ArticlePDF AvailableLiterature Review

The metabolic syndrome—An ongoing story

Authors:

Abstract

The metabolic syndrome refers to the clustering of cardiovascular risk factors that include diabetes, obesity, dyslipidaemia and hypertension. Due to various definitions and unexplained pathophysiology it is still a source of medical controversy. Insulin resistance and visceral obesity have been recognized as the most important pathogenic factors. Insulin resistance could be defined as the inability of insulin to produce its numerous actions, in spite of the unimpaired secretion from the beta cells. Metabolic abnormalities result from the interaction between the effects of insulin resistance located primarily in the muscle and adipose tissue and the adverse impact of the compensatory hyperinsulinaemia on tissues that remain normally insulin-sensitive. The clinical heterogeneity of the syndrome can be explained by its significant impact on glucose, fat and protein metabolism, cellular growth and differentiation, and endothelial function. Visceral fat represents a metabolically active organ, strongly related to insulin sensitivity. Moderating the secretion of adipocytokines like leptin, adiponectin, plasminogen activator inhibitor 1 (PAI-1), tumor necrosis factor alfa (TNF-alfa), interleukin-6 (IL-6) and resistin, it is associated with the processes of inflammation, endothelial dysfunction, hypertension and atherogenesis. In 2005, the International Diabetes Federation (IDF) has proposed a new definition, based on clinical criteria and designed for global application in clinical practice. Visceral obesity measured by waist circumference is an essential requirement for diagnosis; other variables include increased triglyceride and decreased HDL levels, hypertension and glucose impairment. Whatever the uncertainties of definition and etiology, metabolic syndrome represents a useful and simple clinical concept which allows earlier detection of type 2 diabetes and cardiovascular disease.
INTRODUCTION
The metabolic syndrome refers to the clustering of
cardiovascular risk factors that include diabetes, obesity,
dyslipidaemia and hypertension (1, 2). This clustering of risk
factors, which is not thought to be grouped by chance alone, is
frequently seen in everyday clinical practice. Approximately 1
adult in 4 or 5, depending on the country, has metabolic
syndrome. Incidence increases with age; it has been estimated
that in the category over 50 years of age, metabolic syndrome
affects more than 40% of the population in the United States and
nearly 30% in Europe (2, 3).
Metabolic syndrome has been widely accepted as a simple
clinical tool for earlier detection of type 2 diabetes and
cardiovascular disease (4, 5). It has been estimated that people with
the metabolic syndrome are at twice the risk of developing
cardiovascular disease compared with those without the syndrome,
and experience a five-fold increased risk of type 2 diabetes (1, 4).
However, due to unclear underlying pathophysiologic
processes leading to its development, and confusion between the
conceptual definitions, metabolic syndrome continues to be a
source of medical controversy.
Recently, the American Diabetes Association (ADA) and the
European Association for the Study of Diabetes (EASD) have
advised refocusing on the individual components of the
syndrome without regarding the syndrome as an identifiable
target. This statement was not accepted by the International
Diabetes Federation (IDF), which emphasized that regardless of
the uncertainties of definition and aetiology, it is advisable to
regard the metabolic syndrome as a whole (5, 6).
PATHOPHYSIOLOGY
The association of obesity and metabolic abnormalities with
poor cerebrovascular outcome had been recognized long before
the concept of the metabolic syndrome became popular.
However, it was in 1988 when Dr Gerald Reaven postulated “the
syndrome X“, which we now call the metabolic syndrome (7).
Reaven noticed that there were many people who at the same
time had glucose intolerance, hyperinsulinaemia, high
triglycerides (TG), low high-density lipoprotein (HDL)
cholesterol, and hypertension, all being factors leading to the
development of cardiovascular disease. He proposed insulin
resistance as the driving force of the syndrome, which has
enabled more insight into the condition (7, 8).
Over the past decades many other abnormalities, in
particular chronic pro-inflammatory and pro-thrombotic states,
JOURNAL OF PHYSIOLOGY AND PHARMACOLOGY 2009, 60, Suppl 7, 19-24
www.jpp.krakow.pl
L. DUVNJAK, M. DUVNJAK
THE METABOLIC SYNDROME – AN ONGOING STORY
Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Zagreb, and University
Hospital “Sestre Milosrdnice”, Division of Gastroenterology and Hepatology, Zagreb, Croatia
The metabolic syndrome refers to the clustering of cardiovascular risk factors that include diabetes, obesity,
dyslipidaemia and hypertension. Due to various definitions and unexplained pathophysiology it is still a source of
medical controversy. Insulin resistance and visceral obesity have been recognized as the most important pathogenic
factors. Insulin resistance could be defined as the inability of insulin to produce its numerous actions, in spite of the
unimpaired secretion from the beta cells. Metabolic abnormalities result from the interaction between the effects of
insulin resistance located primarily in the muscle and adipose tissue and the adverse impact of the compensatory
hyperinsulinaemia on tissues that remain normally insulin-sensitive. The clinical heterogeneity of the syndrome can be
explained by its significant impact on glucose, fat and protein metabolism, cellular growth and differentiation, and
endothelial function. Visceral fat represents a metabolically active organ, strongly related to insulin sensitivity.
Moderating the secretion of adipocytokines like leptin, adiponectin, plasminogen activator inhibitor 1 (PAI-1), tumor
necrosis factor alfa (TNF-alfa), interleukin-6 (IL-6) and resistin, it is associated with the processes of inflammation,
endothelial dysfunction, hypertension and atherogenesis. In 2005, the International Diabetes Federation (IDF) has
proposed a new definition, based on clinical criteria and designed for global application in clinical practice. Visceral
obesity measured by waist circumference is an essential requirement for diagnosis; other variables include increased
triglyceride and decreased HDL levels, hypertension and glucose impairment. Whatever the uncertainties of definition
and etiology, metabolic syndrome represents a useful and simple clinical concept which allows earlier detection of type
2 diabetes and cardiovascular disease.
Key words: metabolic syndrome, insulin resistance, visceral obesity, type 2 diabetes, cardiovascular disease
were added to the syndrome, rendering the definition more
complex. The issue of abdominal obesity as the core of the
syndrome has gained more attention (9-11). It has been
recognised that metabolic abnormalities linked to insulin
resistance are usually found in patients with abdominal obesity
(12, 13). Although endocrine research had identified insulin
resistance and visceral obesity as important players in its
pathogenesis, they failed to present a unifying hypothesis (Fig.
1). From a practical point of view, it seems that there is no need
to dissociate the two conditions. Insulin resistance is considered
to be at the core of the syndrome, while central obesity is its
most prevalent clinical manifestation (14).
INSULIN RESISTANCE
Insulin resistance can be defined as the inability of insulin to
produce its numerous actions, in spite of the unimpaired
secretion from the beta cells (15-17). Insulin is the most potent
anabolic hormone in our body, which has a significant role in
glucose, fat and protein metabolism, but also influences cellular
growth and differentiation, as well as the endothelial function.
These numerous actions explain the clinical heterogeneity of the
metabolic syndrome (7).
Insulin elicits its various biological responses by binding to
a specific receptor (15, 16). The ability of insulin receptor to
autophosphorylate and phosphorylate intracellular substrates is
crucial for complex cellular responses to insulin (15-17). Insulin
binding to the alfa subunit of insulin receptor results in
conformational changes in the receptor, stimulation of the
tyrosine kinase activity intrinsic to the βsubunit which in turn
triggers the signalling cascades (Fig. 2).
Insulin receptor transphosphorylation of several substrates
including insulin receptor substrate (IRS) proteins 1-4 leads to
the activation of downstream signalling pathways which mediate
insulin actions. The four IRS proteins show tissue-specific
differences in mediating insulin action, with IRS-1 playing a
prominent role in the skeletal muscle and IRS-2 in the liver. Two
major signalling pathways activated by insulin binding to its
receptor are the phosphatidylinositol-3’-kinase (PI3K) pathway
and mitogenic, or mitogen-activated protein kinase (MAPK)
pathway.
PI3K pathway plays a crucial role in the metabolic actions of
insulin, glycogen, lipid and protein synthesis, vasodilatation and
20


8
   1
9
!"
"#+
 9

Figure 1. Pathophysiology of the
metabolic syndrome

# 
0 
3
%3
3+
! 
" 
5."6
7
896!-,
37
Figure 2. Conditions associated with
the metabolic syndrome
anti-inflammatory effects. This pathway has been demonstrated
to be upstream of glucose transporters (GLUT) 4 translocation,
by which insulin promotes glucose uptake by muscle and
adipose tissue. The activation of MAPK pathway is associated
with cell growth and proliferation, decrease in nitric oxide
production and procoagulant effects (17, 18).
Insulin resistance could be caused by various genetic and
acquired conditions. Except in a few rare cases involving
antibodies against insulin receptor or mutations in the insulin
receptor gene, insulin resistance of the metabolic syndrome
results from impairments in cellular events distal to the
interaction between insulin and its surface receptor (7, 8).
Metabolic abnormalities result from the interaction between the
effects of insulin resistance located primarily in the muscle and
adipose tissue and the adverse impact of the compensatory
hyperinsulinaemia on tissues that remain normally insulin
sensitive (15, 16).
VISCERAL ADIPOSITY
Although adiposity has been traditionally defined as an
increase in total body mass, visceral fat accumulation has been
found to correlate with a cluster of metabolic abnormalities
observed among the metabolic syndrome patients (19).
Waist circumference is accepted as an easily obtainable
indicator of visceral adiposity. The standard calls for
measurement at the high point of the iliac crest in the supine
position (19-22).
Visceral fat, in comparison with the subcutaneous tissue,
represents a metabolically active organ, strongly related to
insulin sensitivity (23). Adipocytes from visceral fat have a very
different histology and biology from subcutaneous fat.
Subcutaneous fat tissue, characterised by small, insulin-sensitive
adipocytes, is a storage fat depot, without vascular stroma and
cellular infiltration. Fat taken from visceral compartments and
composed of large, insulin resistant adipocytes, has a well-
developed vasculature with the infiltration of inflammatory
cells. Increased lypolisis in large insulin resistant adipocytes
leads to increased synthesis of very-low-density lipoprotein
(VLDL) and low-density lipoprotein (LDL) in the liver, driving
some of typical changes in the lipoprotein profile.
Inflammatory cells regulate adipocyte behaviour as a source
of hormones and cytokines, called adipokines, with
proinflammatory and proatherogenic effects. Circulating levels
of cytokines including resistin, leptin, TNFα, interleukin -6 (IL-
6), C-reactive protein, fibrinogen and plasminogen activator
inhibitor 1 (PAI-1) are generally increased in obese subjects and
in patients with diabetes (23-26). On the contrary, visceral
adiposity is a state with a relative deficiency of adiponectin, a
tissue-specific circulating hormone with insulin-sensitising and
anti-atherogenic properties. Adiponectin stimulates glucose use
and fatty acid oxidation in the muscle, enhances insulin
sensitivity in the liver, increases free fatty acid (FFA) oxidation,
reduces hepatic glucose output and inhibits monocyte adhesion
and macrophage transformation to foam cells within the vascular
wall (24-26).
DEFINITIONS
Throughout the years several classifications for the
metabolic syndrome have been proposed, emphasising insulin
resistance or visceral obesity. However, there are 3 main ones:
The World Health Organization (WHO) definition, the Adult
Treatment Panel III (ATPIII) Report and the International
Diabetes Federation (IDF) consensus on the metabolic syndrome
(Table 1).
According to the WHO definition from 1999, the syndrome
is present in a person with diabetes, impaired fasting glucose,
impaired glucose tolerance or insulin resistance harbouring at
least two of the following criteria: waist-to-hip ratio >0.90 in
men or >0.85 cm in women, serum triglyceride 150mg/dl or
HDL-C<35mg/dl in men and <39mg/dl in women, urinary
albumin excretion rate >20 mcg/min and blood pressure 140/90
mmHg (27).
21

 !" # ! 
#
$%
&&&
'()
*+

,
#-
#
+..(
'/

0
+..(
'+)
*+

,
%1 









 !
"
#
$%&'
()*+ &

,-.
/$ 
/$&!
*+0&

(',
!)
)(

1
'&
.
+'
!
()*+ &
,-.
/ 
/0&!

*+$&



!

+&
,
.
2
3
0
!4
()*+ &
,-.
/ 
/0&!

*+$&



3

+&
,
$%-2--%34$-2541-64!-7
Table 1. Definitions of the Metabolic Syndrome
In 2001, the National Cholesterol Education Program - Adult
Treatment Panel (NCEP–ATPIII) defined the metabolic
syndrome as having at least three of the following abnormalities:
waist circumference >102 cm in men and >88 cm in women,
serum triglyceride 150mg/dl, HDL-C 40mg/dl in men and
<50mg/dl in women, BP130/85 mmHg and serum glucose
110mg/dl (28). This definition was slightly modified in 2005
(2, 14). That same year, the International Diabetes Federation
(IDF) proposed a new definition based on clinical criteria and
designed for global application in clinical practice. This
definition represents modifications of the WHO and ATP III
definitions and places greater emphasis on visceral obesity as the
core feature of the syndrome. Visceral obesity measured by waist
circumference is an essential requirement for the diagnosis,
while other variables employed by ATP III are slightly (Table 1).
IDF defined visceral obesity for different ethnic populations
based on waist circumference measurements obtained from
epidemiologic data of various ethnic populations (14).
CONDITIONS ASSOCIATED
WITH THE METABOLIC SYNDROME
Type 2 Diabetes
In insulin resistant state primary effects of insulin blood
glucose, i.e. decreased glucose hepatic production and
increased peripheral glucose uptake in the muscle, are
abolished. As long as the pancreatic beta cells are able to secrete
large amounts of insulin needed to prevent increases in plasma
glucose, normal glucose tolerance is maintained. In individuals
with abnormal beta cells due to both genetic and acquired
conditions, frank hyperglycaemia with relative insulin
deficiency will develop (8, 11, 16, 17). Although approximately
25% of insulin resistant patients have normal glucose tolerance
test, this condition significantly increases the risk of developing
type 2 diabetes (16).
Dyslipidaemia
Insulin resistant state is characterised by resistance to
insulin-inhibited lipolysis in the adipose tissue, leading to
overproduction of FFAs in the plasma and increased FFA uptake
by the liver.
FFA leads to increased liver concentrations of TG and
cholesterol esters (CE). High blood TG concentrations in the
form of VLDL induce cholesterol ester transfer protein (CETP)
activity, which promotes transfer of TG from VLDL to HDL and
a subsequent increase in HDL clearance and decreased HDL
concentrations. It also promotes the transfer of TG into LDL in
exchange for LDL cholesterol ester. The triglyceride-rich LDL
can undergo hydrolysis by hepatic lipase or lipoprotein lipase,
which leads to a small, dense, cholesterol-depleted LDL particle
(SD-LDL).
All three components of atherogenic dyslipidaemia are
individually associated with a cardiovascular risk (29, 39).
Hypertension
Insulin resistance and subsequent hyperinsulinaemia induce
blood pressure elevation by activating sympathetic nervous
system and renin-angiotensin-aldosterone system (RAAS)
resulting in sodium retention and volume expansion, and
endothelial and renal dysfunction (12, 16, 18). Hyperinsulinaemia
stimulates the activation of RAAS in blood vessels and the heart,
generating the production of angiotensin II and its pro-
atherogenic effects. At the same time, hyperinsulinaemia in
insulin resistant subjects stimulates the MAPK pathway, which
promotes vascular and cardiac injury (15, 16). The local RAAS in
the visceral adipose tissue exerts more powerful systemic effects
compared with the subcutaneous adipose tissue. Angiotensin II
acts through angiotensin 1 receptors, inhibiting vasodilatatory
effects of insulin on blood vessels and glucose uptake into the
skeletal muscle cells by blocking insulin action on
phosphatydilinositol-3 kinase and protein kinase beta through free
oxygen production (16, 18, 31). This leads to a decrease in nitric
oxide (NO) production in endothelial cells, vasoconstriction in
smooth muscle cells, and GLUT 4 inhibition in the skeletal
muscles. The second mechanism by which insulin resistance
contributes to hypertension includes angiotensin 1 receptor
overactivity, which further leads to vasoconstriction and volume
expansion (12, 17, 18).
Polycystic ovary syndrome (PCOS)
PCOS is the most common endocrine abnormality in
premenopausal women, characterised by oligo/anovulation,
clinical and/or biochemical hyperandrogenism and polycystic
ovarian morphology. Although the pathophysiology is not
completely understood, there is evidence that insulin resistance
and compensatory hyperinsulinaemia play a key role (8, 16, 32,
33). Hyperinsulinaemia acting on normally insulin sensitive
tissues augments androgen production. It has been proposed that
insulin acts directly and indirectly through the pituitary.
Insulin increases LH activity, stimulates ovarian receptors of
insulin and IgF, enhances the amplitude of serum LH pulses,
stimulates adrenal androgen production and suppresses hepatic
production of sex-hormone-binding globulin (SHBG), resulting
in increased testosterone (16, 17, 32-34).
Many studies have proven that women with PCOS are at an
increased risk for the development of type 2 diabetes,
dyslipidaemia, hypertension and cardiovascular disease (33-35).
The prevalence of the metabolic syndrome was found to be
nearly 2-fold higher in women with PCOS than in general
population, matched for age and BMI (34).
Non-alcoholic fatty liver disease
Non-alcoholic fatty liver disease (NAFLD) represents a
spectrum of several non-alcoholic-related steatotic liver
diseases, ranging from benign fatty liver to non-alcoholic
steatohepatitis (NASH), associated with cirrhosis and
hepatocellular carcinoma. Increased prevalence of obesity,
diabetes, hyperlipidaemia, and insulin resistance in patients
with NAFLD implicate a close link with the metabolic
syndrome (35, 36). Insulin resistance, present in 98% of
patients with NAFLD, leads to increased lipolysis and
circulating FFAs, decrease in insulin-mediated glucose
disposal, inhibition of glucose utilization and promotion of
gluconeogenesis (7, 8, 16). Elevated plasma glucose and
insulin concentrations promote de novo fatty acid synthesis
(lipogenesis) and impair β-oxidation, thereby contributing to
the development of hepatic steatosis. Decreased adiponectin
hinders FFAs oxidation contributing to fat accumulation in the
liver (8, 11, 17). However, the reason some patients with benign
disease develop the more aggressive form of NASH is unclear.
It seems reasonable that the development of NASH requires
additional pathophysiologic abnormalities. In the context of
multiple-hit hypothesis oxidative stress and various cytokines
like TNF-αhave been implicated in the progression of fatty
liver to NASH. TNF-α, synthesised by hepatocytes and
Kupffer, cells cause hepatocyte injury and inflammation,
leading to the activation of stellate cells and fibrosis (36, 37).
Various cytokines secreted by adipose tissue contribute to
22
insulin resistance in the muscle and liver (32-38). Recently it
has been postulated that the liver could be the primary source of
systemic insulin resistance. Insulin resistance caused by hepatic
activation of NF-κB promoting systemic inflammation and
insulin resistance in the skeletal muscle was documented in a
mice model (37, 38). Although there is no doubt that insulin
resistance, visceral obesity and fatty liver are strongly
interrelated, it seems that the old question concerning the
metabolic syndrome is raised again: what comes first?
THERAPEUTIC APPROACH TO PATIENTS
WITH THE METABOLIC SYNDROME
The lack of specific algorithm makes the therapeutic
approach to patients with the metabolic syndrome difficult and
heterogeneous. Weight reduction by means of dietary changes
and promotion of physical activity are widely accepted as the
main approaches. Both patients and physicians agree that
unhealthy lifestyle aggravates the underlying pathology (39-41).
However, in clinical practice lifestyle modifications are usually
not sufficient to obtain the target value of individual risk factors.
This fact underlines the therapeutic importance of
pharmacological interventions capable of reducing blood
pressure, dyslipidaemia, glucose metabolism impairment and
other abnormalities related to the metabolic syndrome.
Although a clinical diagnosis of the metabolic syndrome is
not sufficient to assess global risk for cardiovascular disease,
this syndrome involves three or more risk factors, often organ
damage and diabetes (1, 2, 8, 14). For this reason the primary
goal in the treatment of patients with the metabolic syndrome
should be the prevention of major vascular events. To achieve
this goal, physicians should pay attention to the choice of drug
in order to avoid aggravation of metabolic abnormalities.
Drugs that improve insulin sensitivity such as metformin
and glitazones are indicated in the treatment of type 2 diabetes.
They have also shown efficacy in the prevention of diabetes
and treatment of PCOS and NASH (42-44). To control
atherogenic dyslipidaemia, a combination therapy of statin and
fibrates is usually required (45). In the treatment of
hypertension a particular emphasis should be placed on ACE
inhibitors and angiotensin II receptor blockers, as these drugs
have shown efficacy in the prevention of diabetes. Central
sympatholytic agents like moxonidine exert additional
beneficial effects of increasing insulin sensitivity (46). If type
2 diabetes is present, in 2/3 of the patients target blood pressure
values can be achieved only with two or more antihypertensive
drugs (12, 13, 46).
Increased understanding of the mechanisms contributing to
the vicious cycle of the metabolic syndrome, as well as critical
analysis of the results of ongoing trials are important for
developing a logical, evidence-based treatment strategy.
CONCLUSION
Whether or not one accepts this condition as a distinct entity,
and in spite of the controversies surrounding its
pathophysiology, the concept of the metabolic syndrome
continues to gain attention. The prevalence of the metabolic
syndrome is increasing at a disturbing rate and within the context
of a proven association with cardiovascular disease, the leading
cause of mortality in the modern world.
In spite of a recent debate, the metabolic syndrome remains
important in the clinical practice, as it integrates the most
common abnormalities representing major cardiovascular risk
factors. The arguments for and against the significance of the
metabolic syndrome will continue to be a matter for debate. On
the other hand, many other conditions will be added in the
future, creating a vicious cycle of “the ongoing story of the
metabolic syndrome”.
Conflict of interests: None declared.
REFERENCES
1. Grundy SM. Metabolic syndrome pandemic. Arterioscler
Thromb Vasc Biol 2008; 28: 629-636.
2. Cameron AJ, Shaw JE, Zimmet PZ. The metabolic
syndrome: prevalence in worldwide populations. Endocrinol
Metab Clin North Am 2006; 33: 351-375.
3. Ford ES, Giles WH, Mokdad AH. Increasing prevalence of
the metabolic syndrome among U.S. adults. Diabetes Care
2004; 27: 2444-2449.
4. Nesto RW. The relation of insulin resistance syndromes to
risk of cardiovascular disease. Rev Cardiovasc Med 2003; 4:
S11-S18.
5. Alberti KG, Zimmet PZ. Should we dump the metabolic
syndrome? BMJ 2008; 336: 641-643.
6. Kahn R, Ferrannini E, Buse J, Stern M. The metabolic
syndrome: time for a critical appraisal. Diabetes Care 2005;
28: 2289-2304.
7. Reaven GM. Role of insulin resistance in human disease.
Diabetes 1988; 37: 1595-1607.
8. Reaven GM. Pathophysiology of insulin resistance in human
disease. Physiol Rev 1995; 75: 473-486.
9. Shirai K. Obesity as the core of the metabolic syndrome and
the management of coronary heart disease. Curr Med Res
Opin 2004; 20: 295-304.
10. Despres JP, Lemieux I, Bergeron J, et al. Abdominal obesity
and the metabolic syndrome: contribution to global
cardiometabolic risk. Arterioscler Thromb Vasc Biol 2008;
28: 1039-1049.
11. Phillips LK, Prins JB. The link between abdominal obesity
and the metabolic syndrome. Curr Hypertens Rep 2008; 10:
156-164.
12. Sowers JR, Fronlich ED. Insulin and insulin resistance:
impact on blood pressure and cardiovascular disease. Med
Clin North Am 2004; 88: 63-82.
13. Bays H, Abate N, Chandalia M. Adiposopathy: sick fat
causes high blood sugar, high blood pressure and
dyslipidaemia. Future Cardiol 2005; 1: 39-59.
14. Alberti KG, Zimmet P, Shaw J. The metabolic syndrome - a
new worldwide definition. IDF Epidemiology Task Force
Consensus Group. Lancet 2005; 366: 1059-1062.
15. Reaven GM. Insulin resistance, compensatory
hyperinsulinemia, and coronary heart disease: syndrome X
revisited. In Handbook of Physiology, Section 7 The Endocrine
System, V
ol. II The Endocrine Pancreas and Regulation of
Metabolism. LS Jefferson, AD Cherrington, (eds). New York,
Oxford University Press, 2001, pp. 1169-1197.
16. Reaven GM. Insulin resistance and its consequences. In
Diabetes Mellitus: AFundamental and Clinical Text. D
LeRoith, SI Taylor, JM Olefasky (eds). Philadelphia,
Lippincott, Williams and Wilkins, 2004, pp. 899-915.
17. Flakoll PJ, Jensen MD, Cherrington AD. Physiological
action of insulin. In Diabetes Mellitus: AFundamental and
Clinical Text. D LeRoith, SI Taylor, JM Olefasky (eds).
Philadelphia, Lippincott, Williams and Wilkins, 2004, pp.
165-181.
18. Low Wang C, Goalstone ML, Draznin B. Molecular
mechanisms of insulin resistance that impact cardiovacsular
biology. Diabetes 2004; 53: 2735-2740.
23
19. Despres JP. Health consequences of visceral adiposity. Ann
Med 2001; 33: 534-541.
20. Gabriely I, Ma XH, Yang XM, et al. Removal of visceral
fat prevents insulin resistance and glucose intolerance of
aging. An adipokine-mediated process? Diabetes 2002; 51:
2951-2958.
21. Despres JP. Is visceral obesity the cause of the metabolic
syndrome? Ann Med 2006; 38: 52-63.
22. Despres JP. Intra-abdominal obesity: an untreated risk factor
for type 2 diabetes and cardiovascular disease. J Endocrinol
Invest 2006; 29(3 Suppl): 77-82.
23. Wajchenberg BL. Subcutaneous and visceral adipose tissue:
their relation to the metabolic syndrome. Endocr Rev 2000;
21: 697-738.
24. Wellen KE, Hotumisligil GS. Obesity-induced inflammatory
changes in adipose tissue. J Clin Invest 2003; 112: 1785-1788.
25. Nells JG, Olefsky JM. Inflamed fat: what starts the fire? J
Clin Invest 2006; 116: 33-35.
26. Trayhum P, Woods IS. Adipokines: inflammation and the
pleiotropic role of white adipose tissue. Br J Nutr 2004; 92:
347-355.
27. World Health Organization. Definition, diagnosis and
classification of diabetes mellitus and its complications.
Geneva, WHO, 1999
28. Executive summary of the third report of the National
Cholesterol Education Program (NCEP) Expert Panel on
Detection and Treatment of High Blood Cholesterol in Adults
(Adult treatment Pannel III). JAMA 2001; 285: 2468-2497.
29. Caballero AE. Endothelial dysfunction, inflammation and
insulin resistance: a focus on subjects at risk for type 2
diabetes. Curr Diab Rep 2004; 4: 237-246.
30. Deedwania PC. Metabolic syndrome and vascular disease: is
nature or nurture leading the new epidemic of cardiovascular
disease? Circulation 2004; 109(1): 2-4.
31. Rahmouni K, Correia MLG, Haynes WG, Mark AL. Obesity-
associated hypertension- new insights into mechanisms.
Hypertension 2005; 45: 9-14.
32. Cascella T, Palomba S, De Sio I, et al. Visceral fat is
associated with cardiovascular risk in women with
polycystic ovary syndrome. Hum Reprod 2008; 23: 153-159.
33. Dunaif A. Insulin resistance and the polycystic ovary
syndrome: mechanism and implications for pathogenesis.
Endocr Rev 1997; 18: 774-800.
34. Apridonidze T, Essah PA, Iuorno MJ, Nestler JE. Prevalence
and characteristics of the metabolic syndrome in women
with polycystic ovary syndrome. J Clin Endocrinol Metab
2005; 90: 1929-1935.
35. Moran L,Teede H. Metabolic features of the reproductive
phenotypes of polycystic ovary syndrome. Hum Reprod
Update 2009; 15: 477-488.
36. Kotronen A, Yki-Jarvinen H. Fatty liver: A novel component
of the metabolic syndrome. Arterioscler Thromb Vasc Biol
2008; 28: 27-38.
37. Lazar MA. The humoral side of insulin resistance. Nat Med
2006; 12: 43-44.
38. Cai D, Yuan M, Frantz DF, et al. Local and systemic insulin
resistance resulting from hepatic activation of IKK-beta and
NF-kappaB. Nat Med 2005; 11: 183-190.
39. Giugliano D, Ceriello A, Esposito K. The effects of diet on
inflammation: emphasis on the metabolic syndrome. J Am
Coll Cardiol 2006; 48: 677-685.
40. Zwirska-Korczala K, Konturek SJ, Sodowski M, et al. Basal
and postprandial plasma levels of PYY, ghrelin,
cholecystokinin, gastrin and insulin in women with
moderate and morbid obesity and metabolic syndrome. J
Physiol Pharmacol 2007; 58(Suppl. 1): 13-35.
41. Zoladz JA, Pilc A, Majerczak J, Grandys M, Zapart-
Bukowska J, Duda K. Endurance training increases
plasma brain-derived neurotrophic factor concentration in
young healthy men. J Physiol Pharmacol 2008; 59(Suppl.
7): 119-132.
42. Ferrucci A, Sciarretta S, Valenti V, Tocci G. How to manage
metabolic syndrome: clinical approach based on
cardiovascular risk profile. High Blood Press Cardiovasc
Prev 2005; 12: 231-238.
43. Cheang KI, Huszar JM, Best AM, Sharma S, Essah PA,
Nestler JE. Long-term effect of metformin on metabolic
parameters in the polycystic ovary syndrome. Diab Vasc Dis
Res 2009; 6: 110-119.
44. Zwirska-Korczala K, Sodowski A, Konturek SJ, et al.
Postprandial response of ghrelin and PYY and indices of
low-grade chronic inflammation in lean women with
polycytic ovary syndrome. J Physiol Pharmacol 2008;
59(Suppl 2): 161-178.
45. Ascaso J, Gonzalez Santos P, Hernandez MA, et al.
Management of dyslipidemia in the metabolic syndrome:
recommendations of the Spanish HDL-Forum. Am J
Cardiovasc Drugs 2007; 7: 39-42.
46. Bakris GL. Current perspectives on hypertension and
metabolic syndrome. J Manag Care Pharm 2007; 13: S3-S5.
R e c ei v e d: October 15, 2009
Accepted: December 11, 2009
Author’s address: Prof. Lea Duvnjak, MD, PhD, Vuk
Vrhovac University Clinic, Dugi dol 4a, Zagreb, Croatia; E-mail:
lduvnjak@idb.hr
24
... Bu sonuçlar çalışmamızla benzer olarak, gebelikte maternal obezitenin plasental ve fetal metabolik gelişim üzerinde doğrudan bir etkisi olduğunu destekler niteliktedir. (Duvnjak & Duvnjak, 2009). Çalışmada gebelik öncesi ve doğumda obez olanların emzirme başarısının düşük olduğu belirlendi. ...
Article
Full-text available
Objective: This study aimed to determine the effect of maternal BMI on NST parameters, breastfeeding success and postpartum depression. Method: This cross-sectional study was conducted with 427 women between September 2022 and April 2023. Women who gave birth by vaginal or cesarean delivery and agreed to participate in the study were included in the study. The data of the study were collected using the Introductory Information Form, the LATCH Breastfeeding Diagnostic Scale, and the Edinburgh Postpartum Depression Scale (EDSS). Results: In the study, 57.4% of the women were at normal weight according to their pre-pregnancy BMI, 41.9% were at normal weight according to their BMI at birth, and 99.5% of them were overweight or underweight during pregnancy. received was determined. In the NST according to BMI at birth, 92.9% of those with baseline tachycardia were obese, 53.4% of those with a deceleration number of 3 or more were obese, and the difference between the groups was statistically significant. In the study, it was determined that as BMI before pregnancy and at birth increased, the mean breastfeeding score decreased. In the study, it was determined that the success of breastfeeding was low and the risk of postpartum depression was high in those who were obese before pregnancy and at birth (p<.05). Conclusion: As a result, it was determined that high BMI during pregnancy and delivery may cause negative birth outcomes, negatively affect breastfeeding behavior, and increase the risk of postpartum depression. Keywords: Body mass index, nonstress test, pregnancy, maternal, postpartum depression ÖZ Amaç: Bu çalışmada maternal BKİ'nin, NST parametereleri, emzirme başarısı ve doğum sonu depresyona etkisini belirlemek amaçlanmıştır. Yöntem: Kesitsel tipte tasarlanan bu çalışma Eylül 2022-Nisan 2023 tarihleri arasında 427 kadınla yürütüldü. Araştırmaya vajinal veya sezaryen doğum yapan kadınlar alındı. Araştırmanın verileri Tanıtıcı Biligi Formu, LATCH Emzirme Tanılama Ölçeği ve Edinburg Doğum Sonrası Depresyon Ölçeği(EDSDÖ) kullanılarak toplandı. Bulgular: Çalışmada gebelik öncesi BKİ'lerine göre kadınların %57,4'ünün normal kiloda olduğu, doğumdaki BKİ'lerine göre %41,9'unun normal kiloda olduğu, %99,5'inin gebelik sürecinde gereğinden az ya da daha fazla kilo aldığı belirlendi. Doğumdaki BKİ'ne göre NST'de, baseline değeri taşikardik olanların %92,9'unun, deselerasyon sayısı 3 ve üzeri olanların %53,4'ün obez olduğu ve gruplar arasındaki farkın istatistiksel olarak anlamlı olduğu belirlendi. Çalışmada gebelik öncesi ve doğumdaki BKİ arttıkça emzirme puan ortalamasının azaldığı belirlendi. Çalışmada gebelik öncesi ve doğumda obez olanların emzirme başarısının düşük olduğu, doğum sonu depreyon risklerinin yüksek olduğu belirlendi (p<,05). Sonuç: Sonuç olarak gebelik ve doğumdaki yüksek BKİ'nin olumsuz doğum sonuçlarına neden olabileceği, emzirme davranışını olumsuz etkileyebileceği, doğum sonu depresyon riskini artırabileceği belirlendi.
... Excess TG in VLDL will be transferred to HDL to form TG-rich HDL. This TG-rich HDL is then cleared from circulation via renal clearance, causing a decrease in HDL levels in the blood [47]. Therefore, this study found that there was a decrease in serum HDL levels of HCHF-induced MetS rats in week 16. ...
Article
Full-text available
Metabolic syndrome (MetS) is composed of central obesity, hyperglycemia, dyslipidemia and hypertension that increase an individual’s tendency to develop type 2 diabetes mellitus and cardiovascular diseases. Kelulut honey (KH) produced by stingless bee species has a rich phenolic profile. Recent studies have demonstrated that KH could suppress components of MetS, but its mechanisms of action are unknown. A total of 18 male Wistar rats were randomly divided into control rats (C group) (n = 6), MetS rats fed with a high carbohydrate high fat (HCHF) diet (HCHF group) (n = 6), and MetS rats fed with HCHF diet and treated with KH (HCHF + KH group) (n = 6). The HCHF + KH group received 1.0 g/kg/day KH via oral gavage from week 9 to 16 after HCHF diet initiation. Compared to the C group, the MetS group experienced a significant increase in body weight, body mass index, systolic (SBP) and diastolic blood pressure (DBP), serum triglyceride (TG) and leptin, as well as the area and perimeter of adipocyte cells at the end of the study. The MetS group also experienced a significant decrease in serum HDL levels versus the C group. KH supplementation reversed the changes in serum TG, HDL, leptin, adiponectin and corticosterone levels, SBP, DBP, as well as adipose tissue 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1) level, area and perimeter at the end of the study. In addition, histological observations also showed that KH administration reduced fat deposition within hepatocytes, and prevented deterioration of pancreatic islet and renal glomerulus. In conclusion, KH is effective in preventing MetS by suppressing leptin, corticosterone and 11βHSD1 levels while elevating adiponectin levels.
... Type 2 diabetes, is a polygenic disease that is related to insulin resistance condition and, consequently, raise blood glucose concentration. There are several risk factors that result in insulin resistance, such as dyslipidemia by increasing the concentration of lipids such as cholesterol and triglyceride in the blood, obesity, and visceral adiposity (accumulation of fat that wraps around the abdominal cavity) [62,63]. Furthermore, sedentary lifestyle is considered as a factor that resulted in the development of insulin resistance and type 2 diabetes, which is low physical activity that increases the complication of cardiovascular diseases and mortality [55,64]. ...
Article
Full-text available
Carbohydrates are the major energy source for the living cells that synthesized from carbon dioxide and water during photosynthesis process by green plants through absorption of sun light. In human, carbohydrates provide more than 55% of body energy which are mostly come from exogenous source. Carbohydrates can also provide energy from endogenous source such as glycogenolysis process. To be used as source of energy, carbohydrates compounds should undergo series of enzymatic metabolic reactions in the cell. Beside the energy productions, catabolism of carbohydrates provides different metabolites for synthesis of several biomolecules such as fatty acids, amino acids, DNA, and RNA. Among the three main examples of monosaccharide (glucose, galactose, and mannose). Glucose is considered as the central molecule in carbohydrate metabolism that all the major pathways of carbohydrate metabolism relate to glucose molecule such as glycolysis and glycogenesis process. Glucose is also considered as an important component of cellular metabolism in preserving carbon homeostasis. Liver plays a significant role in controlling and stabilizing blood glucose levels; therefore, it can be considered as glucostate monitor. In this article, we will review the major metabolic pathways of carbohydrate metabolism, their biochemical role in cellular energy production, and latest development in the understanding in these fields. Also, we discussed about the factors that participate in regulation of blood glucose concentration. We believe understanding this process is essential for control carbohydrate-related human disorders.
... Obesity is linked with an elevated risk of developing a number of health issues (1). If obesity is accompanied by an accumulation of further cardiometabolic risk factors, including hyperglycemia, dyslipidemia and hypertension (i.e. the metabolic syndrome, MetS), the risk of developing cardiovascular diseases (CVD) and certain types of cancer as well as for mortality is additionally significantly increased (2,3). Additionally, it has been reported that a large proportion of obese people suffer from non-alcoholic fatty liver disease (NAFLD), which is characterized by an increased fat storage in the liver independent of alcohol consumption (4). ...
Article
Full-text available
Non-alcoholic fatty liver disease (NAFLD) and cardiometabolic disorders are highly prevalent in obese individuals. Physical exercise is an important element in obesity and metabolic syndrome (MetS) treatment. However, the vast majority of individuals with obesity do not meet the general physical activity recommendations (i.e. 150 min of moderate activity per week). The present study aimed to investigate the impact of a highly time-saving high-intensity interval training (HIIT) protocol (28 min time requirement per week) on NAFLD fibrosis (NFS) and cardiometabolic risk scores in obese patients with MetS and elevated NFS values. Twenty-nine patients performed HIIT on cycle ergometers (5 x 1 min at an intensity of 80 - 95% maximal heart rate) twice weekly for 12 weeks and were compared to a control group without exercise (CON, n = 17). Nutritional counseling for weight loss was provided to both groups. NFS, cardiometabolic risk indices, MetS z-score, cardiorespiratory fitness (VO2max) and body composition were assessed before and after intervention. The HIIT (-4.3 kg, P < 0.001) and CON (-2.3 kg, P = 0.003) group significantly reduced body weight. There were no significant group differences in relative weight reduction (HIIT: -3.5%, CON: -2.4%). However, only the HIIT group improved NFS (-0.52 units, P = 0.003), MetS z-score (-2.0 units, P < 0.001), glycemic control (HbA1c: -0.20%, P = 0.014) and VO2max (+3.1 mL/kg/min, P < 0.001). Decreases in NFS (-0.50 units, P = 0.025) and MetS z-score (-1.4 units, P = 0.007) and the increment in VO2max (3.3 mL/kg/min, P < 0.001) were significantly larger in the HIIT than in the CON group. In conclusion, only 28 min of HIIT per week can elicit significant improvements in NFS and a several cardiometabolic health indices in obese MetS patients with increased NFS grades. Our results underscore the importance of exercise in NAFLD and MetS treatment and suggest that our low-volume HIIT protocol can be regarded as viable alternative to more time-consuming exercise programs.
... Several studies have proven that insulin resistance (IR) is the main factor to be concerned about in complications of DM [26][27][28]. Besides IR, some of the common risk factors for insulin resistance are oxidative stress, hydrolytic enzymatic inhibition, inflammation, genetic habitual, environmental, dyslipidemia, obesity, and epigenetic modulations [29,30]. Thus, many pathological factors use to contribute to insulin resistance, although the exact mechanism is not clear yet. ...
Article
Full-text available
Mushrooms belong to the family "Fungi" and became famous for their medicinal properties and easy accessibility all over the world. Because of its pharmaceutical properties, including anti-diabetic, anti-inflammatory, anti-cancer, and antioxidant properties, it became a hot topic among scientists. However, depending on species and varieties, most of the medicinal properties became indistinct. With this interest, an attempt has been made to scrutinize the role of edible mushrooms (EM) in diabetes mellitus treatment. A systematic contemporary literature review has been carried out from all records such as Science Direct, PubMed, Embase, and Google Scholar with an aim to represents the work has performed on mushrooms focuses on diabetes, insulin resistance (IR), and preventive mechanism of IR, using different kinds of mushroom extracts. The final review represents that EM plays an important role in anticipation of insulin resistance with the help of active compounds, i.e., polysaccharide, vitamin D, and signifies α-glucosidase or α-amylase preventive activities. Although most of the mechanism is not clear yet, many varieties of mushrooms' medicinal properties have not been studied properly. So, in the future, further investigation is needed on edible medicinal mushrooms to overcome the research gap to use its clinical potential to prevent non-communicable diseases.
... Via adipokine signaling, adipose tissue is able to communicate with many organs like the liver, pancreas, muscle, and brain, and is therefore able to modulate systemic metabolism (3)(4)(5)(6). Thus, adipose tissue dysfunction plays an important role in the pathogenesis of metabolic disorders, such as obesity, cardiovascular disease, insulin resistance, and diabetes mellitus (7)(8)(9). How adipose tissue specifically contributes to the pathogenesis of metabolic diseases is however highly complex and varies between different fat depots (10)(11)(12). It is thought that visceral adipose tissue is more likely to contribute to the pathogenesis of insulin resistance and type 2 diabetes mellitus (13,14), while accumulation of subcutaneous fat has even been reported to reduce metabolic disease risk (15)(16)(17). ...
Article
Full-text available
Adipose tissue (AT) is no longer considered to be responsible for energy storage only but is now recognized as a major endocrine organ that is distributed across different parts of the body and is actively involved in regulatory processes controlling energy homeostasis. Moreover, AT plays a crucial role in the development of metabolic disease such as diabetes. Recent evidence has shown that adipokines have the ability to regulate blood glucose levels and improve metabolic homeostasis. While AT has been studied extensively in the context of type 2 diabetes, less is known about how different AT types are affected by absolute insulin deficiency in type 1 or permanent neonatal diabetes mellitus. Here, we analyzed visceral and subcutaneous AT in a diabetic, insulin-deficient pig model (MIDY) and wild-type (WT) littermate controls by RNA sequencing and quantitative proteomics. Multi-omics analysis indicates a depot-specific dysregulation of crucial metabolic pathways in MIDY AT samples. We identified key proteins involved in glucose uptake and downstream signaling, lipogenesis, lipolysis and β-oxidation to be differentially regulated between visceral and subcutaneous AT in response to insulin deficiency. Proteins related to glycogenolysis, pyruvate metabolism, TCA cycle and lipogenesis were increased in subcutaneous AT, whereas β-oxidation-related proteins were increased in visceral AT from MIDY pigs, pointing at a regionally different metabolic adaptation to master energy stress arising from diminished glucose utilization in MIDY AT. Chronic, absolute insulin deficiency and hyperglycemia revealed fat depot-specific signatures using multi-omics analysis. The generated datasets are a valuable resource for further comparative and translational studies in clinical diabetes research.
... There is a direct influence of maternal obesity during pregnancy on placental and fetal cardiovascular and metabolic development. Within the cluster of cardiometabolic risk factors, obesity is one of the most important causal factors [44]. It has been shown that maternal pre-pregnancy obesity is associated with higher placental weight, placental vascular dysfunction, placental inflammation, and alterations in placental transporter and mitochondrial activity [6-8, 31, 45-47]. ...
Article
Full-text available
Background/Objective Healthy weight maintenance before and during pregnancy has a significant effect on pregnancy outcomes; however, there are no specific guidelines for gestational weight gain in pregnant Korean women. Therefore, we investigated the impact of pre-pregnancy body mass index (BMI) and gestational weight gain on the risk of maternal and infant pregnancy complications in pregnant Korean women. Methods Study participants comprised 3454 singleton pregnant women from the Korean Pregnancy Outcome Study who had baseline examination and pregnancy outcome data. Maternal pre-pregnancy BMI and gestational weight gain were categorized according to the Asia-pacific regional guidelines and the Institute of Medicine recommendations, respectively. The primary outcome was any adverse outcomes, defined as the presence of one or more of the following: hypertensive disorders of pregnancy, gestational diabetes mellitus, peripartum depressive symptom, cesarean delivery, delivery complications, preterm birth, small or large weight infant, neonatal intensive care unit admission, or a congenital anomaly. Multiple logistic regression models were applied to examine the independent and combined impact of pre-pregnancy BMI and gestational weight gain on the risk of maternal and infant outcomes. Results Obesity before pregnancy significantly increased the risk of perinatal adverse outcomes by more than 2.5 times [odds ratio (OR): 2.512, 95% confidence interval (CI): 1.817–3.473]. Compared to that in women with appropriate gestational weight gain, women with excessive weight gain had a 36.4% incremental increase in the risk of any adverse outcomes [OR: 1.364, 95% CI: 1.115–1.670]. Moreover, women who were overweight or obese before pregnancy and had excessive gestational weight gain had a three-fold increase in the risk of adverse outcomes [OR: 3.460, 95% CI: 2.210–5.417]. Conclusion This study highlights the need for appropriate weight recommendations before and during pregnancy to prevent perinatal complications in Korean women of childbearing age.
Article
Globally, the incidence of diabetes mellitus (DM) and Alzheimer's disease (AD) is increasing year by year, causing a huge economic and social burden, and their pathogenesis and aetiology have been proven to have a certain correlation. In recent years, more and more studies have shown that vacuolar adenosine triphosphatases (v‐ATPases) in eukaryotes, which are biomolecules regulating lysosomal acidification and glycolipid metabolism, play a key role in DM and AD. This article describes the role of v‐ATPase in DM and AD, including its role in glycolysis, insulin secretion and insulin resistance (IR), as well as its relationship with lysosomal acidification, autophagy and β‐amyloid (Aβ). In DM, v‐ATPase is involved in the regulation of glucose metabolism and IR. v‐ATPase is closely related to glycolysis. On the one hand, v‐ATPase affects the rate of glycolysis by affecting the secretion of insulin and changing the activities of key glycolytic enzymes hexokinase (HK) and phosphofructokinase 1 (PFK‐1). On the other hand, glucose is the main regulator of this enzyme, and the assembly and activity of v‐ATPase depend on glucose, and glucose depletion will lead to its decomposition and inactivation. In addition, v‐ATPase can also regulate free fatty acids, thereby improving IR. In AD, v‐ATPase can not only improve the abnormal brain energy metabolism by affecting lysosomal acidification and autophagy but also change the deposition of Aβ by affecting the production and degradation of Aβ. Therefore, v‐ATPase may be the bridge between DM and AD.
Article
INTRODUCTION: The inability of the heart to supply enough blood flow to the body's metabolic tissues is known as heart failure (H.F.). A patient's tolerance for physical activity is described by the N.Y.H.A. Classification of H.F., updated in 1994 to include objective assessments of heart function and disease. The phrase "cardiopulmonary disease" is used in medicine to refer to various dangerous diseases that affect the heart and lungs. Cardiovascular Disease (CVD) and Chronic Obstructive Pulmonary Disease (C.O.P.D.) are the two main cardiopulmonary conditions caused by cigarette use (C.O.P.D.). A severe respiratory infection brings on the lung condition known as pneumonia. The tiny sacs called alveoli that make up a healthy person's lungs are filled with air as they breathe. Pneumonia causes the alveoli to fill with pus and fluid, making breathing challenging and limiting oxygen intake. A metabolic syndrome is a group of risk factors associated with cardiovascular disease morbidity and mortality. It is often referred to as Syndrome X. (CVD). Despite being widespread, it has not received enough attention and occasionally goes unnoticed. As primary care practitioners, we must keep an eye out for these people so that early identification, intervention, and prevention can be put into practice.A Chinese lady with metabolic syndrome is described in the following paragraphs. Present complaint and investigation: on March 28, 2022, a 62-year-old female was hospitalized at A.V.B.R. Hospital with breathing difficulty, lower limb swelling, chest pain, mucus in the throat and lungs, loss of appetite, nausea, and vomiting. Twelve days have passed. Investigation: Haemoglobin is decreased by 10.7gm, WBC count 11,000 per microliter, Total R.B.C. count increased 5.84 million /cell, M.C.H. level Decreased 70fl, MCH IS DECREASE 22.8 pico-gm. Therefore CT report should be interpreted in correlation with clinical &pathological findings. PAST HISTORY: he was admitted eight days before outside the hospital for the same complaints on 26/02/2022. Pneumonia one year back for which she symptoms in 2000 for fibroid he took treatment. After the physical examination and investigation, diagnosed RESPIRATORY ACIDOSIS WITH OBSTRUCTIVE SLEEP APNEA WITH HYPERTENSION. Conclusion: He responded well to all medication and doctor care and made a good recovery.
Article
Full-text available
El síndrome metabólico es una condición clínica que incluye un conjuntode anomalías cardiometabólicas como hipertensión arterial, obesidad central, resistencia a la insulina y dislipidemia aterogénica. La prevalencia del síndrome metabólico se ha incrementado en los últimos años en el mundo, representando un problema de salud pública, porque aumenta directamente entre dos y seis veces el riesgo de enfermedades cardiovasculares y diabetes mellitus tipo 2. Los cambios en el estilo de vida pueden revertir los componentes del síndrome metabólico, pero en ocasiones es necesaria la intervención farmacológica para un control más estricto de los factores de riesgo. Este artículo tiene como objetivo explicar de forma clara y precisa los aspectos generales del síndrome metabólico en adultos.
Article
Full-text available
The metabolic syndrome represents the association in a single individual of a cluster of metabolic and hemodynamic factors, leading to an increased risk of type 2 diabetes and/or cardiovascular diseases. Several definitions exist (WHO, EGIR, NCEP-ATP III, AACE), but all of them include a cluster of criteria (hyper glycemia or type 2 diabetes, arterial hypertension, dyslipidemia, abdominal obesity) which increased these risks in parallel to their aggregation. The prevalence of the metabolic syndrome in industrialized countries represents 10 to 30% of the adult population, depending on the definition used and of the range of age, with a regular progression, particularly in women. Thus, it is needed to identify subjects with metabolic syndrome in the general population, and not only in overweight/obese subjects. This review, briefly presents the main definitions, as well as current data on pathophysiology, prevalence and consequences of the metabolic syndrome. Steps to diagnose it and guidance for the therapeutic management of metabolic syndrome in primary care practice are described.
Article
Full-text available
The aim of the study were to answer the question 1.) Whether circulating pro-inflammatory markers of endothelial dysfunction and due to chronic low-grade inflammation of obesity, are altered in untreated lean, young relatively healthy polycystic ovary syndrome (PCOS) patients in comparison with healthy controls; 2.) Whether postprandial plasma concentration pattern of ghrelin and PYY can be predictable as risk factors for atherosclerosis and depend of obesity. Forty young women with PCOS were divided in two groups: 19 lean and 21 obese. The control group included 20 lean, healthy volunteers. Plasma total and active ghrelin, total PYY and PYY(3-36), serum adiponectin and insulin were measured using RIA technique, serum sCD40L, visfatin, sP-, sE-selectins, resistin by EIA. Composition of test meal was: 527 kcal total and consisted of 24.1% fat, 54.4% carbohydrate and 21.5% protein. Total and active ghrelin and total PYY were significantly lower in obese PCOS women, whereas active ghrelin was also significantly lower in lean PCOS women compared to controls. Postprandial plasma total ghrelin levels decrease were blunted in lean and obese compared to controls (12.8 % and 18.2% vs 28.2 %). Postprandial plasma active ghrelin decreased in lean and obese PCOS groups (49.9 % and 44.1 %) and controls (63.8 %). PCOS subjects exhibited smaller rises in postprandial levels of total PYY. Postprandial plasma PYY(3-36) levels increased in obese PCOS women (30.9 %) and controls (41%), whereas lean PCOS women exhibited blunted increase (11.5%). sCD40L levels increased, whereas adiponectin decreased in PCOS groups independently, whereas rise in visfatin, sE- and sP-selectin and the fall in adiponectin was associated with obesity. sP- and sE -selectins correlated positively with obesity. In summary, our study provides the first evidence that lean untreated young PCOS women contribute to the so called "pancreatic islet adaptation to insulin resistance" because of ghrelin and PYY profiles. We confirmed existing of low-grade chronic inflammation in early stage of visceral obesity in lean PCOS patients. The lost endogenous "islet adaptation to insulin resistance" may lead to endothelial dysfunction and promote acceleration of atherosclerosis.
Article
Full-text available
Insulin resistance is a central feature of the PCOS and may increase cardiovascular risk. Owing to insulin resistance, the metabolic syndrome is more prevalent in women with PCOS compared with unaffected women. Metformin improves the metabolic profile in PCOS in short-term studies. In this study, we evaluated the long-term effect of metformin on metabolic parameters in women with PCOS during routine care without a controlled diet.We performed a retrospective medical chart review of 70 women with PCOS receiving metformin from an academic endocrine clinic. Metabolic risk factors were compared before and after metformin treatment. Time trends of these metabolic parameters were also analysed. After a mean follow-up of 36.1 months with metformin treatment, improvements were observed for BMI (-1.09 +/- 3.48 kg/m2, p = 0.0117), diastolic blood pressure (-2.69 +/- 10.35 mmHg, p = 0.0378), and HDL cholesterol (+5.82 +/- 11.02 mg/dL, p <0.0001).The prevalence of metabolic syndrome decreased from 34.3% at baseline to 21.4% (p = 0.0495). The course of BMI reduction during metformin treatment was significantly more pronounced in women with PCOS with metabolic syndrome at baseline, compared with those without the metabolic syndrome (p = 0.0369 for interaction). In conclusion, metformin improved the metabolic profile of women with PCOS over 36.1 months, particularly in HDL cholesterol, diastolic blood pressure and BMI.
Article
Full-text available
Adiposopathy is defined as pathological adipose tissue function that may be promoted and exacerbated by fat accumulation (adiposity) and sedentary lifestyle in genetically susceptible patients. Adiposopathy is a root cause of some of the most common metabolic diseases observed in clinical practice, including Type 2 diabetes mellitus, hypertension and dyslipidemia. The most common term for the metabolic consequences of adiposopathy is currently 'the metabolic syndrome'. Drug usage to treat the metabolic syndrome has focused on the safety and efficacy of treatments directed towards individual components of the metabolic syndrome, and not so much upon adiposopathy itself. However, enough is known about the pathophysiology of adiposopathy to propose diagnostic criteria. Regulatory issues are important obstacles to the research and development of new drug treatments for the metabolic syndrome. It is hoped that these obstacles can, to some extent, be addressed and overcome by clearly defining and increasing our understanding of adiposopathy.
Article
Full-text available
BACKGROUND Polycystic ovary syndrome (PCOS) is a common condition in women of reproductive age with well established metabolic abnormalities. There are numerous diagnostic criteria generating several reproductive diagnostic phenotypes [National Institute of Health (NIH) hyperandrogenic anovulatory PCOS and non-NIH PCOS including hyperandrogenic ovulatory or non-hyperandrogenic anovulatory PCOS]. There is ongoing debate regarding the optimal diagnostic criteria for PCOS and on the metabolic implications of newer non-NIH PCOS phenotypes. METHODS We reviewed the literature on the presence of risk factors for type 2 diabetes (DM2) and cardiovascular disease (CVD) across the reproductive diagnostic phenotypes of PCOS with the aims of comparing the metabolic features of the NIH and non-NIH groups and identifying potential high metabolic risk phenotypes of PCOS. RESULTS NIH PCOS patients present with greater obesity, abdominal obesity, insulin resistance (IR) and risk factors for DM2 and CVD compared with non-NIH ovulatory and non-hyperandrogenic PCOS patients. Where differences in metabolic features exist between the phenotypes, they are generally related to the degree of total and abdominal obesity. There is emerging evidence suggesting ovulatory and non-hyperandrogenic PCOS have greater metabolic abnormalities than controls primarily linked to abdominal adiposity. There is currently no evidence that non-hyperandrogenic PCOS is associated with a less adverse metabolic profile than ovulatory PCOS. CONCLUSIONS Current metabolic evidence appears to justify the inclusion of both non-NIH PCOS groups (ovulatory and non-hyperandrogenic) as PCOS subgroups. NIH PCOS is associated with a more adverse metabolic profile including greater total and abdominal obesity, IR and risk factors for CVD and DM2 than non-NIH phenotypes.
Article
Full-text available
It is believed that brain derived neurotrophic factor (BDNF) plays an important role in neuronal growth, transmission, modulation and plasticity. Single bout of exercise can increase plasma BDNF concentration [BDNF](p) in humans. It was recently reported however, that elevated [BDNF](p) positively correlated with risk factors for metabolic syndrome and type 2 diabetes mellitus in middle age group of subjects. On the other hand it is well established that endurance training decreases the risk of diabetes and development of metabolic syndrome. In the present study we have examined the effect of 5 weeks of moderate intensity endurance training on the basal and the exercise induced changes in [BDNF](p) in humans. Thirteen young, healthy and physically active men (mean +/- S.E: age 22.7 +/- 0.5 yr, body height 180.2 +/- 1.7 cm, body weight 77.0 +/- 2.5 kg, V(O2max) 45.29 +/- 0.93 ml x kg-1 x min(-1)) performed a five week endurance cycling training program, composed mainly of moderate intensity bouts. Before training [BDNF]p at rest have amounted to 10.3 +/- 1.4 pg x ml(-1). No effect of a single maximal incremental cycling up to V(O2max) on its concentration was found (10.9 +/- 2.3 pg x ml(-1), P=0.74). The training resulted in a significant (P=0.01) increase in [BDNF]p at rest to 16.8 +/- 2.1 pg x ml(-1), as well as in significant (P=0.0002) exercise induced increase in the [BDNF](p) (10.9 +/- 2.3 pg x ml(-1) before training vs. 68.4 +/- 16.0 pg x ml(-1) after training). The training induced increase in resting [BDNF](p) was accompanied by a slight decrease in insulin resistance (P=0.25), calculated using the homeostatic model assessment version 2 (HOMA2-IR), amounting to 1.40 +/- 0.13 before and 1.15 +/- 0.13 after the training. Moreover, we have found that the basal [BDNF](p) in athletes (n=16) was significantly higher than in untrained subjects (n=13) (29.5 +/- 9.5 pg x ml(-1) vs. 10.3 +/- 1.4 pg x ml(-1), P=0.013). We have concluded that endurance training of moderate intensity increases both basal as well as the end-exercise [BDNF](p) in young healthy men. This adaptive response, contrariwise to the recent findings in patients with metabolic disorders, was accompanied by a slight decrease in insulin resistance.
Article
Background The term ‘metabolic syndrome’ refers to a clustering of specific cardiovascular disease (CVD) risk factors whose underlying pathophysiology is thought to be related to insulin resistance. Methods Since the term is widely used in research and clinical practice, we undertook an extensive review of the literature in relation to the syndrome’s definition, underlying pathogenesis, association with cardiovascular disease and to the goals and impact of treatment. Discussion While there is no question that certain CVD risk factors are prone to cluster, we found that the metabolic syndrome has been imprecisely defined, there is a lack of certainty regarding its pathogenesis, and there is considerable doubt regarding its value as a CVD risk marker. Our analysis indicates that too much critically important information is missing to warrant its designation as a ‘syndrome’. Conclusion Until much-needed research is completed, clinicians should evaluate and treat all CVD risk factors without regard to whether a patient meets the criteria for diagnosis of the ‘metabolic syndrome’.
Article
Resistance to insulin-stimulated glucose uptake is present in the majority of patients with impaired glucose tolerance (IGT) or non-insulin-dependent diabetes mellitus (NIDDM) and in ∼25% of nonobese individuals with normal oral glucose tolerance. In these conditions, deterioration of glucose tolerance can only be prevented if the β-cell is able to increase its insulin secretory response and maintain a state of chronic hyperinsulinemia. When this goal cannot be achieved, gross decompensation of glucose homeostasis occurs. The relationship between insulin resistance, plasma insulin level, and glucose intolerance is mediated to a significant degree by changes in ambient plasma free-fatty acid (FFA) concentration. Patients with NIDDM are also resistant to insulin suppression of plasma FFA concentration, but plasma FFA concentrations can be reduced by relatively small increments in insulin concentration.Consequently, elevations of circulating plasma FFA concentration can be prevented if large amounts of insulin can be secreted. If hyperinsulinemia cannot be maintained, plasma FFA concentration will not be suppressed normally, and the resulting increase in plasma FFA concentration will lead to increased hepatic glucose production. Because these events take place in individuals who are quite resistant to insulinstimulated glucose uptake, it is apparent that even small increases in hepatic glucose production are likely to lead to significant fasting hyperglycemia under these conditions. Although hyperinsulinemia may prevent frank decompensation of glucose homeostasis in insulin-resistant individuals, this compensatory response of the endocrine pancreas is not without its price. Patients with hypertension, treated or untreated, are insulin resistant, hyperglycemic, and hyperinsulinemic. In addition, a direct relationship between plasma insulin concentration and blood pressure has been noted. Hypertension can also be produced in normal rats when they are fed a fructose-enriched diet, an intervention that also leads to the development of insulin resistance and hyperinsulinemia. The development of hypertension in normal rats by an experimental manipulation known to induce insulin resistance and hyperinsulinemia provides further support for the view that the relationship between the three variables may be a causal one. However, even if insulin resistance and hyperinsulinemia are not involved in the etiology of hypertension, it is likely that the increased risk of coronary artery disease (CAD) in patients with hypertension and the fact that this risk if not reduced with antihypertensive treatment are due to the clustering of risk factors for CAD, in addition to high blood pressure, associated with insulin resistance. These include hyperinsulinemia, IGT, increased plasma triglyceride concentration, and decreased high-density lipoprotein cholesterol concentration, all of which are associated with increased risk for CAD. It is likely that the same risk factors play a significant role in the genesis of CAD in the population as a whole. Based on these considerations the possibility is raised that resistance to insulin-stimulated glucose uptake and hyperinsulinemia are involved in the etiology and clinical course of three major related diseases— NIDDM, hypertension, and CAD.
Article
LR: 20061115; JID: 7501160; 0 (Antilipemic Agents); 0 (Cholesterol, HDL); 0 (Cholesterol, LDL); 57-88-5 (Cholesterol); CIN: JAMA. 2001 Nov 21;286(19):2401; author reply 2401-2. PMID: 11712930; CIN: JAMA. 2001 Nov 21;286(19):2400-1; author reply 2401-2. PMID: 11712929; CIN: JAMA. 2001 Nov 21;286(19):2400; author reply 2401-2. PMID: 11712928; CIN: JAMA. 2001 Nov 21;286(19):2400; author reply 2401-2. PMID: 11712927; CIN: JAMA. 2001 May 16;285(19):2508-9. PMID: 11368705; CIN: JAMA. 2003 Apr 16;289(15):1928; author reply 1929. PMID: 12697793; CIN: JAMA. 2001 Aug 1;286(5):533-5. PMID: 11476650; CIN: JAMA. 2001 Nov 21;286(19):2401-2. PMID: 11712931; ppublish