to environmental demands. Critical studies of these 10. Barroso I, Gurnell M, Crowley VE, Agostini M, Schwabe gene-environment interactions is likely to be a fruitful area JW, Soos MA, Maslen GL, Williams TD, Lewis H, Schafer of investigation in the years to come. AJ, Chatterjee VK, O’Rahilly S 1999 Dominant negative mutations in human PPAR␥ associated with severe insulin resistance, diabetes mellitus and hypertension. Nature 402: 880–883 Acknowledgments 11. Fajans SS, Bell GI, Polonsky KS 2001 Molecular mecha- nisms and clinical pathophysiology of maturity-onset dia- Address all correspondence and requests for reprints to: Mary-Elizabeth betes of the young. N Engl J Med 345:971–980 Patti, Joslin Diabetes Center, One Joslin Place, Room 620, Boston, Mas- 12. Florez JC, Jablonski KA, Bayley N, Pollin TI, de Bakker PI, sachusetts 02215. E-mail: [email protected]; or Shuldiner AR, Knowler WC, Nathan DM, Altshuler D Silvia Corvera, Program in Molecular Medicine, 373 Plantation Street, 2006 TCF7L2 polymorphisms and progression to diabetes Suite 107, Worcester, Massachusetts 01605. E-mail: silvia.corvera@ in the Diabetes Prevention Program. N Engl J Med 355: umassmed.edu. 241–250 The authors gratefully acknowledge research support from National 13. Grant SF, Thorleifsson G, Reynisdottir I, Benediktsson R, Institutes of Health Grants DK062948 (to M.-E.P.), DK080366 (to Manolescu A, Sainz J, Helgason A, Stefansson H, Emilsson S.C.), the Graetz Foundation (to M.-E.P.), LM008748 (to M.-E.P.), V, Helgadottir A, Styrkarsdottir U, Magnusson KP, DK060837 (Diabetes Genome Anatomy Project, to M.-E.P., and S.C.), Walters GB, Palsdottir E, Jonsdottir T, Gudmundsdottir M01 RR001032 (General Clinical Research Center), DK36836 (Diabe- T, Gylfason A, Saemundsdottir J, Wilensky RL, Reilly tes and Endocrinology Research Center, Joslin Diabetes Center), and MP, Rader DJ, Bagger Y, Christiansen C, Gudnason V, DK32520 (Diabetes Endocrinology Research Center, University of Mas- Sigurdsson G, Thorsteinsdottir U, Gulcher JR, Kong A, sachusetts Medical School). Stefansson K 2006 Variant of transcription factor 7-like 2 Disclosure Summary: The authors have nothing to disclose. (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet 38:320–323 14. Horikawa Y, Oda N, Cox NJ, Li X, Orho-Melander M, Hara M, Hinokio Y, Lindner TH, Mashima H, Schwarz References PE, del Bosque-Plata L, Horikawa Y, Oda Y, Yoshiuchi I, Colilla S, Polonsky KS, Wei S, Concannon P, Iwasaki N, 1. American Diabetes Association 2008 Diabetes statistics. Schulze J, Baier LJ, Bogardus C, Groop L, Boerwinkle E, http://www.diabetes.org/diabetes-basics/diabetes-statistics/ Hanis CL, Bell GI 2000 Genetic variation in the gene en- 386 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395

coding calpain-10 is associated with type 2 diabetes mel- Kamal M, Patterson N, Lander ES, Mann M 2003 In- litus. Nat Genet 26:163–175 tegrated analysis of protein composition, tissue diver- 15. O’Rahilly S, Barroso I, Wareham NJ 2005 Genetic factors sity, and gene regulation in mouse mitochondria. Cell in type 2 diabetes: the end of the beginning? Science 307: 115:629–640 370–373 33. Starkov AA 2008 The role of mitochondria in reactive ox- 16. Owen KR, McCarthy MI 2007 Genetics of type 2 diabetes. ygen species metabolism and signaling. Ann NY Acad Sci Curr Opin Genet Dev 17:239–244 1147:37–52 17. Knowler WC, Pettitt DJ, Saad MF, Bennett PH 1990 34. Murphy MP 2009 How mitochondria produce reactive Diabetes mellitus in the Pima Indians: incidence, risk fac- oxygen species. Biochem J 417:1–13 tors and pathogenesis. Diabetes Metab Rev 6:1–27 35. Murgia M, Giorgi C, Pinton P, Rizzuto R 2009 Controlling 18. Poulsen P, Kyvik KO, Vaag A, Beck-Nielsen H 1999 Her- metabolism and cell death: at the heart of mitochondrial itability of type II (non-insulin-dependent) diabetes melli- calcium signalling. J Mol Cell Cardiol 46:781–788

tus and abnormal glucose tolerance—a population-based 36. Rimessi A, Giorgi C, Pinton P, Rizzuto R 2008 The versa- Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 twin study. Diabetologia 42:139–145 tility of mitochondrial calcium signals: from stimulation of 19. Barker DJ, Hales CN, Fall CH, Osmond C, Phipps K, Clark cell metabolism to induction of cell death. Biochim Biophys PM 1993 Type 2 (non-insulin-dependent) diabetes melli- Acta 1777:808–816 tus, hypertension and hyperlipidaemia (syndrome X): re- 37. Lill R, Mu¨ hlenhoff U 2008 Maturation of iron-sulfur pro- lation to reduced fetal growth. Diabetologia 36:62–67 teins in : mechanisms, connected processes, and 20. Dabelea D, Hanson RL, Lindsay RS, Pettitt DJ, Imperatore diseases. Annu Rev Biochem 77:669–700 G, Gabir MM, Roumain J, Bennett PH, Knowler WC 2000 38. Miller WL 2008 Steroidogenic enzymes. Endocr Dev Intrauterine exposure to diabetes conveys risks for type 2 13:1–18 diabetes and obesity: a study of discordant sibships. Dia- 39. Miller WL 2007 Steroidogenic acute regulatory protein betes 49:2208–2211 (StAR), a novel mitochondrial cholesterol transporter. Bio- 21. Rich-Edwards JW, Colditz GA, Stampfer MJ, Willett WC, chim Biophys Acta 1771:663–676 Gillman MW, Hennekens CH, Speizer FE, Manson JE 40. Miller WL 2005 Minireview: regulation of steroidogenesis 1999 Birthweight and the risk for type 2 diabetes mellitus by electron transfer. Endocrinology 146:2544–2550 in adult women. Ann Intern Med 130:278–284 41. Gonzalez IL 2005 Barth syndrome: TAZ gene mutations, 22. Despre´s JP 1993 Abdominal obesity as important compo- mRNAs, and evolution. Am J Med Genet A 134:409–414 nent of insulin-resistance syndrome. Nutrition 9:452–459 42. Muravchick S 2008 Clinical implications of mitochondrial 23. Lazar MA 2005 How obesity causes diabetes: not a tall disease. Adv Drug Deliv Rev 60:1553–1560 tale. Science 307:373–375 43. Gargus JJ 2009 Genetic calcium signaling abnormalities in 24. Manson JE, Rimm EB, Stampfer MJ, Colditz GA, Willett the central nervous system: seizures, migraine, and autism. WC, Krolewski AS, Rosner B, Hennekens CH, Speizer FE Ann NY Acad Sci 1151:133–156 1991 Physical activity and incidence of non-insulin-depen- 44. Gandhi S, Wood-Kaczmar A, Yao Z, Plun-Favreau H, dent diabetes mellitus in women. Lancet 338:774–778 Deas E, Klupsch K, Downward J, Latchman DS, Tabrizi SJ, 25. Buchanan TA, Xiang AH 2005 Gestational diabetes mel- Wood NW, Duchen MR, Abramov AY 2009 PINK1- litus. J Clin Invest 115:485–491 associated Parkinson’s disease is caused by neuronal vul- 26. Lo¨ nnroth P, Smith U 1986 Aging enhances the insulin re- nerability to calcium-induced cell death. Mol Cell 33: sistance in obesity through both receptor and postreceptor 627–638 alterations. J Clin Endocrinol Metab 62:433–437 45. Benard G, Rossignol R 2008 Ultrastructure of the mito- 27. Eriksson J, Franssila-Kallunki A, Ekstrand A, Saloranta C, chondrion and its bearing on function and bioenergetics. Wide´n E, Schalin C, Groop L 1989 Early metabolic defects Antioxid Redox Signal 10:1313–1342 in persons at increased risk for non-insulin-dependent 46. Stuart RA 2008 Supercomplex organization of the oxida- diabetes mellitus. N Engl J Med 321:337–343 tive phosphorylation enzymes in yeast mitochondria. 28. Martin BC, Warram JH, Krolewski AS, Bergman RN, J Bioenerg Biomembr 40:411–417 Soeldner JS, Kahn CR 1992 Role of glucose and insulin 47. Vonck J, Scha¨fer E 2009 Supramolecular organization of resistance in development of type 2 diabetes mellitus: re- protein complexes in the mitochondrial inner membrane. sults of a 25-year follow-up study. Lancet 340:925–929 Biochim Biophys Acta 1793:117–124 29. Matthews DR, Levy JC 2009 Impending type 2 diabetes. 48. Shaw JM, Nunnari J 2002 Mitochondrial dynamics and Lancet 373:2178–2179 division in budding yeast. Trends Cell Biol 12:178–184 30. Goldfine AB, Bouche C, Parker RA, Kim C, Kerivan A, 49. Chen H, Chan DC 2005 Emerging functions of mamma- Soeldner JS, Martin BC, Warram JH, Kahn CR 2003 In- lian mitochondrial fusion and fission. Hum Mol Genet 14 sulin resistance is a poor predictor of type 2 diabetes in Spec No. 2:R283–R289 individuals with no family history of disease. Proc Natl 50. Parone PA, Da Cruz S, Tondera D, Mattenberger Y, James Acad Sci USA 100:2724–2729 DI, Maechler P, Barja F, Martinou JC 2008 Preventing 31. Pagliarini DJ, Calvo SE, Chang B, Sheth SA, Vafai SB, Ong mitochondrial fission impairs mitochondrial function and SE, Walford GA, Sugiana C, Boneh A, Chen WK, Hill DE, leads to loss of mitochondrial DNA. PLoS ONE 3:e3257 Vidal M, Evans JG, Thorburn DR, Carr SA, Mootha VK 51. Soubannier V, McBride HM 2009 Positioning mitochon- 2008 A mitochondrial protein compendium elucidates drial plasticity within cellular signaling cascades. Biochim complex I disease biology. Cell 134:112–123 Biophys Acta 1793:154–170 32. Mootha VK, Bunkenborg J, Olsen JV, Hjerrild M, 52. Bach D, Pich S, Soriano FX, Vega N, Baumgartner B, Wisniewski JR, Stahl E, Bolouri MS, Ray HN, Sihag S, Oriola J, Daugaard JR, Lloberas J, Camps M, Zierath JR, Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 387

Rabasa-Lhoret R, Wallberg-Henriksson H, Laville M, 65. Vianna CR, Huntgeburth M, Coppari R, Choi CS, Lin J, Palacín M, Vidal H, Rivera F, Brand M, Zorzano A 2003 Krauss S, Barbatelli G, Tzameli I, Kim YB, Cinti S, Shulman Mitofusin-2 determines mitochondrial network archi- GI, Spiegelman BM, Lowell BB 2006 Hypomorphic muta- tecture and mitochondrial metabolism. A novel regula- tion of PGC-1␤ causes mitochondrial dysfunction and liver tory mechanism altered in obesity. J Biol Chem 278: insulin resistance. Cell Metab 4:453–464 17190–17197 66. Lelliott CJ, Medina-Gomez G, Petrovic N, Kis A, Feldmann 53. Scarpulla RC 1997 Nuclear control of respiratory chain HM, Bjursell M, Parker N, Curtis K, Campbell M, Hu P, expression in mammalian cells. J Bioenerg Biomembr Zhang D, Litwin SE, Zaha VG, Fountain KT, Boudina S, 29:109–119 Jimenez-Linan M, Blount M, Lopez M, Meirhaeghe A, 54. Virbasius JV, Scarpulla RC 1994 Activation of the human Bohlooly-Y M, Storlien L, Stro¨ mstedt M, Snaith M, Oresic mitochondrial transcription factor A gene by nuclear re- M, Abel ED, Cannon B, Vidal-Puig A 2006 Ablation of spiratory factors: a potential regulatory link between nu- PGC-1␤ results in defective mitochondrial activity, thermo-

clear and mitochondrial gene expression in organelle bio- genesis, hepatic function, and cardiac performance. PLoS Biol Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 genesis. Proc Natl Acad Sci USA 91:1309–1313 4:e369 55. Scarpulla RC 2008 Transcriptional paradigms in mamma- 67. Leone TC, Lehman JJ, Finck BN, Schaeffer PJ, Wende AR, lian mitochondrial biogenesis and function. Physiol Rev Boudina S, Courtois M, Wozniak DF, Sambandam N, 88:611–638 Bernal-Mizrachi C, Chen Z, Holloszy JO, Medeiros DM, 56. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Schmidt RE, Saffitz JE, Abel ED, Semenkovich CF, Kelly Mootha V, Troy A, Cinti S, Lowell B, Scarpulla RC, DP 2005 PGC-1␣ deficiency causes multi-system energy Spiegelman BM 1999 Mechanisms controlling mitochon- metabolic derangements: muscle dysfunction, abnormal drial biogenesis and respiration through the thermogenic weight control and hepatic steatosis. PLoS Biol 3:e101 coactivator PGC-1. Cell 98:115–124 68. Arany Z, He H, Lin J, Hoyer K, Handschin C, Toka O, 57. Lin J, Puigserver P, Donovan J, Tarr P, Spiegelman BM Ahmad F, Matsui T, Chin S, Wu PH, Rybkin II, Shelton JM, 2002 Peroxisome proliferator-activated receptor ␥ coacti- Manieri M, Cinti S, Schoen FJ, Bassel-Duby R, Rosenzweig vator 1␤ (PGC-1␤), a novel PGC-1-related transcription A, Ingwall JS, Spiegelman BM 2005 Transcriptional coacti- coactivator associated with host cell factor. J Biol Chem vator PGC-1 ␣ controls the energy state and contractile func- 277:1645–1648 tion of cardiac muscle. Cell Metab 1:259–271 58. Andersson U, Scarpulla RC 2001 Pgc-1-related coactiva- 69. Lin J, Wu PH, Tarr PT, Lindenberg KS, St-Pierre J, Zhang tor, a novel, serum-inducible coactivator of nuclear respi- CY, Mootha VK, Ja¨ger S, Vianna CR, Reznick RM, Cui L, ratory factor 1-dependent transcription in mammalian Manieri M, Donovan MX, Wu Z, Cooper MP, Fan MC, cells. Mol Cell Biol 21:3738–3749 Rohas LM, Zavacki AM, Cinti S, Shulman GI, Lowell BB, 59. Sonoda J, Mehl IR, Chong LW, Nofsinger RR, Evans RM Krainc D, Spiegelman BM 2004 Defects in adaptive energy 2007 PGC-1␤ controls mitochondrial metabolism to mod- metabolism with CNS-linked hyperactivity in PGC-1␣ null ulate circadian activity, adaptive thermogenesis, and he- mice. Cell 119:121–135 patic steatosis. Proc Natl Acad Sci USA 104:5223–5228 70. Vercauteren K, Gleyzer N, Scarpulla RC 2009 Short hair- 60. Leick L, Wojtaszewski JF, Johansen ST, Kiilerich K, Comes pin RNA-mediated silencing of PRC (PGC-1-related coac- G, Hellsten Y, Hidalgo J, Pilegaard H 2008 PGC-1␣ is not tivator) results in a severe respiratory chain deficiency as- mandatory for exercise- and training-induced adaptive sociated with the proliferation of aberrant mitochondria. gene responses in mouse skeletal muscle. Am J Physiol En- J Biol Chem 284:2307–2319 docrinol Metab 294:E463–E474 71. Christian M, White R, Parker MG 2006 Metabolic regu- 61. Lehman JJ, Boudina S, Banke NH, Sambandam N, Han X, lation by the nuclear receptor corepressor RIP140. Trends Young DM, Leone TC, Gross RW, Lewandowski ED, Abel Endocrinol Metab 17:243–250 ED, Kelly DP 2008 The transcriptional coactivator 72. Powelka AM, Seth A, Virbasius JV, Kiskinis E, Nicoloro PGC-1␣ is essential for maximal and efficient cardiac mi- SM, Guilherme A, Tang X, Straubhaar J, Cherniack AD, tochondrial fatty acid oxidation and lipid homeostasis. Parker MG, Czech MP 2006 Suppression of oxidative me- Am J Physiol Heart Circ Physiol 295:H185–H196 tabolism and mitochondrial biogenesis by the transcrip- 62. LaiL,LeoneTC,ZechnerC,SchaefferPJ,KellySM,Flanagan tional corepressor RIP140 in mouse adipocytes. J Clin DP, Medeiros DM, Kovacs A, Kelly DP 2008 Transcriptional Invest 116:125–136 coactivators PGC-1␣ and PGC-l␤ control overlapping pro- 73. Debevec D, Christian M, Morganstein D, Seth A, Herzog grams required for perinatal maturation of the heart. Genes B, Parker M, White R 2007 Receptor interacting protein Dev 22:1948–1961 140 regulates expression of uncoupling protein 1 in adi- 63. Handschin C, Chin S, Li P, Liu F, Maratos-Flier E, pocytes through specific peroxisome proliferator activated Lebrasseur NK, Yan Z, Spiegelman BM 2007 Skeletal receptor isoforms and estrogen-related receptor ␣. Mol muscle fiber-type switching, exercise intolerance, and Endocrinol 21:1581–1592 myopathy in PGC-1␣ muscle-specific knock-out ani- 74. Leonardsson G, Steel JH, Christian M, Pocock V, Milligan mals. J Biol Chem 282:30014–30021 S, Bell J, So PW, Medina-Gomez G, Vidal-Puig A, White R, 64. Handschin C, Choi CS, Chin S, Kim S, Kawamori D, Kurpad Parker MG 2004 Nuclear receptor corepressor RIP140 AJ, Neubauer N, Hu J, Mootha VK, Kim YB, Kulkarni RN, regulates fat accumulation. Proc Natl Acad Sci USA 101: Shulman GI, Spiegelman BM 2007 Abnormal glucose ho- 8437–8442 meostasis in skeletal muscle-specific PGC-1␣ knockout mice 75. Parker MG, Christian M, White R 2006 The nuclear re- reveals skeletal muscle-pancreatic ␤-cell crosstalk. J Clin ceptor co-repressor RIP140 controls the expression of met- Invest 117:3463–3474 abolic gene networks. Biochem Soc Trans 34:1103–1106 388 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395

76. Seth A, Steel JH, Nichol D, Pocock V, Kumaran MK, Fritah in young nonobese Caucasian first-degree relatives of pa- A, Mobberley M, Ryder TA, Rowlerson A, Scott J, Poutanen tients with non-insulin-dependent diabetes mellitus. J Clin M, White R, Parker M 2007 The transcriptional corepressor Invest 89:782–788 RIP140 regulates oxidative metabolism in skeletal muscle. 93. Pratipanawatr W, Pratipanawatr T, Cusi K, Berria R, Adams Cell Metab 6:236–245 JM, Jenkinson CP, Maezono K, DeFronzo RA, Mandarino LJ 77. Baker MJ, Frazier AE, Gulbis JM, Ryan MT 2007 Mito- 2001 Skeletal muscle insulin resistance in normoglyce- chondrial protein-import machinery: correlating structure mic subjects with a strong family history of type 2 dia- with function. Trends Cell Biol 17:456–464 betes is associated with decreased insulin-stimulated in- 78. MacKenzie JA, Payne RM 2007 Mitochondrial protein im- sulin receptor substrate-1 tyrosine phosphorylation. port and human health and disease. Biochim Biophys Acta Diabetes 50:2572–2578 1772:509–523 94. Jacob S, Machann J, Rett K, Brechtel K, Volk A, Renn W, 79. Mokranjac D, Neupert W 2008 Energetics of protein trans- Maerker E, Matthaei S, Schick F, Claussen CD, Ha¨ring HU

location into mitochondria. Biochim Biophys Acta 1777: 1999 Association of increased intramyocellular lipid con- Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 758–762 tent with insulin resistance in lean nondiabetic offspring of 80. Walther DM, Rapaport D 2009 Biogenesis of mitochon- type 2 diabetic subjects. Diabetes 48:1113–1119 drial outer membrane proteins. Biochim Biophys Acta 95. Malenfant P, Joanisse DR, The´riault R, Goodpaster BH, 1793:42–51 Kelley DE, Simoneau JA 2001 Fat content in individual 81. Hatch GM 2004 Cell biology of cardiac mitochondrial muscle fibers of lean and obese subjects. Int J Obes Relat phospholipids. Biochem Cell Biol 82:99–112 Metab Disord 25:1316–1321 82. Mileykovskaya E, Zhang M, Dowhan W 2005 Cardiolipin 96. Dresner A, Laurent D, Marcucci M, Griffin ME, Dufour S, in energy transducing membranes. Biochemistry (Mosc) Cline GW, Slezak LA, Andersen DK, Hundal RS, Rothman 70:154–158 DL, Petersen KF, Shulman GI 1999 Effects of free fatty 83. Schlame M, Rua D, Greenberg ML 2000 The biosyn- acids on glucose transport and IRS-1-associated phospha- thesis and functional role of cardiolipin. Prog Lipid Res tidylinositol 3-kinase activity. J Clin Invest 103:253–259 39:257–288 97. Savage DB, Petersen KF, Shulman GI 2007 Disordered 84. Osman C, Haag M, Potting C, Rodenfels J, Dip PV, Wieland lipid metabolism and the pathogenesis of insulin resistance. FT, Bru¨ gger B, Westermann B, Langer T 2009 The genetic Physiol Rev 87:507–520 interactome of prohibitins: coordinated control of cardiolipin 98. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M, and phosphatidylethanolamine by conserved regulators in Ilkayeva O, Bain J, Stevens R, Dyck JR, Newgard CB, mitochondria. J Cell Biol 184:583–596 Lopaschuk GD, Muoio DM 2008 Mitochondrial overload 85. Kutik S, Rissler M, Guan XL, Guiard B, Shui G, Gebert N, and incomplete fatty acid oxidation contribute to skeletal Heacock PN, Rehling P, Dowhan W, Wenk MR, Pfanner muscle insulin resistance. Cell Metab 7:45–56 N, Wiedemann N 2008 The translocator maintenance pro- 99. Holland WL, Brozinick JT, Wang LP, Hawkins ED, Sargent tein Tam41 is required for mitochondrial cardiolipin bio- KM, Liu Y, Narra K, Hoehn KL, Knotts TA, Siesky A, Nelson synthesis. J Cell Biol 183:1213–1221 DH, Karathanasis SK, Fontenot GK, Birnbaum MJ, 86. Xu Y, Condell M, Plesken H, Edelman-Novemsky I, Ma J, Summers SA 2007 Inhibition of ceramide synthesis amelio- Ren M, Schlame M 2006 A Drosophila model of Barth rates glucocorticoid-, saturated-fat-, and obesity-induced in- syndrome. Proc Natl Acad Sci USA 103:11584–11588 sulin resistance. Cell Metab 5:167–179 87. Becker T, Pfannschmidt S, Guiard B, Stojanovski D, 100. Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y, Milenkovic D, Kutik S, Pfanner N, Meisinger C, Bergeron R, Kim JK, Cushman SW, Cooney GJ, Atcheson Wiedemann N 2008 Biogenesis of the mitochondrial B, White MF, Kraegen EW, Shulman GI 2002 Mechanism TOM complex: Mim1 promotes insertion and assembly by which fatty acids inhibit insulin activation of insulin of signal-anchored receptors. J Biol Chem 283:120–127 receptor substrate-1 (IRS-1)-associated phosphatidylino- 88. Claypool SM, Oktay Y, Boontheung P, Loo JA, Koehler sitol 3-kinase activity in muscle. J Biol Chem 277:50230– CM 2008 Cardiolipin defines the interactome of the major 50236 ADP/ATP carrier protein of the mitochondrial inner mem- 101. Chibalin AV, Leng Y, Vieira E, Krook A, Bjo¨ rnholm M, brane. J Cell Biol 182:937–950 Long YC, Kotova O, Zhong Z, Sakane F, Steiler T, Nyle´n 89. Xu Y, Sutachan JJ, Plesken H, Kelley RI, Schlame M 2005 C, Wang J, Laakso M, Topham MK, Gilbert M, Wallberg- Characterization of lymphoblast mitochondria from pa- Henriksson H, Zierath JR 2008 Downregulation of diac- tients with Barth syndrome. Lab Invest 85:823–830 ylglycerol kinase ␦ contributes to hyperglycemia-induced 90. Luft R, Ikkos D, Palmieri G, Ernster L, Afzelius B 1962 A insulin resistance. Cell 132:375–386 case of severe hypermetabolism of nonthyroid origin with 102. Houstis N, Rosen ED, Lander ES 2006 Reactive oxygen a defect in the maintenance of mitochondrial respiratory species have a causal role in multiple forms of insulin re- control: a correlated clinical, biochemical, and morpho- sistance. Nature 440:944–948 logical study. J Clin Invest 41:1776–1804 103. Lanner JT, Bruton JD, Katz A, Westerblad H 2008 Ca(2ϩ) 91. Ducluzeau PH, Perretti N, Laville M, Andreelli F, Vega N, and insulin-mediated glucose uptake. Curr Opin Pharma- Riou JP, Vidal H 2001 Regulation by insulin of gene ex- col 8:339–345 pression in human skeletal muscle and adipose tissue. Ev- 104. Lebeche D, Davidoff AJ, Hajjar RJ 2008 Interplay between idence for specific defects in type 2 diabetes. Diabetes 50: impaired calcium regulation and insulin signaling abnor- 1134–1142 malities in diabetic cardiomyopathy. Nat Clin Pract Car- 92. Vaag A, Henriksen JE, Beck-Nielsen H 1992 Decreased diovasc Med 5:715–724 insulin activation of glycogen synthase in skeletal muscles 105. Park S, Scheffler TL, Gunawan AM, Shi H, Zeng C, Hannon Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 389

KM, Grant AL, Gerrard DE 2009 Chronic elevated calcium Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstråle M, blocks AMPK-induced GLUT-4 expression in skeletal mus- Laurila E, Houstis N, Daly MJ, Patterson N, Mesirov JP, cle. Am J Physiol Cell Physiol 296:C106–C115 Golub TR, Tamayo P, Spiegelman B, Lander ES, Hirschhorn 106. Kacerovsky-Bielesz G, Chmelik M, Ling C, Pokan R, JN, Altshuler D, Groop LC 2003 PGC-1␣-responsive genes Szendroedi J, Farukuoye M, Kacerovsky M, Schmid AI, involved in oxidative phosphorylation are coordinately Gruber S, Wolzt M, Moser E, Pacini G, Smekal G, Groop downregulated in human diabetes. Nat Genet 34:267–273 L, Roden M 2009 Short-term exercise training does not 122. Patti ME, Butte AJ, Crunkhorn S, Cusi K, Berria R, stimulate skeletal muscle ATP synthesis in relatives of hu- Kashyap S, Miyazaki Y, Kohane I, Costello M, Saccone R, mans with type 2 diabetes. Diabetes 58:1333–1341 Landaker EJ, Goldfine AB, Mun E, DeFronzo R, Finlayson 107. Simoneau JA, Colberg SR, Thaete FL, Kelley DE 1995 J, Kahn CR, Mandarino LJ 2003 Coordinated reduction of Skeletal muscle glycolytic and oxidative enzyme capacities genes of oxidative metabolism in humans with insulin re- are determinants of insulin sensitivity and muscle compo- sistance and diabetes: potential role of PGC1 and NRF1.

sition in obese women. FASEB J 9:273–278 Proc Natl Acad Sci USA 100:8466–8471 Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 108. Simoneau JA, Bouchard C 1995 Skeletal muscle metabo- 123. Sreekumar R, Halvatsiotis P, Schimke JC, Nair KS 2002 Gene lism and body fat content in men and women. Obes Res expression profile in skeletal muscle of type 2 diabetes and the 3:23–29 effect of insulin treatment. Diabetes 51:1913–1920 109. Simoneau JA, Kelley DE 1997 Altered glycolytic and ox- 124. Morino K, Petersen KF, Dufour S, Befroy D, Frattini J, idative capacities of skeletal muscle contribute to insulin Shatzkes N, Neschen S, White MF, Bilz S, Sono S, Pypaert resistance in NIDDM. J Appl Physiol 83:166–171 M, Shulman GI 2005 Reduced mitochondrial density and 110. Bruce CR, Anderson MJ, Carey AL, Newman DG, Bonen increased IRS-1 serine phosphorylation in muscle of insu- A, Kriketos AD, Cooney GJ, Hawley JA 2003 Muscle ox- lin-resistant offspring of type 2 diabetic parents. J Clin idative capacity is a better predictor of insulin sensitivity Invest 115:3587–3593 than lipid status. J Clin Endocrinol Metab 88:5444–5451 125. Nair KS, Bigelow ML, Asmann YW, Chow LS, Coenen- 111. Kelley DE, Goodpaster B, Wing RR, Simoneau JA 1999 Schimke JM, Klaus KA, Guo ZK, Sreekumar R, Irving BA Skeletal muscle fatty acid metabolism in association with 2008 Asian Indians have enhanced skeletal muscle mito- insulin resistance, obesity, and weight loss. Am J Physiol chondrial capacity to produce ATP in association with se- 277:E1130–E1141 vere insulin resistance. Diabetes 57:1166–1175 112. Kelley DE, Mandarino LJ 2000 Fuel selection in human 126. Frederiksen CM, Højlund K, Hansen L, Oakeley EJ, skeletal muscle in insulin resistance: a reexamination. Hemmings B, Abdallah BM, Brusgaard K, Beck-Nielsen Diabetes 49:677–683 H, Gaster M 2008 Transcriptional profiling of myo- 113. Zurlo F, Lillioja S, Esposito-Del Puente A, Nyomba BL, tubes from patients with type 2 diabetes: no evidence for Raz I, Saad MF, Swinburn BA, Knowler WC, Bogardus C, a primary defect in oxidative phosphorylation genes. Ravussin E 1990 Low ratio of fat to carbohydrate oxida- Diabetologia 51:2068–2077 tion as predictor of weight gain: study of 24-h RQ. Am J 127. Crunkhorn S, Dearie F, Mantzoros C, Gami H, da Silva Physiol 259:E650–E657 WS, Espinoza D, Faucette R, Barry K, Bianco AC, Patti ME 114. Ritov VB, Menshikova EV, He J, Ferrell RE, Goodpaster 2007 Peroxisome proliferator activator receptor ␥ coacti- BH, Kelley DE 2005 Deficiency of subsarcolemmal mito- vator-1 expression is reduced in obesity: potential patho- chondria in obesity and type 2 diabetes. Diabetes 54:8–14 genic role of saturated fatty acids and p38 mitogen-acti- 115. Boushel R, Gnaiger E, Schjerling P, Skovbro M, Kraunsøe vated activation. J Biol Chem 282:15439– R, Dela F 2007 Patients with type 2 diabetes have normal 15450 mitochondrial function in skeletal muscle. Diabetologia 128. Sparks LM, Xie H, Koza RA, Mynatt R, Hulver MW, Bray 50:790–796 GA, Smith SR 2005 A high-fat diet coordinately down- 116. Kelley DE, He J, Menshikova EV, Ritov VB 2002 Dys- regulates genes required for mitochondrial oxidative phos- function of mitochondria in human skeletal muscle in type phorylation in skeletal muscle. Diabetes 54:1926–1933 2 diabetes. Diabetes 51:2944–2950 129. Richardson DK, Kashyap S, Bajaj M, Cusi K, Mandarino 117. Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI SJ, Finlayson J, DeFronzo RA, Jenkinson CP, Mandarino 2004 Impaired mitochondrial activity in the insulin-resis- LJ 2005 Lipid infusion decreases the expression of nuclear tant offspring of patients with type 2 diabetes. N Engl encoded mitochondrial genes and increases the expression J Med 350:664–671 of extracellular matrix genes in human skeletal muscle. 118. Befroy DE, Petersen KF, Dufour S, Mason GF, de Graaf J Biol Chem 280:10290–10297 RA, Rothman DL, Shulman GI 2007 Impaired mitochon- 130. Garcia-Roves P, Huss JM, Han DH, Hancock CR, Iglesias- drial substrate oxidation in muscle of insulin-resistant off- Gutierrez E, Chen M, Holloszy JO 2007 Raising plasma spring of type 2 diabetic patients. Diabetes 56:1376–1381 fatty acid concentration induces increased biogenesis of 119. Mogensen M, Sahlin K, Fernstro¨ m M, Glintborg D, Vind mitochondria in skeletal muscle. Proc Natl Acad Sci USA BF, Beck-Nielsen H, Højlund K 2007 Mitochondrial res- 104:10709–10713 piration is decreased in skeletal muscle of patients with 131. Hancock CR, Han DH, Chen M, Terada S, Yasuda T, type 2 diabetes. Diabetes 56:1592–1599 Wright DC, Holloszy JO 2008 High-fat diets cause insulin 120. Petersen KF, Dufour S, Shulman GI 2005 Decreased insu- resistance despite an increase in muscle mitochondria. Proc lin-stimulated ATP synthesis and phosphate transport in Natl Acad Sci USA 105:7815–7820 muscle of insulin-resistant offspring of type 2 diabetic par- 132. Sreekumar R, Unnikrishnan J, Fu A, Nygren J, Short KR, ents. PLoS Med 2:e233 Schimke J, Barazzoni R, Nair KS 2002 Effects of caloric 121. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, restriction on mitochondrial function and gene tran- 390 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395

scripts in rat muscle. Am J Physiol Endocrinol Metab PGC-1␣ promoter through DNMT3B controls mitochon- 283:E38–E43 drial density. Cell Metab 10:189–198 133. Parikh H, Nilsson E, Ling C, Poulsen P, Almgren P, Nittby 147. Lerin C, Rodgers JT, Kalume DE, Kim SH, Pandey A, H, Eriksson KF, Vaag A, Groop LC 2008 Molecular cor- Puigserver P 2006 GCN5 acetyltransferase complex controls relates for maximal oxygen uptake and type 1 fibers. Am J glucose metabolism through transcriptional repression of Physiol Endocrinol Metab 294:E1152–E1159 PGC-1␣. Cell Metab 3:429–438 134. Wisløff U, Najjar SM, Ellingsen O, Haram PM, Swoap S, 148. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Al-Share Q, Fernstro¨ m M, Rezaei K, Lee SJ, Koch LG, Lerin C, Daussin F, Messadeq N, Milne J, Lambert P, Britton SL 2005 Cardiovascular risk factors emerge after Elliott P, Geny B, Laakso M, Puigserver P, Auwerx J 2006 artificial selection for low aerobic capacity. Science 307: Resveratrol improves mitochondrial function and protects 418–420 against metabolic disease by activating SIRT1 and PGC- 135. Wright DC, Han DH, Garcia-Roves PM, Geiger PC, Jones 1␣. Cell 127:1109–1122

TE, Holloszy JO 2007 Exercise-induced mitochondrial 149. Astrup A 1986 Thermogenesis in human brown adipose Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 ␣ biogenesis begins before the increase in muscle PGC-1 tissue and skeletal muscle induced by sympathomimetic expression. J Biol Chem 282:194–199 stimulation. Acta Endocrinol Suppl (Copenh) 278:1–32 136. Ro¨ nn T, Poulsen P, Hansson O, Holmkvist J, Almgren P, 150. Cannon B, Nedergaard J 1985 The biochemistry of an Nilsson P, Tuomi T, Isomaa B, Groop L, Vaag A, Ling C inefficient tissue: brown adipose tissue. Essays Biochem 2008 Age influences DNA methylation and gene expres- 20:110–164 sion of COX7A1 in human skeletal muscle. Diabetologia 151. Cypess AM, Lehman S, Williams G, Tal I, Rodman D, 51:1159–1168 Goldfine AB, Kuo FC, Palmer EL, Tseng YH, Doria A, 137. Ling C, Poulsen P, Carlsson E, Ridderstråle M, Almgren P, Kolodny GM, Kahn CR 2009 Identification and impor- Wojtaszewski J, Beck-Nielsen H, Groop L, Vaag A 2004 tance of brown adipose tissue in adult humans. N Engl Multiple environmental and genetic factors influence skel- J Med 360:1509–1517 ␣ ␤ etal muscle PGC-1 and PGC-1 gene expression in twins. 152. Weber WA 2004 Brown adipose tissue and nuclear med- J Clin Invest 114:1518–1526 icine imaging. J Nucl Med 45:1101–1103 138. Ro¨ nn T, Poulsen P, Tuomi T, Isomaa B, Groop L, Vaag A, 153. Bouillaud F, Ricquier D, Thibault J, Weissenbach J 1985 Ling C 2009 Genetic variation in ATP5O is associated with Molecular approach to thermogenesis in brown adipose skeletal muscle ATP50 mRNA expression and glucose up- tissue: cDNA cloning of the mitochondrial uncoupling take in young twins. PLoS ONE 4:e4793 protein. Proc Natl Acad Sci USA 82:445–448 139. Ling C, Groop L 2009 Epigenetics: a molecular link be- 154. Jacobsson A, Stadler U, Glotzer MA, Kozak LP 1985 Mi- tween environmental factors and type 2 diabetes. Diabetes tochondrial uncoupling protein from mouse brown fat. 58:2718–2725 Molecular cloning, genetic mapping, and mRNA expres- 140. Petersen KF, Befroy D, Dufour S, Dziura J, Ariyan C, Roth- sion. J Biol Chem 260:16250–16254 man DL, DiPietro L, Cline GW, Shulman GI 2003 Mitochon- 155. Ricquier D, Bouillaud F 1997 The mitochondrial uncou- drial dysfunction in the elderly: possible role in insulin resis- pling protein: structural and genetic studies. Prog Nucleic tance. Science 300:1140–1142 Acid Res Mol Biol 56:83–108 141. Yechoor VK, Patti ME, Saccone R, Kahn CR 2002 Coor- dinated patterns of gene expression for substrate and en- 156. Luo GF, Yu TY, Wen XH, Li Y, Yang GS 2008 Alteration ergy metabolism in skeletal muscle of diabetic mice. Proc of mitochondrial oxidative capacity during porcine prea- Natl Acad Sci USA 99:10587–10592 dipocyte differentiation and in response to leptin. Mol Cell 142. Karakelides H, Asmann YW, Bigelow ML, Short KR, Biochem 307:83–91 Dhatariya K, Coenen-Schimke J, Kahl J, Mukhopadhyay D, 157. Kim BW, Choo HJ, Lee JW, Kim JH, Ko YG 2004 Extra- Nair KS 2007 Effect of insulin deprivation on muscle mito- cellular ATP is generated by ATP synthase complex in adi- chondrial ATP production and gene transcript levels in type pocyte lipid rafts. Exp Mol Med 36:476–485 1 diabetic subjects. Diabetes 56:2683–2689 158. Wilson-Fritch L, Burkart A, Bell G, Mendelson K, Leszyk 143. Meugnier E, Faraj M, Rome S, Beauregard G, Michaut A, J, Nicoloro S, Czech M, Corvera S 2003 Mitochondrial Pelloux V, Chiasson JL, Laville M, Clement K, Vidal H, biogenesis and remodeling during adipogenesis and in re- Rabasa-Lhoret R 2007 Acute hyperglycemia induces a sponse to the insulin sensitizer rosiglitazone. Mol Cell Biol global downregulation of gene expression in adipose tis- 23:1085–1094 sue and skeletal muscle of healthy subjects. Diabetes 159. Owen OE, Kalhan SC, Hanson RW 2002 The key role of 56:992–999 anaplerosis and cataplerosis for citric acid cycle function. 144. Rabøl R, Højberg PM, Almdal T, Boushel R, Haugaard J Biol Chem 277:30409–30412 SB, Madsbad S, Dela F 2009 Effect of hyperglycemia on 160. Koh EH, Park JY, Park HS, Jeon MJ, Ryu JW, Kim M, Kim mitochondrial respiration in type 2 diabetes. J Clin Endo- SY, Kim MS, Kim SW, Park IS, Youn JH, Lee KU 2007 crinol Metab 94:1372–1378 Essential role of mitochondrial function in adiponectin 145. Southgate RJ, Bruce CR, Carey AL, Steinberg GR, Walder K, synthesis in adipocytes. Diabetes 56:2973–2981 Monks R, Watt MJ, Hawley JA, Birnbaum MJ, Febbraio MA 161. Shi X, Burkart A, Nicoloro SM, Czech MP, Straubhaar J, 2005 PGC-1␣ gene expression is down-regulated by Akt-me- Corvera S 2008 Paradoxical effect of mitochondrial respi- diated phosphorylation and nuclear exclusion of FoxO1 in ratory chain impairment on insulin signaling and glucose insulin-stimulated skeletal muscle. FASEB J 19:2072–2074 transport in adipose cells. J Biol Chem 283:30658–30667 146. Barre`s R, Osler ME, Yan J, Rune A, Fritz T, Caidahl K, 162. Marette A, Tulp OL, Bukowiecki LJ 1991 Mechanism Krook A, Zierath JR 2009 Non-CpG methylation of the linking insulin resistance to defective thermogenesis in Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 391

brown adipose tissue of obese diabetic SHR/N-cp rats. Int ated with obesity and treatment with rosiglitazone. J Clin J Obes 15:823–831 Invest 114:1281–1289 163. Nedergaard J, Bengtsson T, Cannon B 2007 Unexpected 178. Maassen JA, ’t Hart LM, Janssen GM, Reiling E, Romijn evidence for active brown adipose tissue in adult humans. JA, Lemkes HH 2006 Mitochondrial diabetes and its les- Am J Physiol Endocrinol Metab 293:E444–E452 sons for common type 2 diabetes. Biochem Soc Trans 34: 164. Saito M, Okamatsu-Ogura Y, Matsushita M, Watanabe K, 819–823 Yoneshiro T, Nio-Kobayashi J, Iwanaga T, Miyagawa M, 179. Maasen JA 2008 Mitochondria, body fat and type 2 dia- Kameya T, Nakada K, Kawai Y, Tsujisaki M 2009 High betes: what is the connection? Minerva Med 99:241–251 incidence of metabolically active brown adipose tissue in 180. Heilbronn L, Smith SR, Ravussin E 2004 Failure of fat cell healthy adult humans: effects of cold exposure and adi- proliferation, mitochondrial function and fat oxidation re- posity. Diabetes 58:1526–1531 sults in ectopic fat storage, insulin resistance and type II 165. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders diabetes mellitus. Int J Obes Relat Metab Disord 28(Suppl

NM, Drossaerts JM, Kemerink GJ, Bouvy ND, Schrauwen 4):S12–S21 Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 P, Teule GJ 2009 Cold-activated brown adipose tissue in 181. Frayn KN, Langin D, Karpe F 2008 Fatty acid-induced healthy men. N Engl J Med 360:1500–1508 mitochondrial uncoupling in adipocytes is not a promising 166. Virtanen KA, Lidell ME, Orava J, Heglind M, Westergren target for treatment of insulin resistance unless adipocyte R, Niemi T, Taittonen M, Laine J, Savisto NJ, Enerba¨ckS, oxidative capacity is increased. Diabetologia 51:394–397 Nuutila P 2009 Functional brown adipose tissue in healthy 182. de Ferranti S, Mozaffarian D 2008 The perfect storm: obe- adults. N Engl J Med 360:1518–1525 sity, adipocyte dysfunction, and metabolic consequences. 167. Zingaretti MC, Crosta F, Vitali A, Guerrieri M, Frontini A, Clin Chem 54:945–955 Cannon B, Nedergaard J, Cinti S 2009 The presence of 183. Rossmeisl M, Flachs P, Brauner P, Sponarova J, Matejkova UCP1 demonstrates that metabolically active adipose tis- O, Prazak T, Ruzickova J, Bardova K, Kuda O, Kopecky sue in the neck of adult humans truly represents brown J 2004 Role of energy charge and AMP-activated protein adipose tissue. FASEB J 23:3113–3120 kinase in adipocytes in the control of body fat stores. Int J 168. Cinti S 2002 Adipocyte differentiation and transdifferen- Obes Relat Metab Disord 28(Suppl 4):S38–S44 184. Harper JA, Dickinson K, Brand MD 2001 Mitochondrial tiation: plasticity of the adipose organ. J Endocrinol Invest uncoupling as a target for drug development for the treat- 25:823–835 ment of obesity. Obes Rev 2:255–265 169. Deveaud C, Beauvoit B, Salin B, Schaeffer J, Rigoulet M 185. Stefl B, Janovska´ A, Hodny´ Z, Rossmeisl M, Hora´kova´M, 2004 Regional differences in oxidative capacity of rat Syrovy´I,Be´mova´ J, Bendlova´ B, Kopecky´J1998 Brown white adipose tissue are linked to the mitochondrial con- fat is essential for cold-induced thermogenesis but not for tent of mature adipocytes. Mol Cell Biochem 267:157–166 obesity resistance in aP2-Ucp mice. Am J Physiol 274: 170. Bogacka I, Xie H, Bray GA, Smith SR 2005 Pioglitazone E527–E533 induces mitochondrial biogenesis in human subcutaneous 186. Rossmeisl M, Syrovy´ I, Baumruk F, Flachs P, Janovska´P, adipose tissue in vivo. Diabetes 54:1392–1399 Kopecky´J2000 Decreased fatty acid synthesis due to mi- 171. Himms-Hagen J, Melnyk A, Zingaretti MC, Ceresi E, tochondrial uncoupling in adipose tissue. FASEB J 14: Barbatelli G, Cinti S 2000 Multilocular fat cells in WAT of 1793–1800 CL-316243-treated rats derive directly from white adipo- 187. Si Y, Palani S, Jayaraman A, Lee K 2007 Effects of forced cytes. Am J Physiol Cell Physiol 279:C670–C681 uncoupling protein 1 expression in 3T3–L1 cells on mito- 172. Loncar D 1991 Convertible adipose tissue in mice. Cell chondrial function and lipid metabolism. J Lipid Res 48: Tissue Res 266:149–161 826–836 173. Orci L, Cook WS, Ravazzola M, Wang MY, Park BH, 188. Choo HJ, Kim JH, Kwon OB, Lee CS, Mun JY, Han SS, Montesano R, Unger RH 2004 Rapid transformation of Yoon YS, Yoon G, Choi KM, Ko YG 2006 Mitochondria white adipocytes into fat-oxidizing machines. Proc Natl are impaired in the adipocytes of type 2 diabetic mice. Acad Sci USA 101:2058–2063 Diabetologia 49:784–791 174. Stro¨ m K, Hansson O, Lucas S, Nevsten P, Fernandez C, 189. Okamoto Y, Higashiyama H, Rong JX, McVey MJ, Klint C, Move´rare-Skrtic S, Sundler F, Ohlsson C, Holm C Kinoshita M, Asano S, Hansen MK 2007 Comparison of 2008 Attainment of brown adipocyte features in white adi- mitochondrial and macrophage content between subcu- pocytes of hormone-sensitive lipase null mice. PLoS ONE taneous and visceral fat in db/db mice. Exp Mol Pathol 3:e1793 83:73–83 175. Tiraby C, Tavernier G, Lefort C, Larrouy D, Bouillaud F, 190. Rong JX, Qiu Y, Hansen MK, Zhu L, Zhang V, Xie M, Ricquier D, Langin D 2003 Acquirement of brown fat cell Okamoto Y, Mattie MD, Higashiyama H, Asano S, Strum features by human white adipocytes. J Biol Chem 278: JC, Ryan TE 2007 Adipose mitochondrial biogenesis is 33370–33376 suppressed in db/db and high-fat diet-fed mice and im- 176. Toh SY, Gong J, Du G, Li JZ, Yang S, Ye J, Yao H, Zhang proved by rosiglitazone. Diabetes 56:1751–1760 Y, Xue B, Li Q, Yang H, Wen Z, Li P 2008 Up-regulation 191. Sutherland LN, Capozzi LC, Turchinsky NJ, Bell RC, of mitochondrial activity and acquirement of brown adi- Wright DC 2008 Time course of high-fat diet-induced re- pose tissue-like property in the white adipose tissue of ductions in adipose tissue mitochondrial proteins: poten- fsp27 deficient mice. PLoS ONE 3:e2890 tial mechanisms and the relationship to glucose intoler- 177. Wilson-Fritch L, Nicoloro S, Chouinard M, Lazar MA, ance. Am J Physiol Endocrinol Metab 295:E1076–E1083 Chui PC, Leszyk J, Straubhaar J, Czech MP, Corvera S 192. Kaaman M, Sparks LM, van Harmelen V, Smith SR, Sjo¨ lin 2004 Mitochondrial remodeling in adipose tissue associ- E, Dahlman I, Arner P 2007 Strong association between 392 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395

mitochondrial DNA copy number and lipogenesis in hu- Kasuga M 2008 FSP27 contributes to efficient energy stor- man white adipose tissue. Diabetologia 50:2526–2533 age in murine white adipocytes by promoting the forma- 193. Dahlman I, Forsgren M, Sjo¨ gren A, Nordstro¨ m EA, Kaaman tion of unilocular lipid droplets. J Clin Invest 118:2808– M, Na¨slund E, Attersand A, Arner P 2006 Downregulation 2821 of electron transport chain genes in visceral adipose tissue in 206. Semple RK, Crowley VC, Sewter CP, Laudes M, type 2 diabetes independent of obesity and possibly involving Christodoulides C, Considine RV, Vidal-Puig A, O’Rahilly tumor necrosis factor-␣. Diabetes 55:1792–1799 S 2004 Expression of the thermogenic nuclear hormone 194. Boden G, Homko C, Mozzoli M, Showe LC, Nichols C, receptor coactivator PGC-1␣ is reduced in the adipose tis- Cheung P 2005 Thiazolidinediones upregulate fatty acid sue of morbidly obese subjects. Int J Obes Relat Metab uptake and oxidation in adipose tissue of diabetic patients. Disord 28:176–179 Diabetes 54:880–885 207. Kotronen A, Seppa¨la¨-Lindroos A, Bergholm R, Yki-Ja¨rvinen 195. Bogacka I, Ukropcova B, McNeil M, Gimble JM, Smith SR H 2008 Tissue specificity of insulin resistance in humans: fat

2005 Structural and functional consequences of mitochon- in the liver rather than muscle is associated with features of the Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 drial biogenesis in human adipocytes in vitro. J Clin En- metabolic syndrome. Diabetologia 51:130–138 docrinol Metab 90:6650–6656 208. Michael MD, Kulkarni RN, Postic C, Previs SF, Shulman 196. Digby JE, Montague CT, Sewter CP, Sanders L, Wilkison GI, Magnuson MA, Kahn CR 2000 Loss of insulin signal- WO, O’Rahilly S, Prins JB 1998 Thiazolidinedione expo- ing in hepatocytes leads to severe insulin resistance and sure increases the expression of uncoupling protein 1 in progressive hepatic dysfunction. Mol Cell 6:87–97 cultured human preadipocytes. Diabetes 47:138–141 209. Biddinger SB, Hernandez-Ono A, Rask-Madsen C, Haas 197. Goetzman ES 2009 The regulation of acyl-CoA dehydro- JT, Alema´n JO, Suzuki R, Scapa EF, Agarwal C, Carey genases in adipose tissue by rosiglitazone. Obesity (Silver MC, Stephanopoulos G, Cohen DE, King GL, Ginsberg Spring) 17:196–198 HN, Kahn CR 2008 Hepatic insulin resistance is sufficient 198. Laplante M, Festuccia WT, Soucy G, Ge´linas Y, Lalonde J, to produce dyslipidemia and susceptibility to atheroscle- Berger JP, Deshaies Y 2006 Mechanisms of the depot rosis. Cell Metab 7:125–134 specificity of peroxisome proliferator-activated recep- 210. Veltri KL, Espiritu M, Singh G 1990 Distinct genomic copy tor ␥ action on adipose tissue metabolism. Diabetes 55: number in mitochondria of different mammalian organs. 2771–2778 J Cell Physiol 143:160–164 199. Granneman JG, Li P, Zhu Z, Lu Y 2005 Metabolic and 211. Benard G, Faustin B, Passerieux E, Galinier A, Rocher C, cellular plasticity in white adipose tissue I: effects of ␤3- Bellance N, Delage JP, Casteilla L, Letellier T, Rossignol R adrenergic receptor activation. Am J Physiol Endocrinol 2006 Physiological diversity of mitochondrial oxidative Metab 289:E608–E616 phosphorylation. Am J Physiol Cell Physiol 291:C1172– 200. Tedesco L, Valerio A, Cervino C, Cardile A, Pagano C, C1182 Vettor R, Pasquali R, Carruba MO, Marsicano G, Lutz B, 212. Schmid AI, Chmelík M, Szendroedi J, Krssa´k M, Brehm A, Pagotto U, Nisoli E 2008 Cannabinoid type 1 receptor Moser E, Roden M 2008 Quantitative ATP synthesis in blockade promotes mitochondrial biogenesis through en- human liver measured by localized 31P spectroscopy using dothelial nitric oxide synthase expression in white adipo- the magnetization transfer experiment. NMR Biomed 21: cytes. Diabetes 57:2028–2036 437–443 201. Katic M, Kennedy AR, Leykin I, Norris A, McGettrick A, 213. Agarwal AK, Garg A 2006 Genetic disorders of adipose Gesta S, Russell SJ, Bluher M, Maratos-Flier E, Kahn CR tissue development, differentiation, and death. Annu Rev 2007 Mitochondrial gene expression and increased oxida- Genomics Hum Genet 7:175–199 tive metabolism: role in increased lifespan of fat-specific 214. Kotronen A, Juurinen L, Tiikkainen M, Vehkavaara S, insulin receptor knock-out mice. Aging Cell 6:827–839 Yki-Ja¨rvinen H 2008 Increased liver fat, impaired insulin 202. Seale P, Kajimura S, Yang W, Chin S, Rohas LM, Uldry M, clearance, and hepatic and adipose tissue insulin resistance Tavernier G, Langin D, Spiegelman BM 2007 Transcrip- in type 2 diabetes. Gastroenterology 135:122–130 tional control of brown fat determination by PRDM16. 215. Petersen KF, Dufour S, Befroy D, Lehrke M, Hendler RE, Cell Metab 6:38–54 Shulman GI 2005 Reversal of nonalcoholic hepatic steato- 203. Seale P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, Scime` sis, hepatic insulin resistance, and hyperglycemia by mod- A, Devarakonda S, Conroe HM, Erdjument-Bromage H, erate weight reduction in patients with type 2 diabetes. Tempst P, Rudnicki MA, Beier DR, Spiegelman BM 2008 Diabetes 54:603–608 PRDM16 controls a brown fat/skeletal muscle switch. Na- 216. Koska J, Stefan N, Permana PA, Weyer C, Sonoda M, ture 454:961–967 Bogardus C, Smith SR, Joanisse DR, Funahashi T, Krakoff 204. Timmons JA, Wennmalm K, Larsson O, Walden TB, J, Bunt JC 2008 Increased fat accumulation in liver may Lassmann T, Petrovic N, Hamilton DL, Gimeno RE, link insulin resistance with subcutaneous abdominal adi- Wahlestedt C, Baar K, Nedergaard J, Cannon B 2007 pocyte enlargement, visceral adiposity, and hypoadi- Myogenic gene expression signature establishes that ponectinemia in obese individuals. Am J Clin Nutr 87: brown and white adipocytes originate from distinct cell 295–302 lineages. Proc Natl Acad Sci USA 104:4401–4406 217. Hwang JH, Stein DT, Barzilai N, Cui MH, Tonelli J, 205. Nishino N, Tamori Y, Tateya S, Kawaguchi T, Shibakusa Kishore P, Hawkins M 2007 Increased intrahepatic tri- T, Mizunoya W, Inoue K, Kitazawa R, Kitazawa S, Mat- glyceride is associated with peripheral insulin resistance: in suki Y, Hiramatsu R, Masubuchi S, Omachi A, Kimura K, vivo MR imaging and spectroscopy studies. Am J Physiol Saito M, Amo T, Ohta S, Yamaguchi T, Osumi T, Cheng Endocrinol Metab 293:E1663–E1669 J, Fujimoto T, Nakao H, Aiba A, Okamura H, Fushiki T, 218. Korenblat KM, Fabbrini E, Mohammed BS, Klein S 2008 Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 393

Liver, muscle, and adipose tissue insulin action is directly oxidative phosphorylation in livers of diabetic patients. related to intrahepatic triglyceride content in obese sub- Obesity (Silver Spring) 16:2601–2609 jects. Gastroenterology 134:1369–1375 232. Pihlajama¨ki J, Boes T, Kim EY, Dearie F, Kim BW, 219. Kim JK, Fillmore JJ, Chen Y, Yu C, Moore IK, Pypaert M, Schroeder J, Mun E, Nasser I, Park PJ, Bianco AC, Lutz EP, Kako Y, Velez-Carrasco W, Goldberg IJ, Breslow Goldfine AB, Patti ME 2009 Thyroid hormone-related JL, Shulman GI 2001 Tissue-specific overexpression of li- regulation of gene expression in human fatty liver. J Clin poprotein lipase causes tissue-specific insulin resistance. Endocrinol Metab 94:3521–3529 Proc Natl Acad Sci USA 98:7522–7527 233. Pe´rez-Carreras M, Del Hoyo P, Martín MA, Rubio JC, 220. Nakamura S, Takamura T, Matsuzawa-Nagata N, Martín A, Castellano G, Colina F, Arenas J, Solis-Herruzo Takayama H, Misu H, Noda H, Nabemoto S, Kurita S, JA 2003 Defective hepatic mitochondrial respiratory chain Ota T, Ando H, Miyamoto K, Kaneko S 2009 Palmitate in patients with nonalcoholic steatohepatitis. Hepatology induces insulin resistance in H4IIEC3 hepatocytes through 38:999–1007

reactive oxygen species produced by mitochondria. J Biol 234. Greenfield V, Cheung O, Sanyal AJ 2008 Recent advances Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 Chem 284:14809–14818 in nonalcoholic fatty liver disease. Curr Opin Gastroen- 221. Ruddock MW, Stein A, Landaker E, Park J, Cooksey RC, terol 24:320–327 McClain D, Patti ME 2008 Saturated fatty acids inhibit 235. Sanyal AJ, Campbell-Sargent C, Mirshahi F, Rizzo WB, hepatic insulin action by modulating insulin receptor Contos MJ, Sterling RK, Luketic VA, Shiffman ML, Clore expression and post-receptor signalling. J Biochem 144: JN 2001 Nonalcoholic steatohepatitis: association of in- 599–607 sulin resistance and mitochondrial abnormalities. Gastro- 222. Hertz R, Magenheim J, Berman I, Bar-Tana J 1998 Fatty enterology 120:1183–1192 acyl-CoA thioesters are ligands of hepatic nuclear factor- 236. Kohjima M, Enjoji M, Higuchi N, Kato M, Kotoh K, 4␣. Nature 392:512–516 Yoshimoto T, Fujino T, Yada M, Yada R, Harada N, 223. Lin J, Tarr PT, Yang R, Rhee J, Puigserver P, Newgard CB, Takayanagi R, Nakamuta M 2007 Re-evaluation of Spiegelman BM 2003 PGC-1␤ in the regulation of he- fatty acid metabolism-related gene expression in nonal- patic glucose and energy metabolism. J Biol Chem 278: coholic fatty liver disease. Int J Mol Med 20:351–358 30843–30848 237. O’Connor MD, Landahl H, Grodsky GM 1980 Compar- 224. Xu HE, Lambert MH, Montana VG, Parks DJ, Blanchard ison of storage- and signal-limited models of pancreatic SG, Brown PJ, Sternbach DD, Lehmann JM, Wisely GB, insulin secretion. Am J Physiol 238:R378–R389 Willson TM, Kliewer SA, Milburn MV 1999 Molecular 238. Hasan NM, Longacre MJ, Stoker SW, Boonsaen T, recognition of fatty acids by peroxisome proliferator-ac- Jitrapakdee S, Kendrick MA, Wallace JC, MacDonald tivated receptors. Mol Cell 3:397–403 MJ 2008 Impaired anaplerosis and insulin secretion in 225. Duplus E, Glorian M, Forest C 2000 Fatty acid regulation insulinoma cells caused by small interfering RNA-me- of gene transcription. J Biol Chem 275:30749–30752 diated suppression of pyruvate carboxylase. J Biol Chem 226. Seppa¨la¨-Lindroos A, Vehkavaara S, Ha¨kkinen AM, Goto 283:28048–28059 T, Westerbacka J, Sovija¨rvi A, Halavaara J, Yki-Ja¨rvinen 239. Evans JL, Goldfine ID, Maddux BA, Grodsky GM 2003 H 2002 Fat accumulation in the liver is associated with Are oxidative stress-activated signaling pathways media- defects in insulin suppression of glucose production and tors of insulin resistance and ␤-cell dysfunction? Diabetes serum free fatty acids independent of obesity in normal 52:1–8 men. J Clin Endocrinol Metab 87:3023–3028 240. Leloup C, Tourrel-Cuzin C, Magnan C, Karaca M, Castel J, 227. Park SY, Cho YR, Kim HJ, Hong EG, Higashimori T, Lee Carneiro L, Colombani AL, Ktorza A, Casteilla L, Pe´nicaud SJ, Goldberg IJ, Shulman GI, Najjar SM, Kim JK 2006 L 2009 Mitochondrial reactive oxygen species are obligatory Mechanism of glucose intolerance in mice with dominant signals for glucose-induced insulin secretion. Diabetes negative mutation of CEACAM1. Am J Physiol Endocrinol 58:673–681 Metab 291:E517–E524 241. Silva JP, Ko¨ hler M, Graff C, Oldfors A, Magnuson MA, 228. Liu HY, Yehuda-Shnaidman E, Hong T, Han J, Pi J, Liu Z, Berggren PO, Larsson NG 2000 Impaired insulin secretion Cao W 2009 Prolonged exposure to insulin suppresses mi- and ␤-cell loss in tissue-specific knockout mice with mito- tochondrial production in primary hepatocytes. J Biol chondrial diabetes. Nat Genet 26:336–340 Chem 284:14087–14095 242. Gauthier BR, Wiederkehr A, Baquie´ M, Dai C, Powers AC, 229. Cheng Z, Guo S, Copps K, Dong X, Kollipara R, Rodgers Kerr-Conte J, Pattou F, MacDonald RJ, Ferrer J, Wollheim JT, Depinho RA, Puigserver P, White MF 2009 Foxo1 CB 2009 PDX1 deficiency causes mitochondrial dysfunc- integrates insulin signaling with mitochondrial function in tion and defective insulin secretion through TFAM sup- the liver. Nat Med 15:1307–1311 pression. Cell Metab 10:110–118 230. Misu H, Takamura T, Matsuzawa N, Shimizu A, Ota T, 243. Kadowaki T, Kadowaki H, Mori Y, Tobe K, Sakuta R, Sakurai M, Ando H, Arai K, Yamashita T, Honda M, Suzuki Y, Tanabe Y, Sakura H, Awata T, Goto Y, Yamashita T, Kaneko S 2007 Genes involved in oxidative Hayakawa T, Matsuoka K, Kawamori R, Kamada T, Horai phosphorylation are coordinately upregulated with fasting S, Nonaka I, Hagura R, Akanuma Y, Yazaki Y 1994 A sub- hyperglycaemia in livers of patients with type 2 diabetes. type of diabetes mellitus associated with a mutation of mito- Diabetologia 50:268–277 chondrial DNA. N Engl J Med 330:962–968 231. Takamura T, Misu H, Matsuzawa-Nagata N, Sakurai M, 244. Maassen JA, Jahangir Tafrechi RS, Janssen GM, Raap AK, Ota T, Shimizu A, Kurita S, Takeshita Y, Ando H, Honda Lemkes HH, ’t Hart LM 2006 New insights in the molec- M, Kaneko S 2008 Obesity upregulates genes involved in ular pathogenesis of the maternally inherited diabetes and 394 Patti and Corvera Mitochondria and Type 2 DM Endocrine Reviews, June 2010, 31(3):364–395

deafness syndrome. Endocrinol Metab Clin North Am 35: Brownlee M, Corkey BE, Collins S 2009 Persistent oxida- 385–396, x–xi tive stress due to absence of uncoupling protein 2 associ- 245. Tawata M, Hayashi JI, Isobe K, Ohkubo E, Ohtaka M, ated with impaired pancreatic ␤-cell function. Endocrinol- Chen J, Aida K, Onaya T 2000 A new mitochondrial DNA ogy 150:3040–3048 mutation at 14577 T/C is probably a major pathogenic 260. Koshkin V, Wang X, Scherer PE, Chan CB, Wheeler MB mutation for maternally inherited type 2 diabetes. Diabetes 2003 Mitochondrial functional state in clonal pancre- 49:1269–1272 atic ␤-cells exposed to free fatty acids. J Biol Chem 278: 246. Maassen JA, ‘T Hart LM, Van Essen E, Heine RJ, Nijpels 19709–19715 G, Jahangir Tafrechi RS, Raap AK, Janssen GM, Lemkes 261. Benton CR, Nickerson JG, Lally J, Han XX, Holloway GP, HH 2004 Mitochondrial diabetes: molecular mecha- Glatz JF, Luiken JJ, Graham TE, Heikkila JJ, Bonen A nisms and clinical presentation. Diabetes 53(Suppl 1): 2008 Modest PGC-1␣ overexpression in muscle in vivo is S103–S109 sufficient to increase insulin sensitivity and palmitate ox- ␤

247. Maechler P, Carobbio S, Rubi B 2006 In -cells, mito- idation in subsarcolemmal, not intermyofibrillar, mito- Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 chondria integrate and generate metabolic signals control- chondria. J Biol Chem 283:4228–4240 ling insulin secretion. Int J Biochem Cell Biol 38:696–709 262. Liang H, Balas B, Tantiwong P, Dube J, Goodpaster BH, 248. de Andrade PB, Rubi B, Frigerio F, van den Ouweland JM, O’Doherty RM, DeFronzo RA, Richardson A, Musi N, Maassen JA, Maechler P 2006 Diabetes-associated mito- Ward WF 2009 Whole body overexpression of PGC-1␣ chondrial DNA mutation A3243G impairs cellular meta- has opposite effects on hepatic and muscle insulin sensi- bolic pathways necessary for ␤-cell function. Diabetologia tivity. Am J Physiol Endocrinol Metab 296:E945–E954 49:1816–1826 263. Miura S, Kai Y, Ono M, Ezaki O 2003 Overexpression of 249. Hou N, Torii S, Saito N, Hosaka M, Takeuchi T 2008 peroxisome proliferator-activated receptor ␥ Reactive oxygen species-mediated pancreatic ␤-cell death coactivator-1␣ down-regulates GLUT4 mRNA in skeletal is regulated by interactions between stress-activated pro- muscles. J Biol Chem 278:31385–31390 tein , p38 and c-Jun N-terminal kinase, and mito- 264. Miura S, Tomitsuka E, Kamei Y, Yamazaki T, Kai Y, gen-activated protein kinase phosphatases. Endocrinology Tamura M, Kita K, Nishino I, Ezaki O 2006 Overexpres- 149:1654–1665 sion of peroxisome proliferator-activated receptor ␥ 250. Lim M, Park L, Shin G, Hong H, Kang I, Park Y 2008 co-activator-1␣ leads to muscle atrophy with depletion of Induction of apoptosis of ␤-cells of the pancreas by ad- ATP. Am J Pathol 169:1129–1139 vanced glycation end-products, important mediators of 265. Wende AR, Schaeffer PJ, Parker GJ, Zechner C, Han DH, chronic complications of diabetes mellitus. Ann NY Acad Chen MM, Hancock CR, Lehman JJ, Huss JM, McClain Sci 1150:311–315 DA, Holloszy JO, Kelly DP 2007 A role for the transcrip- 251. Newsholme P, Haber EP, Hirabara SM, Rebelato EL, tional coactivator PGC-1␣ in muscle refueling. J Biol Chem Procopio J, Morgan D, Oliveira-Emilio HC, Carpinelli 282:36642–36651 AR, Curi R 2007 Diabetes associated cell stress and dys- 266. Russell LK, Mansfield CM, Lehman JJ, Kovacs A, Courtois function: role of mitochondrial and non-mitochondrial M, Saffitz JE, Medeiros DM, Valencik ML, McDonald JA, ROS production and activity. J Physiol 583:9–24 Kelly DP 2004 Cardiac-specific induction of the transcrip- 252. Nishikawa T, Araki E 2007 Impact of mitochondrial ROS tional coactivator peroxisome proliferator-activated re- production in the pathogenesis of diabetes mellitus and its ceptor ␥ coactivator-1␣ promotes mitochondrial biogen- complications. Antioxid Redox Signal 9:343–353 esis and reversible cardiomyopathy in a developmental 253. Robertson RP 2009 ␤-Cell deterioration during diabetes: stage-dependent manner. Circ Res 94:525–533 what’s in the gun? Trends Endocrinol Metab 20:388–393 267. Calvo JA, Daniels TG, Wang X, Paul A, Lin J, Spiegelman 254. Li N, Frigerio F, Maechler P 2008 The sensitivity of BM, Stevenson SC, Rangwala SM 2008 Muscle-specific pancreatic ␤-cells to mitochondrial injuries triggered by expression of PPAR␥ coactivator-1␣ improves exercise lipotoxicity and oxidative stress. Biochem Soc Trans 36: performance and increases peak oxygen uptake. J Appl 930–934 Physiol 104:1304–1312 255. Rachek LI, Thornley NP, Grishko VI, LeDoux SP, Wilson 268. Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, Boss O, Michael GL 2006 Protection of INS-1 cells from free fatty acid- LF, Puigserver P, Isotani E, Olson EN, Lowell BB, Bassel- induced apoptosis by targeting hOGG1 to mitochondria. Duby R, Spiegelman BM 2002 Transcriptional co-activa- Diabetes 55:1022–1028 tor PGC-1 ␣ drives the formation of slow-twitch muscle 256. Simmons RA, Suponitsky-Kroyter I, Selak MA 2005 Pro- fibers. Nature 418:797–801 gressive accumulation of mitochondrial DNA mutations 269. Sandri M, Lin J, Handschin C, Yang W, Arany ZP, Lecker and decline in mitochondrial function lead to ␤-cell failure. SH, Goldberg AL, Spiegelman BM 2006 PGC-1␣ protects J Biol Chem 280:28785–28791 skeletal muscle from atrophy by suppressing FoxO3 action 257. Brand MD 2005 The efficiency and plasticity of mitochon- and atrophy-specific gene transcription. Proc Natl Acad drial energy transduction. Biochem Soc Trans 33:897–904 Sci USA 103:16260–16265 258. Zhang CY, Baffy G, Perret P, Krauss S, Peroni O, Grujic D, 270. Choi CS, Befroy DE, Codella R, Kim S, Reznick RM, Hagen T, Vidal-Puig AJ, Boss O, Kim YB, Zheng XX, Hwang YJ, Liu ZX, Lee HY, Distefano A, Samuel VT, Wheeler MB, Shulman GI, Chan CB, Lowell BB 2001 Un- Zhang D, Cline GW, Handschin C, Lin J, Petersen KF, coupling protein-2 negatively regulates insulin secretion Spiegelman BM, Shulman GI 2008 Paradoxical effects of and is a major link between obesity, ␤-cell dysfunction, and increased expression of PGC-1␣ on muscle mitochondrial type 2 diabetes. Cell 105:745–755 function and insulin-stimulated muscle glucose metabo- 259. Pi J, Bai Y, Daniel KW, Liu D, Lyght O, Edelstein D, lism. Proc Natl Acad Sci USA 105:19926–19931 Endocrine Reviews, June 2010, 31(3):364–395 edrv.endojournals.org 395

271. Arany Z, Lebrasseur N, Morris C, Smith E, Yang W, Ma 283. Hance N, Ekstrand MI, Trifunovic A 2005 Mitochondrial Y, Chin S, Spiegelman BM 2007 The transcriptional co- DNA polymerase ␥ is essential for mammalian embryo- activator PGC-1␤ drives the formation of oxidative type genesis. Hum Mol Genet 14:1775–1783 IIX fibers in skeletal muscle. Cell Metab 5:35–46 284. Trifunovic A, Wredenberg A, Falkenberg M, Spelbrink JN, 272. Kamei Y, Ohizumi H, Fujitani Y, Nemoto T, Tanaka T, Rovio AT, Bruder CE, Bohlooly-Y M, Gidlo¨ f S, Oldfors A, Takahashi N, Kawada T, Miyoshi M, Ezaki O, Kakizuka Wibom R, To¨ rnell J, Jacobs HT, Larsson NG 2004 Pre- A 2003 PPAR␥ coactivator 1␤/ERR ligand 1 is an ERR mature ageing in mice expressing defective mitochondrial protein ligand, whose expression induces a high-energy DNA polymerase. Nature 429:417–423 expenditure and antagonizes obesity. Proc Natl Acad Sci 285. Tyynismaa H, Sembongi H, Bokori-Brown M, Granycome USA 100:12378–12383 C, Ashley N, Poulton J, Jalanko A, Spelbrink JN, Holt IJ, 273. Espinoza DO, Boros LG, Crunkhorn S, Gami H, Patti ME Suomalainen A 2004 Twinkle helicase is essential for mtDNA 11 November 2009 Dual modulation of both lipid oxida- maintenance and regulates mtDNA copy number. Hum Mol

tion and synthesis by peroxisome proliferator-activated re- Genet 13:3219–3227 Downloaded from https://academic.oup.com/edrv/article/31/3/364/2354793 by guest on 23 September 2021 ␥ ␣ ␤ ceptor- coactivator-1 and -1 in cultured myotubes. 286. Stump CS, Short KR, Bigelow ML, Schimke JM, Nair KS FASEB J doi: 10.1096/fj.09-133728 2003 Effect of insulin on human skeletal muscle mitochon- 274. Pospisilik JA, Knauf C, Joza N, Benit P, Orthofer M, Cani drial ATP production, protein synthesis, and mRNA tran- PD, Ebersberger I, Nakashima T, Sarao R, Neely G, scripts. Proc Natl Acad Sci USA 100:7996–8001 Esterbauer H, Kozlov A, Kahn CR, Kroemer G, Rustin P, 287. Anderson EJ, Kypson AP, Rodriguez E, Anderson CA, Burcelin R, Penninger JM 2007 Targeted deletion of AIF Lehr EJ, Neufer PD 2009 Substrate-specific derangements decreases mitochondrial oxidative phosphorylation and in mitochondrial metabolism and redox balance in the protects from obesity and diabetes. Cell 131:476–491 atrium of the type 2 diabetic human heart. J Am Coll Car- 275. Mootha VK, Handschin C, Arlow D, Xie X, St Pierre J, diol 54:1891–1898 Sihag S, Yang W, Altshuler D, Puigserver P, Patterson N, 288. Bonnard C, Durand A, Peyrol S, Chanseaume E, Chauvin Willy PJ, Schulman IG, Heyman RA, Lander ES, MA, Morio B, Vidal H, Rieusset J 2008 Mitochondrial ␣ Spiegelman BM 2004 ERR and Gabpa/b specify PGC- dysfunction results from oxidative stress in the skeletal 1␣-dependent oxidative phosphorylation gene expression muscle of diet-induced insulin-resistant mice. J Clin Invest that is altered in diabetic muscle. Proc Natl Acad Sci USA 118:789–800 101:6570–6575 289. Yokota T, Kinugawa S, Hirabayashi K, Matsushima S, 276. Soriano FX, Liesa M, Bach D, Chan DC, Palacín M, Zorzano Inoue N, Ohta Y, Hamaguchi S, Sobirin MA, Ono T, Suga A 2006 Evidence for a mitochondrial regulatory pathway T, Kuroda S, Tanaka S, Terasaki F, Okita K, Tsutsui H defined by peroxisome proliferator-activated receptor-␥ co- 2009 Oxidative stress in skeletal muscle impairs mitochon- activator-1 ␣, estrogen-related receptor-␣, and mitofusin 2. drial respiration and limits exercise capacity in type 2 di- Diabetes 55:1783–1791 abetic mice. Am J Physiol Heart Circ Physiol 297:H1069– 277. Luo J, Sladek R, Carrier J, Bader JA, Richard D, Gigue`re V H1077 2003 Reduced fat mass in mice lacking orphan nuclear re- 290. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, ceptor estrogen-related receptor ␣. Mol Cell Biol 23:7947– Lien LF, Haqq AM, Shah SH, Arlotto M, Slentz CA, 7956 278. Liu D, Zhang Z, Teng CT 2005 Estrogen-related recep- Rochon J, Gallup D, Ilkayeva O, Wenner BR, Yancy Jr WS, tor-␥ and peroxisome proliferator-activated receptor-␥ co- Eisenson H, Musante G, Surwit RS, Millington DS, Butler activator-1␣ regulate estrogen-related receptor-␣ gene ex- MD, Svetkey LP 2009 A branched-chain -re- pression via a conserved multi-hormone response element. lated metabolic signature that differentiates obese and lean J Mol Endocrinol 34:473–487 humans and contributes to insulin resistance. Cell Metab 279. Rampersaud E, Damcott CM, Fu M, Shen H, McArdle P, 9:311–326 Shi X, Shelton J, Yin J, Chang YP, Ott SH, Zhang L, Zhao 291. Shah SH, Hauser ER, Bain JR, Muehlbauer MJ, Haynes C, Y, Mitchell BD, O’Connell J, Shuldiner AR 2007 Identifi- Stevens RD, Wenner BR, Dowdy ZE, Granger CB, Ginsburg cation of novel candidate genes for type 2 diabetes from a GS, Newgard CB, Kraus WE 2009 High heritability of genome-wide association scan in the Old Order Amish: evi- metabolomic profiles in families burdened with premature dence for replication from diabetes-related quantitative traits cardiovascular disease. Mol Syst Biol 5:258 and from independent populations. Diabetes 56:3053–3062 292. Lerin C, Goldfine AB, Tatro E, Boes T, Schroeder J, Patti 280. Freyer C, Larsson NG 2007 Is energy deficiency good in ME 2009 Reductions in branched chain amino acid oxi- moderation? Cell 131:448–450 dation gene expression is a signature of insulin resistance in 281. Larsson NG, Wang J, Wilhelmsson H, Oldfors A, Rustin humans. Diabetes 58:A298 (Supplement) P, Lewandoski M, Barsh GS, Clayton DA 1998 Mito- 293. Shaham O, Wei R, Wang TJ, Ricciardi C, Lewis GD, Vasan chondrial transcription factor A is necessary for mtDNA RS, Carr SA, Thadhani R, Gerszten RE, Mootha VK 2008 maintenance and embryogenesis in mice. Nat Genet 18: Metabolic profiling of the human response to a glucose 231–236 challenge reveals distinct axes of insulin sensitivity. Mol 282. Wang J, Wilhelmsson H, Graff C, Li H, Oldfors A, Rustin Syst Biol 4:214 P, Bru¨ ning JC, Kahn CR, Clayton DA, Barsh GS, Thore´n 294. Bajaj M, Medina-Navarro R, Suraamornkul S, Meyer C, P, Larsson NG 1999 Dilated cardiomyopathy and atrio- DeFronzo RA, Mandarino LJ 2007 Paradoxical changes in ventricular conduction blocks induced by heart-specific in- muscle gene expression in insulin-resistant subjects after activation of mitochondrial DNA gene expression. Nat sustained reduction in plasma free fatty acid concentra- Genet 21:133–137 tion. Diabetes 56:743–752