Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Annals of Medicine

ISSN: 0785-3890 (Print) 1365-2060 (Online) Journal homepage: www.tandfonline.com/journals/iann20

Glitazones: clinical effects and molecular


mechanisms

Michael Stumvoll & Hans-Ulrich Häring

To cite this article: Michael Stumvoll & Hans-Ulrich Häring (2002) Glitazones: clinical effects
and molecular mechanisms, Annals of Medicine, 34:3, 217-224, DOI: 10.1080/ann.34.3.217.224

To link to this article: https://doi.org/10.1080/ann.34.3.217.224

Published online: 08 Jul 2009.

Submit your article to this journal

Article views: 910

View related articles

Citing articles: 10 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iann20
^ WHAT IS NEW IN MOLECULAR ENDOCRINOLOGY? ^

Glitazones: clinical effects and molecular


mechanisms
Michael Stumvoll and Hans-Ulrich HaÈring

With the thiazolidinediones rosiglitazone and pioglitazone a with the thiazolidinediones a novel concept for the
novel treatment modality for type 2 diabetes has become treatment of insulin resistance is available that in theory
available in many countries. As monotherapy, fasting blood could also be used for prevention of type 2 diabetes. Long-
glucose and glycosylated hemoglobin (HbA1c), on average, term data are indispensable for a ®nal risk-bene®t assess-
can be improved by approximately 40 mg/dl and almost 1%, ment of these substances.
respectively. In combination with other agents their ef®cacy
Keywords: adipose tissue; insulin resistance; obesity; pioglitazone; peroxisome
is additive. Thiazolidinediones reduce insulin resistance not
proliferator-activated receptor; rosiglitazone; type 2 diabetes.
only in type 2 diabetes but also in non-diabetic conditions
associated with insulin resistance such as obesity. The Ann Med 2002; 34: 217±224
mechanism of action involves binding to the peroxisome
(PPAR)g, a transcription
proliferator-activated receptor (PPAR)g
factor that regulates the expression of speci®c genes Insulin resistance
especially in fat cells but also in other tissues. It is likely
that thiazolidinediones primarily act in adipose tissue where Insulin resistance is said to be present in an individual
PPARg is predominantly expressed. Thiazolidinediones have
PPARg when the biological effects of insulin are less than
been shown to interfere with expression and release of expected. The tissues through which alterations in
mediators of insulin resistance originating in adipose tissue insulin sensitivity immediately in¯uence glucose
(e.g. free fatty acids, adipocytokines such as tumor necrosis homeostasis are muscle (glucose disposal), liver and
factor a, resistin, adiponectin) in a way that results in net to a minor extent kidney (suppression of endogenous
improvement of insulin sensitivity (i.e. in muscle and liver). glucose production). Thus, the two resulting abnorm-
Nevertheless, a direct molecular effect in skeletal muscle alities consistently observed in glucose-intolerant
cannot be excluded. Interference with transcription entails a states are increased rates of glucose production and
potential for side-effect risk, that cannot de®nitively be decreased peripheral glucose disposal (1, 2). Further-
assessed yet. For example, the in-vitro stimulation of more, suppression of lipolysis in adipose tissue is
adipogenic differentiation may underlie the clinical ob- highly sensitive to insulin. In patients with type 2
servation of weight gain. Theoretically, this may turn out to diabetes impaired insulin suppression of lipolysis has
be counterproductive in the long run. However, there is not been reported (3). A novel concept recently emerging
suf®cient evidence from humans at the moment, especially from knockout animals also suggests a pathophysio-
no long-term data, to allow a conclusive statement. The logical role of insulin resistance of the central nervous
hepatotoxicity observed with troglitazone, on the other system (4) (Fig. 1).
PPARg-mediated but secondary
hand, does not seem to be PPARg Besides disturbing glucose homeostasis, insulin
to toxic metabolites. Based on differences in drug metab- resistance contributes to the pathogenesis of other
olism this problem is relatively unlikely to occur with clinically relevant disorders. Hypertension associated
rosiglitazone or pioglitazone. Unexplained but not unim- with insulin resistance appears to be a consequence,
portant is the propensity for ¯uid retention. In summary, among many other causes, also of the impaired
vasodilating effect of insulin (5). Hepatic insulin
resistance contributes to dyslipidemias characterized
by decreased high-density lipoprotein (HDL) concen-
From the Medizinische Klinik, Abteilung fuÈr Endokrinologie,
Stoffwechsel und Pathobiochemie, Eberhard-Karls-UniversitaÈt,
trations and increased low-density lipoprotein (LDL)
TuÈbingen, Germany. (6). (Furthermore, recent results suggest that insulin
Correspondence: Dr. Michael Stumvoll, Medizinische Univer- resistance is also associated with increased aggreg-
sitaÈtsklinik, Otfried-MuÈller-Str. 10, DE-72076 TuÈbingen, Germany. ability of platelets (7) and with endothelial dysfunc-
Fax: +49-7071-295277, Email: michael.stumvoll@med.uni- tion (8). Thus, insulin resistance can be made
tuebingen.de
responsible, at least in part, for the pro-arteriosclero-

# 2002 Taylor & Francis. ISSN 0785-3890 Annals of Medicine 34


218 STUMVOLL . HAÈRING

Key messages
. With the thiazolidinediones rosiglitazone and
pioglitazone, a novel treatment modality for
type 2 diabetes has become available.
. The mechanism of action of these compounds

involves binding to the peroxisome proliferator-


activated receptor (PPAR) gamma , a transcrip-
tion factor that regulates the expression of
speci®c genes especially in fat cells but also in
other tissues.
. As monotherapy, glycosylated hemoglobin
Figure 1. Insulin secretion and resistance. Organ manifesta- (HbA1c) on average can be improved by almost
tions of insulin resistance and metabolic consequences: adipose
tissue — increased lipolysis and excessive FFA release; liver (and
1%.
. Thiazolidinediones reduce insulin resistance
kidney) — increased glucose production; muscle — decreased
glucose uptake. The role of insulin resistance in brain is theoretical not only in type 2 diabetes but also in non-
at present since evidence is available only from a brain-specific
diabetic conditions associated with insulin
insulin receptor knockout mouse (4).
resistance such as obesity.

tic milieu of hyperglycemia, arterial hypertension,


dyslipidemia and hypercoagulability seen in type 2
diabetes and related disorders. It therefore represents fact that local levels of TNFa bound to its circulating
a highly rewarding target for pharmacological inter- receptor correlates with the degree of adiposity in
vention. humans (18) suggests that paracrine mechanisms may
be operative. Studies in cell models suggest that
molecular mechanisms of TNFa-induced insulin
Role of adipose tissue and obesity resistance involve activation of inhibitory serine
kinases (19) or tyrosine phosphatases which lead to
The striking association between insulin resistance inhibition of the insulin signaling pathway (20, 21).
and obesity (9) produces the important question Ð Resistin, a protein that is increasingly released from
which factors originating from adipose tissue are white adipocytes during differentiation was recently
responsible and have the potential for pharmacologi- identi®ed. In rodent models of obesity resistin mRNA
cal intervention? In addition to free fatty acids (FFAs) is increased in adipose tissue. Intravenous adminis-
whose role is ®rmly established (10), advances in tration of the recombinant protein causes glucose
molecular biology and genomic research have facili- intolerance and insulin resistance in mice. On the
tated the identi®cation of peptides released from other hand, neutralizing antibodies reduce insulin
adipocytes (so-called adipocytokines) that potentially resistance and hyperglycemia in a mouse model with
in¯uence insulin sensitivity. The ob gene product dietary obesity and type 2 diabetes (22). Although
leptin whose primary function is to signal replenished these data appear promising, evidence for the applic-
energy stores from the adipocyte to the hypothalamus ability to the human situation is still pending. At
(11) has been shown to interfere with insulin signaling present, it is actually unclear whether a human
and action in cell models (12±14). Tumor necrosis resistin homologue is suf®ciently expressed in adipose
factor-a (TNFa) is a cytokine that is primarily tissue and circulates in human plasma.
produced in macrophages and involved in host Recently, a novel adipocyte-derived hormone,
defense. Since fat tissue is also a signi®cant source adiponectin was identi®ed and characterized (23,
of this cytokine and TNFa expression in adipose 24). With a concentration of around 5 mg/ml in
tissue is elevated in obesity (15), it represents a human plasma, adiponectin represents one of the
reasonable candidate for linking obesity to insulin most abundant circulating proteins. Interestingly, in
resistance. However, evidence from euglycemic obesity, a condition highly associated with insulin
clamps showing an effect of circulating TNFa on resistance, plasma adiponectin levels are signi®cantly
insulin sensitivity independent of other factors decreased (25, 26). Adiponectin concentrations are
released from adipose tissue is lacking. While in positively correlated with insulin sensitivity, decrease
obese rats neutralization of TNFa resulted in signi®cantly with deteriorating glucose tolerance (25)
increased insulin sensitivity (16) this was not the case and increase after weight reduction (27). Further-
in patients with type 2 diabetes (17). Nevertheless, the more, intravenous administration of recombinant

# 2002 Taylor & Francis. ISSN 0785-3890 Annals of Medicine 34


GLITAZONES 219

fasting insulin levels ranging from 2 to 10 mU/ml


(35, 36). The alterations in glycemic parameters are
strongly suggestive of an improvement in insulin
sensitivity in patients with type 2 diabetes. Taking
into account differences in study design (pretreatment
glycemic control, duration, dose) rosiglitazone and
pioglitazone are similarly ef®cacious (1). To date no
study has directly compared the clinical ef®cacy of
two or more thiazolidinediones. The effect on
glycemic control was dose dependent and leveled off
with daily doses >600 mg for troglitazone, >8 mg for
rosiglitazone and >30 mg for pioglitazone (35±37).
Overall, the improvement in glycemic control using a
single agent appears to be less pronounced with
thiazolidinediones than with sulfonylureas or metfor-
min.
Interestingly, other abnormalities commonly asso-
Figure 2. Chemical structure of thiazolidinediones and ciated with insulin resistance, such as dyslipidemia
vitamin E. and arterial hypertension, also appeared to be
improved by thiazolidinediones (35, 38, 39). While
rosiglitazone did not alter the LDL-HDL ratio (35),
adiponectin to rodent models of insulin resistance pioglitazone treatment reduced LDL-cholesterol and
restored normal insulin sensitivity (28). Thus, in increased HDL-cholesterol (36). No signi®cant effect
contrast to previously discussed adipocytokines, on triglyceride levels was reported for either of the
adiponectin seems to protect from insulin resistance compounds. Troglitazone, which unlike other thia-
and type 2 diabetes. zolidinediones carries an antioxidant vitamin E
moiety, in healthy volunteers also reduced LDL lipid
hydroperoxides (40) which are thought to be of
Thiazolidinediones Ð historical aspects and particular atherogenic potential (41). Other reported
clinical ef®cacy non-hypoglycemic effects of thiazolidinedines include
improvement of ®brinolysis and decrease in carotid
For a long time, only biguanides such as metformin, artery intima media thickness (reviewed in (42)).
have been used for the treatment of insulin resistance.
It was not until 1997, when the thiazolidinediones (=
`glitazones’) also became available for treating insulin Mechanism of action
resistance. They are a new class of agents which were
Metabolic studies
originally developed in the early 1980s in Japan as
antioxidants (29). Soon after the synthesis of the ®rst The concept, that a decrease in blood sugar, accom-
thiazolidinedione, ciglitazone, the blood glucose low- panied by a decrease in insulin concentrations,
ering potential of these compounds was observed in represents improvement of insulin sensitivity, was
animals with particularly pronounced effects in systematically examined in metabolic studies in vivo.
animals with genetic insulin resistance such as KK, Thus, a 25% decrease in blood sugar and 30% lower
db/db and ob/ob mice and fa/fa rats (30±32). The insulin could be shown during a meal-tolerance-test
observation that glycemia improved in the absence of and an oral glucose tolerance test in patients with
increasing insulin and the lack of effect in insulin type-2-diabetes and patients with impaired glucose
de®cient animals (29, 33) led to the conclusion that tolerance after treatment with troglitazone (43, 44).
thiazolidinediones improved insulin resistance, hence For pioglitazone a signi®cant improvement of the
the nickname `insulin sensitizers’. Due to an unto- insulin sensitivity could also be observed in a clamp
ward risk-bene®t ratio (hepatotoxic side effects), experiment in patients with type-2-diabetes (45). In
however, troglitazone had to be withdrawn from non-diabetic, adipose patients treated with troglita-
the market and currently the newer compounds zone an improvement of insulin sensitivity of about
rosiglitazone and pioglitazone are the two thiazolidi- 80% was measured with the hyperinsulinemic-eugly-
nediones available in most countries (34) (Fig. 2). cemic clamp in the absence of changes in fasting
With thiazolidinedione treatment an average glucose (46). The pathologically increased endogen-
improvement of fasting plasma glucose levels by ous glucose production in type 2 diabetic patients was
about 40 mg/dl was observed in patients with type 2 reduced after therapy with troglitazone to near-
diabetes accompanied by a signi®cant reduction in normal rates (43). This indicates improved effective-

# 2002 Taylor & Francis. ISSN 0785-3890 Annals of Medicine 34


220 STUMVOLL . HAÈRING

Figure 4. Cellular mechanism of action of thiazolidine-


Figure 3. Family of PPARs and ligand specificity. Font size diones. TZD = thiazolidinedione; Ins = Insulin; IR = insulin recep-
reflects expression levels in specific tissues. tor; RXR = retinoid X receptor; see text for details.

ness of insulin also in glucose producing tissues


expressed in fat cells makes it somewhat dif®cult to
(mainly liver). However, this effect seems to be less
reconcile cellular and metabolic effects. In addition,
pronounced in humans (47) compared to animal
considering the well known connection between
models (48, 49).
obesity and insulin resistance it seems paradoxical
that an agent which promotes adipogenesis should
improve insulin sensitivity. A number of hypothetical
In-vitro studies
schemas to explain the available data have been put
The cellular mechanism of action of the thiazolidine- forward. Firstly, PPARg is also expressed in skeletal
diones is mediated by binding to and activating the muscle (55) and the minute quantities may be
peroxisomal proliferator-activated receptor (PPAR)g. suf®cient to mediate the thiazolidinedione effect
PPARg is a nuclear receptor that acts as transcription directly. It is unclear what these muscle-speci®c
factor upon activation by regulating the transcription effects could be but in a fat-cell-free primary human
and expression of speci®c genes. Together with the muscle cell culture system troglitazone enhanced
isoforms PPARa and PPAR¯ (Fig. 3) it belongs to the insulin-stimulated glycogen synthesis and phosphati-
same superfamily as thyroid hormone and steroid dyl-inositol-3 kinase activity (16, 56). Direct effects in
receptors (50). Expression levels of the nuclear muscle are also supported by in-vivo observations in a
receptor PPARg are highest in adipocytes, intestinal lipoatrophic mouse model where thiazolidinedione
cells and macrophages but very low in most other treatment improved insulin sensitivity in the absence
tissues including muscle. Endogenous ligands of or near absence of adipose tissue (57).
PPARg include long-chain unsaturated fatty acids Secondly, the effect of thiazolidinediones on insulin
and prostanoids such as 15-deoxy-delta12, 14-prosta- stimulated glucose disposal may indeed be secondary
glandin J2 (51). Upon activation, PPARg heterodi- to changes in adipose tissue where PPARg is pre-
merizes with the retinoid X receptor (itself activated dominantly expressed and involve factors which alter
by speci®c ligands) and the activated complex subse- peripheral insulin sensitivity. Thiazolidinediones have
quently binds to speci®c DNA segments to induce been shown to selectively stimulate lipogenic activities
transcription of PPAR-responsive elements (PPRE) in fat cells (58) resulting in greater insulin suppression
(®g. 4). In various cell models (preadipocytes, of lipolysis (59). This would leave less FFAs available
®broblasts, myoblasts) thiazolidinedione treatment for other tissues (12). Thus, insulin desensitizing
resulted in expression of a number of adipocyte effects of FFAs in muscle and liver (10) would be
speci®c genes (lipoprotein lipase, fatty-acid binding reduced as a consequence of thiazolidinedione treat-
protein, glucose free fatty acids (GLUT4), acylco- ment. This was originally proposed as `fatty-acid-steal
enzyme A (CoA) synthetase, etc.) so that PPARg phenomenon’ (60) and recently con®rmed in humans
activation and/or overexpression has essentially been (61). Moreover, thiazolidinediones have been shown
associated with adipogenic differentiation (52±54). to alter expression and release of adipocytokines.
The clinical observations that treatment with Resistin, TNFa and leptin which have the potential to
thiazolidinediones improves insulin-stimulated (i.e. decrease insulin sensitivity (see above) are reduced
muscle) glucose uptake while PPARg is mainly following incubation with thiazolidinediones (22, 62±

# 2002 Taylor & Francis. ISSN 0785-3890 Annals of Medicine 34


GLITAZONES 221

ing of fat depots away from metabolically unfavor-


able (i.e. visceral) to more innocent sites (i.e.
subcutaneous) has been con®rmed in humans by
computer tomography (75±78).
Nevertheless, novel concerns regarding the poten-
tial of these agents to affect cellular differentiation
originated from the observation that PPARg-activa-
tion causes fatty transformation of bone marrow
stromal cells (79). Furthermore, PPARg was recently
shown to be involved in differentiation of monocytes
into macrophages and uptake of oxidized LDL by
macrophages/monocytes, suggesting that PPARg is
an important regulator of gene expression during
athero-genesis (80). On the other hand, transcrip-
Figure 5. Interference of thiazolidinediones with factors
tional alterations with the potential to prevent
released from adipose tissue. see text for details.
atherogenesis and in¯ammation have also been
reported (81). Moreover, in mice PPARg-agonists
were shown to promote carcinomatous growth in
64). Conversely, expression and release of adiponectin colon epithelium (82, 83) while human colonic cancer
is increased upon thiazolidinedione treatment (58, 65). cells transplanted into mice showed a signi®cant
Thus, all adipocytokines implicated in insulin sensi- growth retardation following treatment with thiazo-
tivity are altered in a way that would ultimately lidinediones (84). Conversely, PPARg-agonists have
improve insulin sensitivity. Which of these mechan- been shown to inhibit growth of a human monocytic
isms plays the most important role in vivo is unclear at leukemia cell line (85) setting the stage for a novel
present but since they are not mutually exclusive all of treatment concept of malignant diseases. However, at
them may be involved (Fig. 5). the present moment lack of clinical data makes it
impossible to determine the human relevance for any
of these mechanisms.
Side effects

In the early studies with troglitazone the thiazoli- Conclusion


dinediones appeared to be well tolerated and main
adverse events included ¯uid retention and reversible Thiazolidinediones represent a novel pharmacologi-
reductions in hemoglobin, hematocrit and neutro- cal principle in the treatment of type 2 diabetes. With
phil counts. The liver toxicity that resulted in respect to both mechanism of action and clinical
withdrawal of troglitazone from most markets does ef®cacy they are complementary or additive to
not seem to be a problem with rosiglitazone or metformin and sulfonylureas. Evidently, thiazolidi-
pioglitazone (66) apart from anecdotal reports (67± nediones improve insulin sensitivity not only of type 2
69). This difference is probably the result of different diabetes but also of primarily non-diabetic conditions
metabolic degradation by the cytochrome P450 such as: polycystic ovary syndrome (72), Werner
enzyme complex (70) and the vitamin E moiety of syndrome (genetic disease characterized by acceler-
troglitazone (71). ated aging) (86) and simple obesity (46). Theoreti-
The adipogenic potential of thiazolidinediones in cally, this opens up the possibility of pharmacological
preadipocytes in vitro and in therapeutic doses in prevention of type 2 diabetes and associated diseases.
animals in vivo (72) has been of some concern. The experimental veri®cation of this hypothesis on a
Clinical use also demonstrated signi®cant weight large scale by the American diabetes prevention trial
gain (73). The fact that an agent causes an increase in (87) had to be aborted following a fatal case of liver-
body fat and at the same time improves insulin toxicity in the troglitazone arm.
sensitivity cannot easily be reconciled. It has been The mechanism of action involves binding to the
suggested that preadipocytes in adult humans could peroxisome proliferator-activated receptor (PPAR)g,
be relatively resistant to the adipogenic effect of a transcription factor with pleiotropic functions that
thiazolidinediones or, alternatively, that increased regulates the expression of speci®c genes especially in
adipogenesis per se need not necessarily cause fat cells but also in other tissues. It is likely that
obesity (53). It is also of note that thiazolidinediones thiazolidinediones primarily act in adipose tissue
preferentially induce terminal differentiation of where PPARg is predominantly expressed. Thiazoli-
preadipocytes of subcutaneous origin but not of dinediones have been shown to interfere with expres-
preadipocytes of visceral origin (74). This remodel- sion and release of mediators between adipose tissue

# 2002 Taylor & Francis. ISSN 0785-3890 Annals of Medicine 34


222 STUMVOLL . HAÈRING

and insulin resistance (e.g. free fatty acids, adipocy- with the newer agents, weight gain and ¯uid retention
tokines such as tumor necrosis factor a, resistin, are probably class effects. Outside the diabetes and
adiponectin) in a way that ultimately improves insulin metabolism ®eld, thiazolidinediones may become a
action. Nevertheless, a direct effect in skeletal muscle novel therapeutic approach for selected forms of
cannot be completely excluded. cancer.
Interference with transcription contains a potential Although currently only rosiglitazone and pio-
risk for side effects, whose clinical relevance cannot glitazone are available for clinical use a number of
yet be de®nitively assessed. While the hepatotoxicity interesting newer thiazolidinediones or related com-
observed with troglitazone does not seem to occur pounds are in development (88±91).

References

1. Matthaei S, Stumvoll M, Kellerer M, Häring HU. Pathoph- insulin sensitivity and glycemic control in patients with
siology and pharmacological treatment of insulin resistance. NIDDM. Diabetes 1996; 45: 881±5.
Endocr Rev 2000; 21: 585±618. 18. Mohamed-Ali V, Goodrick S, Bulmer K, Holly JM, Yudkin
2. Dinneen S, Gerich J, Rizza R. Carbohydrate metabolism in JS, Coppack SW. Production of soluble tumor necrosis factor
non-insulin-dependent diabetes mellitus. N Engl J Med 1992; receptors by human subcutaneous adipose tissue in vivo. Am J
327: 707±13. Physiol Endocrinol Metab 1999; 277: E971±5.
3. Groop LC, Bonadonna RC, DelPrato S, Ratheiser K, 19. Rui L, Aguirre V, Kim JK, Shulman GI, Lee A, Corbould A,
Zyck K, Ferrannini E, et al. Glucose and free fatty acid et al. Insulin/IGF-1 and TNF-alpha stimulate phosphorylation
metabolism in non-insulin-dependent diabetes mellitus. Evi- of IRS-1 at inhibitory Ser307 via distinct pathways. J Clin
dence for multiple sites of insulin resistance. J Clin Invest 1989; Invest 2001; 107: 181±9.
84: 205±13. 20. Kroder G, Bossenmaier B, Kellerer M, Capp E, Stoyanov B,
4. Bruning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Muhlhofer A, et al. Tumor necrosis factor-alpha- and
Orban PC, et al. Role of brain insulin receptor in control of hyperglycemia-induced insulin resistance. Evidence for differ-
body weight and reproduction. Science 2000; 289: 2122±5. ent mechanisms and different effects on insulin signaling. J
5. Hsueh W, Law RE, Saad M, Dy J, Feener E, King G. Insulin Clin Invest 1996; 97: 1471±7.
resistance and macrovascular disease. Curr Opin Endocrinol 21. Hotamisligil GS, Murray DL, Choy LN, Spiegelman BM.
Diab 1996; 3: 346±54. Tumor necrosis factor alpha inhibits signaling from the insulin
6. Taskinen M-R. Hyperlipidaemia in diabetes. Baillieres Best receptor. Proc Natl Acad Sci USA 1994; 91: 4854±8.
Pract Res Clin Endocrinol Metab 1990; 4: 743±75. 22. Steppan CM, Bailey ST, Bhat S, Brown EJ, Baernee RR,
7. Trovati M, Anfossi G. Insulin, insulin resistance and platelet Wright CM, et al. The hormone resistin links obesity to
function: similarities with insulin effects on cultured vascular diabetes. Nature 2001; 409: 307±12.
smooth muscle cells. Diabetologia 1998; 41: 609±22. 23. Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-
8. Balletshofer BM, Rittig K, Enderle MD, Volk A, Maerker speci®c gene dysregulated in obesity. J Biol Chem 1996; 271:
E, Jacob S, et al. Endothelial dysfunction is detectable in 10697±703.
young normotensive ®rst-degree relatives of subjects with type 24. Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF.
2 diabetes in association with insulin resistance. Circulation A novel serum protein similar to C1q, produced exclusively in
2000; 101: 1780±4. adipocytes. J Biol Chem 1995; 270: 26746±9.
9. Must A, Spadano J, Coakley EH, Field AE, Colditz G, Dietz 25. Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y,
WH. The disease burden associated with overweight and Pratley RE, et al. Hypoadiponectinemia in obesity and type 2
obesity. JAMA 1999; 282: 1523±9. diabetes: close association with insulin resistance and hyper-
10. Boden G. Role of fatty acids in the pathogenesis of insulin insulinemia. J Clin Endocrinol Metab 2001; 86: 1930±5.
resistance and NIDDM. Diabetes 1997; 46: 3±10. 26. Takahashi M, Arita Y, Yamagata K, Matsukawa Y, Oku-
11. Friedman JM, Halaas JL. Leptin and the regulation of body tomi K, Horie M, et al. Genomic structure and mutations in
weight in mammals. Nature 1998; 395: 763±70. adipose-speci®c gene, adiponectin. Int J Obes Relat Metab
12. Berti L, Kellerer M, Capp E, Häring HU. Leptin stimulates Disord 2000; 24: 861±8.
glucose transport and glycogen synthesis in C2C12 myotubes: 27. Yang WS, Lee WJ, Funahashi T, Tanaka S, Matsuzawa, Y,
evidence for a P13-kinase mediated effect. Diabetologia 1997; Chao CL, et al. Weight reduction increases plasma levels of an
40: 606±9. adipose-derived anti-in¯ammatory protein, adiponectin. J Clin
13. Cohen B, Novick D, Rubinstein M. Modulation of insulin Endocrinol Metab 2001; 86: 3815±9.
activities by leptin. Science 1996; 274: 1185±8. 28. Yamauchi T, Kamon J, Waki H, Terauchi K, Kubota N,
14. Sempoux C, Guiot Y, Dubois D, Moulin P, Rahier J. Human Hara K, et al. The fat-derived hormone adiponectin reverses
type 2 diabetes: morphological evidence for abnormal beta-cell insulin resistance associated with both lipoatrophy and
function. Diabetes 2001; 50 Suppl 1: S172±7. obesity. Nat Med 2001; 7: 941±6.
15. Hotamisligil GS, Arner P, Atkinson RL, Spiegelman BM. 29. Fujita T, Sugiyama Y, Taketomi S, Sohda T, Kawamatsu Y,
Differential regulation of the p80 tumor necrosis factor Iwatsuka H, et al. Reduction of insulin resistance in obese
receptor in human obesity and insulin resistance. Diabetes and/or diabetic animals by 5-[4-(1-methylcyclohexylmethoxy)-
1997; 46: 451±5. benzyl]-thiazolidine-2, 4-dione (ADD-3878, U-63, 287, ciglita-
16. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose zone), a new antidiabetic agent. Diabetes 1983; 32: 804±10.
expression of tumor necrosis factor-alpha: direct role in 30. Fujiwara T, Okuno A, Yoshioka S, Horikoshi H. Suppres-
obesity-linked insulin resistance. Science 1993; 259: 87±91. sion of hepatic gluconeogenesis in long-term Troglitazone
17. Ofei F, Hurel S, Newkirk J, Sopwith M, Taylor R. Effects of treated diabetic KK and C57BL/KsJ-db/db mice. Metabolism
an engineered human anti-TNF-alpha antibody (CDP571) on 1995; 44: 486±90.

# 2002 Taylor & Francis. ISSN 0785-3890 Annals of Medicine 34


GLITAZONES 223

31. Stevenson RW, Hutson NJ, Krupp MN, Volkmann RA, double-blind, placebo-controlled trial. Ann Intern Med 1998;
Holland GF, Eggler JF, et al. Actions of novel antidiabetic 128: 176±85.
agent englitazone in hyperglycemic hyperinsulinemic ob/ob 48. Tominaga M, Igarashi M, Daimon M, Eguchi H, Matsu-
mice. Diabetes 1990; 39: 1218±27. moto M, Sekikawa A, et al. Thiazolidinediones (AD-4833
32. Kirsch DM, Bachmann W, Häring HU. Ciglitazone reverses and CS-045) improve hepatic insulin resistance in streptozo-
cAMP-induced post-insulin receptor resistance in rat adipo- tocin-induced diabetic rats. Endocr J 1993; 40: 343±9.
cytes. FEBS Lett 1984; 176: 49±54. 49. Lee MK, Olefsky JM. Acute effects of troglitazone on in vivo
33. Fujiwara T, Wada M, Fukuda K, Fukami M, Yoshioka S, insulin action in normal rats. Metabolism 1995; 44: 1166±9.
Yoshioka T, et al. Characterization of CS-045, a new oral 50. Desvergne B, Wahli W. Peroxisome proliferator-activated
antidiabetic agent, II. Effects on glycemic control and receptors: nuclear control of metabolism. Endocr Rev 1999; 20:
pancreatic islet structure at a late stage of the diabetic 649±88.
syndrome in C57BL/KsJ-db/db mice. Metabolism 1991; 40: 51. Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman
1213±8. BM, Evans RM. 15-Deoxy-delta 12, 14-prostaglandin J2 is a
34. Gale EA. Lessons from the glitazones: a story of drug ligand for the adipocyte determination factor PPAR gamma.
development. Lancet 2001; 357: 1870±5. Cell 1995; 83: 803±12.
35. Raskin P, Rendell M, Riddle MC, Dole JF, Freed MI, 52. Tafuri SR. Troglitazone enhances differentiation, basal
Rosenstock J. A randomized trial of rosiglitazone therapy in glucose uptake, and Glut1 protein levels in 3T3-L1 adipocytes.
patients with inadequately controlled insulin-treated type 2 Endocrinology 1996; 137: 4706±12.
diabetes. Diabetes Care 2001; 24: 1226±32. 53. Teboul L, Gaillard D, Staccini L, Inadera H, Amri EZ,
36. Aronoff S, Rosenblatt S, Braithwaite S, Egan JW, Mathisen Grimaldi PA. Thiazolidinediones and fatty acids convert
AL, Schneider RL. Pioglitazone hydrochloride monotherapy myogenic cells into adipose-like cells. J Biol Chem 1995; 270:
improves glycemic control in the treatment of patients with 28183±7.
type 2 diabetes: a 6-month randomized placebo-controlled 54. Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipo-
dose-response study. The Pioglitazone 001 Study Group. genesis in ®broblasts by PPAR gamma 2, a lipid-activated
Diabetes Care 2000; 23: 1605±11. transcription factor. Cell 1994; 79: 1147±56.
37. Horton ES, Whitehouse F, Ghazzi MN, Venable TC, 55. Loviscach M, Rehman N, Carter L, Mudaliar S, Mohadeen
Whitcomb RW. Troglitazone in combination with sulfony- P, Ciaraldi TP, et al. Distribution of peroxisome proliferator-
lurea restores glycemic control in patients with type 2 diabetes. activated receptors (PPARs) in human skeletal muscle and
Diabetes Care 1998; 21: 1462±9. adipose tissue: relation to insulin action. Diabetologia 2000;
38. Wang M, Wise SC, Leff T, Su T-Z. Troglitazone, an 43: 304±11.
antidiabetic agent, inhibits cholesterol biosynthesis through a 56. Park KS, Ciaraldi TP, Abrams Carter L, Mudaliar S,
mechanism independent of peroxisome proliferator-activated Nikoulina SE, Henry RR. Troglitazone regulation of glucose
receptor-gamma. Diabetes 1999; 48: 254±60. metabolism in human skeletal muscle cultures from obese type
39. Buchanan TA, Meehan WP, Jeng YY, Yang D, Chan TM, II diabetic subjects. J Clin Endocrinol Metab 1998; 83: 1636±
Nadler JL, et al. Blood pressure lowering by pioglitazone. 43.
Evidence for a direct vascular effect. J Clin Invest 1995; 96: 57. Burant CF, Sreenan S, Hirano K, Tai TA, Lohmiller J,
354±60. Lukens J, et al. Troglitazone action is independent of adipose
40. Cominacini L, Young MMR, Capriati A, Garbin U, Fratta tissue. J Clin Invest 1997; 100: 2900±8.
Pasini A, Campagnola M, et al. Troglitazone increases the 58. Yamauchi T, Kamon J, Waki H, Murakami K, Motojima K,
resistance of low densitiy lipoprotein to oxidation in healthy Komeda K, et al. The mechanisms by which both hetero-
volunteers. Diabetologia 1997; 40: 1211±8. zygous PPARgamma de®ciency and PPARgamma agonist
41. Steinberg D, Parthasarath y S, Carew TE, Khoo JC, improve insulin resistance. J Biol Chem 2001; 276: 41245±54.
Witztum JL. Beyond cholesterol. Modi®cations of low-density 59. Oakes ND, Thalen PG, Jacinto SM, Ljung B. Thiazolidine-
lipoprotein that increase its atherogenicity. N Engl J Med diones increase plasma-adipose tissue FFA exchange capacity
1989; 320: 915±24. and enhance insulin-mediated control of systemic FFA avail-
42. Porte D, Jr., Kahn SE. beta-cell dysfunction and failure in type ability. Diabetes 2001; 50: 1158±65.
2 diabetes: potential mechanisms. Diabetes 2001; 50 Suppl 1: 60. Schoonjans K, Martin G, Staels B, Auwerx J. Peroxisome
S160±3. proliferator-activated receptors, orphans with ligands and
43. Suter SL, Nolan JJ, Wallace P, Gumbiner B, Olefsky JM. functions. Curr Opin Lipidol 1997; 8: 159±66.
Metabolic effects of new oral hypoglycemic agent CS-045 in 61. Miyazaki Y, Glass L, Triplitt C, Matsuda M, Cusi K,
NIDDM subjects. Diabetes Care 1992; 15: 193±203. Mahankali A, et al. Effect of rosiglitazone on glucose and
44. Antonucci T, Whitcomb R, McLain R, Lockwood D. non-esteri®ed fatty acid metabolism in Type II diabetic
Impaired glucose tolerance is normalized by treatment with patients. Diabetologia 2001; 44: 2210±9.
the thiazolidinedione troglitazone. Diabetes Care 1997; 20: 62. Hofmann C, Lorenz K, Braithwaite SS, Colca JR, Palazuk
188±93. BJ, Hotamisligil GS, et al. Altered gene expression for tumor
45. Kawamori R, Matsuhisa M, Kinoshita J, Mochizuki K, necrosis factor-? and its receptor during drug and dietary
Niwa M, Arisaka T, et al. Pioglitazone enhances splanchnic modulation of insulin resistance. Endocrinology 1994; 134:
glucose uptake as well as peripheral glucose uptake in non- 264±70.
insulin-dependent diabetes mellitus. AD-4833 Clamp-OGL 63. De Vos P, Lefebvre AM, Miller SG, Guerre-Millo M,
Study Group. Diabetes Res Clin Pract 1998; 41: 35±43. Wong K, Saladin R, et al. Thiazolidinediones repress ob gene
46. Nolan JJ, Ludvik B, Beerdsen P, Joyce M, Olefsky J. expression in rodents via activation of peroxisome prolifera-
Improvement in glucose tolerance and insulin resistance in tor-activated receptor gamma. J Clin Invest 1996; 98: 1004±9.
obese subjects treated with troglitazone. N Engl J Med 1994; 64. Kallen CB, Lazar MA. Antidiabetic thiazolidinediones inhibit
331: 1188±93. leptin (ob) gene expression in 3T3-L1 adipocytes. Proc Natl
47. Maggs DG, Buchanan TA, Burant CF, Cline G, Gumbiner Acad Sci USA 1996; 93: 5793±6.
B, Hsueh WA, et al. Metabolic effects of troglitazone 65. Maeda N, Takahashi M, Funahashi T, Kihara S, Nishizawa
monotherapy in type 2 diabetes mellitus. A randomized, H, Kishida K, et al. PPARgamma ligands increase expression

# 2002 Taylor & Francis. ISSN 0785-3890 Annals of Medicine 34


224 STUMVOLL . HAÈRING

and plasma concentrations of adiponectin, an adipose-derived in bone marrow stromal cells. Mol Pharmacol 1996; 50: 1087±
protein. Diabetes 2001; 50: 2094±9. 94.
66. Scheen AJ. Thiazolidinediones and liver toxicity. Diabetes 80. Tontonoz P, Nagy L, Alvarez JGA, Thomazy VA, Evans
Metab 2001; 27: 305±13. RM. PPAR gamma promotes monocyte/macrophage differ-
67. Maeda K. Hepatocellular injury in a patient receiving entiation and uptake of oxidized LDL. Cell 1998; 93: 241±52.
pioglitazone. Ann Intern Med 2001; 135: 306. 81. Kato K, Satoh H, Endo Y, Yamada D, Midorikawa S,
68. Al Salman J, Arjomand H, Kemp DG, Mittal M. Hepato- Sato W, et al. Thiazolidinediones down-regulate plasminogen
cellular injury in a patient receiving rosiglitazone. A case activator inhibitor type 1 expression in human vascular
report. Ann Intern Med 2000; 132: 121±4. endothelial cells: A possible role for PPARgamma in endothe-
69. Forman LM, Simmons DA, Diamond RH. Hepatic failure in lial function. Biochem Biophys Res Commun 1999; 258: 431±5.
a patient taking rosiglitazone. Ann Intern Med 2000; 132: 118± 82. Saez E, Tontonoz P, Nelson MC, Alvarez JG, Ming UT,
21. Baird SM, et al. Activators of the nuclear receptor PPAR-
70. Baldwin SJ, Clarke SE, Chenery RJ. Characterization of the gamma enhance colon polyp formation. Nat Med 1998; 4:
cytochrome P450 enzymes involved in the in vitro metabolism 1058±61.
of rosiglitazone. Br J Clin Pharmacol 1999; 48: 424±32. 83. Lefebvre AM, Chen I, Desreumaux P, Najib J, Fruchart JC,
71. Isley WL, Oki JC. Rosiglitazone and liver failure. Ann Intern Geboes K, et al. Activation of the peroxisome proliferator-
Med 2000; 133: 393. activated receptor gamma promotes the development of colon
72. Dunaif A, Scott D, Finegood D, Quintana B, Whitcomb R. tumors in C57BL/6J- APCMin/+ mice. Nat Med 1998; 4:
The insulin-sensitizing agent troglitazone improves metabolic 1053±7.
and reproductive abnormalities in the polycystic ovary 84. Sarraf P, Mueller E, Jones D, King FJ, DeAngelo DJ,
syndrome. J Clin Endocrinol Metab 1996; 81: 3299±306. Partridge JB, et al. Differentiation and reversal of malignant
changes in colon cancer through PPARgamma. Nat Med 1998;
73. Patel J, Anderson RJ, Rappaport EB. Rosiglitazone mono-
4: 1046±52.
therapy improves glycemic control in patients with type 2
85. Zhu L, Gong B, Bisgaier CL, Aviram M, Newton RS.
diabetes: a twelve-week, randomized, placebo-controlled
Induction of PPARgamma1 expression in human THP-1
study. Diab Obes Metab 1999; 1: 165±72.
monocytic leukemia by 9-cis-retinoic acid is associated with
74. Adams M, Montague CT, Prins JB, Holder JC, Smith SA,
cellular growth suppression. Biochem Biophys Res Commun
Sanders L, et al. Activators of peroxisome proliferator-
1998; 251: 842±8.
activated receptor gamma have depot-speci®c effects on
86. Imano E, Kanda T, Kawamori R, Kajimoto Y, Yamasaki Y.
human preadipocyte differentiation. J Clin Invest 1997; 100:
Pioglitazone-reduced insulin resistance in patient with Werner
3149±53. syndrome. Lancet 1997; 350: 1365.
75. Akazawa S, Sun F, Ito M, Kawasaki E, Eguchi K. Ef®cacy of 87. Adler A, Turner RC. The diabetes prevention program.
troglitazone on body fat distribution in type 2 diabetes. Diabetes Care 1999; 22: 543±5.
Diabetes Care 2000; 23: 1067±71. 88. Elbrecht A, Chen Y, Adams A, Berger J, GrifŽn P, Klatt T, et
76. Kawai T, Takei I, Oguma Y, Ohashi N, Tokui M, Oguchi S, al. L-764406 is a partial agonist of human peroxisome
et al. Effects of troglitazone on fat distribution in the proliferator-activated receptor gamma. The role of Cys313
treatment of male type 2 diabetes. Metabolism 1999; 48: in ligand binding. J Biol Chem 1999; 274: 7913±22.
1102±7. 89. Mukherjee R, Hoener PA, Jow L, Bilakovics J, Klausing K,
77. Kelly IE, Han TS, Walsh K, Lean ME. Effects of a Mias DE, et al. A selective peroxisome proliferator-activated
thiazolidinedione compound on body fat and fat distribution receptor-gamma (PPARgamma) modulator blocks adipocyte
of patients with type 2 diabetes [published erratum appears in differentiation but stimulates glucose uptake in 3T3-L1
Diabetes Care 1999 Mar;22(3):536]. Diabetes Care 1999; 22: adipocytes. Mol Endocrinol 2000; 14: 1425±33.
288±93. 90. OberŽeld JL, Collins JL, Holmes CP, Goreham DM,
78. Mori Y, Murakawa Y, Okada K, Horikoshi H, Yokoyama Cooper JP, Cobb JE, et al. A peroxisome proliferator-
J, Tajima N, et al. Effect of troglitazone on body fat activated receptor gamma ligand inhibits adipocyte differen-
distribution in type 2 diabetic patients. Diabetes Care 1999; tiation. Proc Natl Acad Sci USA 1999; 96: 6102±6.
22: 908±12. 91. Reginato MJ, Bailey ST, Krakow SL, Minami C, Ishii S,
79. Gimble JM, Robinson CE, Wu X, Kelly KA, Rodriguez BR, Tanaka H, et al. A potent antidiabetic thiazolidinedione with
Kliewer SA, et al. Peroxisome proliferator-activated receptor- unique peroxisome proliferator-activated receptor gamma-
gamma activation by thiazolidinediones induces adipogenesis activating properties. J Biol Chem 1998; 273: 32679±84.

# 2002 Taylor & Francis. ISSN 0785-3890 Annals of Medicine 34

You might also like