ArticlePDF Available

Age-Related Hypercholesterolemia and HMG-CoA Reductase Dysregulation: Sex Does Matter (A Gender Perspective)

Wiley
Current Gerontology and Geriatrics Research
Authors:

Abstract and Figures

Although cardiovascular diseases are less prevalent in premenopausal women than in men, their occurrence in women increases at the onset of menopause, and the loss of female sex hormones contributes to the striking increase in cardiovascular morbidity and mortality in postmenopausal women. We present here a description of age-related disruption of lipid homeostasis, which particularly affects 3-hydroxy 3-methylglutaryl Coenzyme A reductase, the key rate-limiting enzyme in the cholesterol biosynthetic pathway. We further discuss the age- and gender-related dysregulation of this enzyme, providing new evidence for the different mechanisms driving dyslipidemia in elderly men and women. In addition, we introduce pharmacological methods of regulating HMGR and maintaining cholesterol homeostasis.
Content may be subject to copyright.
Hindawi Publishing Corporation
Current Gerontology and Geriatrics Research
Volume 2010, Article ID 420139, 7pages
doi:10.1155/2010/420139
Review Article
Age-Related Hypercholesterolemia and HMG-CoA Reductase
Dysregulation: Sex Does Matter (A Gender Perspective)
Laura Trapani and Valentina Pallottini
Department of Biology, University of Roma Tre, Viale Marconi, 446, 00146 Rome, Italy
Correspondence should be addressed to Valentina Pallottini, vpallott@uniroma3.it
Received 29 October 2009; Revised 26 January 2010; Accepted 18 February 2010
Academic Editor: Jacek Witkowski
Copyright © 2010 L. Trapani and V. Pallottini. This is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
cited.
Although cardiovascular diseases are less prevalent in premenopausal women than in men, their occurrence in women increases
at the onset of menopause, and the loss of female sex hormones contributes to the striking increase in cardiovascular morbidity
and mortality in postmenopausal women. We present here a description of age-related disruption of lipid homeostasis, which
particularly aects 3-hydroxy 3-methylglutaryl Coenzyme A reductase, the key rate-limiting enzyme in the cholesterol biosynthetic
pathway. We further discuss the age- and gender-related dysregulation of this enzyme, providing new evidence for the dierent
mechanisms driving dyslipidemia in elderly men and women. In addition, we introduce pharmacological methods of regulating
HMGR and maintaining cholesterol homeostasis.
1. Introduction
Aging has been defined as the series of the deteriorative
changes occurring during the adult period of life that
underlie increased vulnerability to challenges and decreased
survival [1]. This deterioration is responsible for both the
commonly recognized sequential changes that accompany
advancing age and the progressive increase in the chance of
disease and death and is usually manifested as a progressive
decrease in physiological functions.
Aging is characterized by the loss of homeostasis [2]
that leads to changes in the biochemical composition
of tissues [35], reduced ability to respond adaptively
to environmental stimuli [6], and increased susceptibility
and vulnerability to diseases [7] including coronary artery
diseases (CAD). The term CAD refers to pathologic changes
within the coronary artery walls that result in diminished
blood flow through these vessels. CAD can cause myocardial
ischemia and possibly lead to acute myocardial infarction
through three mechanisms—profound vascular spasm of the
coronary arteries, formation of atherosclerotic plaques, and
thromboembolism.
Although it is widely accepted that abnormal levels of
lipids and/or lipoproteins in blood are modifiable risk factors
for CAD [8,9], the importance of lipid levels as prognostic
factors in older adults is controversial. Several studies have
suggested that the association between cholesterol concen-
tration and atherosclerotic CAD weakens with age, and that
screening and treating older adults for dyslipidemia provides
little potential benefit [10,11]. In contrast, other reports
suggest that lipoprotein levels remain a significant risk factor
for CAD in the elderly and that treatment of dyslipidemia in
the elderly may have a greater impact on CAD mortality than
in younger people because the total attributable risk from
dyslipidemia is greater in the older age group [12,13].
The mechanisms behind this age-related dyslipidemia
are incompletely characterized. Some evidence demonstrates
that the causes of age-related disruption of lipid homeostasis
include the gradual decline in fractional clearance of LDL
with increasing age, the progressively reduced ability to
remove cholesterol through conversion to bile acids, and the
decreased activity of the rate-limiting enzyme in bile acid
biosynthesis, cholesterol 7α-hydroxylase (C7αOH). More-
over, an interesting hypothesis states that critical changes
in cholesterol and lipoprotein metabolism depend on the
progressive decrease in growth hormone (GH) secretion, a
characteristic feature of aging. GH plays an important role in
cholesterol homeostasis by either modulating the expression
of hepatic LDLr [14] or controlling the activity of cholesterol
7α-hydroxylase [15].
2 Current Gerontology and Geriatrics Research
Cholesterol
Heme A Isoprenylated
proteins Dolichol
2,3 -Oxiydosqualene
Squalene Geranylgeranyl-PP
Farnesyl-PP
Geranyl-PP
Isopentenyl-PP
Mevalonate
HMG CoA
Acetyl CoA
HMGR
Figure 1: Schematic illustration of the biosynthetic pathway of HMGR end-products.
CAD is less prevalent in premenopausal women than in
men, but its occurrence in women increases at the onset of
menopause, and the loss of female sex hormones contributes
to the striking increase in the incidence of cardiovascular
morbidity and mortality in postmenopausal women. Estro-
gen replacement therapy results in improved lipoprotein
profiles in postmenopausal women, but these improved
profiles account for less than half of the cardioprotective
eects of estrogen replacement therapy. The cardioprotective
eects of estrogens include beneficial changes in plasma lipid
levels (increased high-density lipoprotein (HDL), decreased
total cholesterol and low-density lipoprotein (LDL), and
decreased LDL oxidation) [16,17], antiplatelet and antiox-
idant eects, and preservation of endothelium-mediated
vasodilation [18,19]. Providing further evidence for the car-
dioprotective role of estrogen, studies performed in estrogen-
deficient animal models have demonstrated a disruption of
lipid homeostasis [20].
Cholesterol plays an essential role in the synthesis of
new membranes, the turnover of lipids in existing mem-
branes, and the biosynthesis of products such as steroid
hormones and bile acids [21]. Cholesterol homeostasis is
maintained by a feedback regulatory system that senses the
level of cholesterol in cell membranes and modulates both
the transcription of genes encoding proteins involved in
cholesterol biosynthesis and posttranscriptional events along
with the uptake of cholesterol from plasma lipoproteins
[22]. Maintenance of cholesterol homeostasis is regulated
by both the receptor-mediated endocytosis of LDL by LDL
receptors (LDLr) and de novo cholesterol synthesis via the
rate-limiting enzyme 3-hydroxy-3-methylglutaryl coenzyme
A reductase (HMGR) [23]. Because of the pivotal role of
HMGR in cholesterol and nonsterol isoprenoid compound
biosynthesis, most of the mechanisms controlling cholesterol
homeostasis are related to short- and long-term regulation of
HMGR.
To provide new evidence for the dierent mechanisms
driving dyslipidemia in elderly men and women, this review
will focus on age-related disruption of lipid homeostasis, and
in particular on the age- and gender-related dysregulation
of HMGR, the key rate-limiting enzyme in the cholesterol
biosynthetic pathway.
2. HMGR Regulation in Adults
Cholesterol biosynthesis occurs through a tightly regulated
pathway that employs multiple feedback mechanisms to
maintain homeostasis [24].Over the past several decades,
much work has focused on the regulation of HMGR, which
catalyzes the conversion of HMG-CoA to mevalonate (MVA)
through a four-electron oxidoreduction. This reaction is
the rate-limiting step in the synthesis of cholesterol and
other isoprenoids such as dolichol, isopentenyladenine,
which is present in some tRNAs, heme A, ubiquinone, and
prenylated proteins such as Ras and Rab proteins (Figure 1)
[24].
Encoded by the HMGR gene located on chromosome
5 of human genome, HMGR consists of a single 888
amino acid polypeptide chain. The N-terminal membrane
domain contains 339 hydrophobic residues that span the
endoplasmic reticulum (ER) membrane and contains the
sterol-sensing domain (SSD), which is responsible for the
binding of sterols and other MVA derivatives that accelerate
enzyme degradation [25], while the catalytic site is located in
the hydro-soluble C-terminal cytoplasmic domain. A linker
region (residues 340–459) connects these two portions of the
protein [2].
Current Gerontology and Geriatrics Research 3
Short-term regulation of HMGR is achieved through its
phosphorylation and dephosphorylation, both of which can
aect enzyme activity. Phosphorylation of residue S872 of
HMGR decreases its catalytic activity, and removal of this
phosphate results in reactivation [26,27]. AMP-activated
kinase (AMPK) appears to be the major HMGR kinase
in the liver, where cholesterologenesis takes place. AMPK
is a heterotrimeric serine/threonine kinase consisting of a
catalytic αsubunit and regulatory βand γsubunits [28].
AMPK is activated by phosphorylation of the αsubunit at a
specific threonine residue (Thr172) [29]. HMGR activation
is mediated by its dephosphorylation by protein phosphatase
2A (PP2A), which regulates a significant network of cellular
events [30].
In addition to this short-term regulation, HMGR is sub-
ject to transcriptional, translational, and posttranslational
control [31].Theselevelsofcontrol,whicharemediatedby
changes in intracellular sterol levels and cholesterol uptake by
LDLr, can result in changes of over 200-fold in HMGR levels
[32]. Both LDLr and HMGR are produced in response to
activation of the Sterol Regulatory Element Binding Proteins
(SREBPs), and particularly SREBP-2, in the liver [33,34].
Long-term regulation of HMGR is mediated by a pair
of membrane-bound proteins, SREBP cleavage activating
protein (Scap) and Insulin-induced gene (Insig), which
directly bind sterols and thereby sense sterol concentration in
the membranes of the ER. As a result of these binding events,
both Scap and Insig undergo conformational changes that
initiate a series of molecular events blocking Scap’s ability
to transport SREBPs to the Golgi, terminating cholesterol
synthesis and uptake [32]. Furthermore, the intracellular
accumulation of sterols induces HMGR to bind Insig,
promoting ubiquitination and proteasomal degradation of
HMGR [35].
Several hormones, including insulin, glucagon, gluco-
corticoids, thyroid hormone, and estrogen, regulate the
expression of hepatic HMGR in animals. Insulin likely
stimulates HMGR expression by increasing its rate of
transcription, while glucagon opposes this eect. Hepatic
HMGR activity undergoes significant diurnal variations due
to changes in the levels of immunoreactive proteins, which
are primarily mediated by changes in insulin and glucagon
levels. Thyroid hormone increases hepatic HMGR levels by
acting to increase both transcription and mRNA stability,
while glucocorticoids decrease hepatic HMGR expression by
destabilizing HMGR mRNA [36]. The eects of estrogen
on HMGR expression are still debated. Some studies sug-
gest that estrogens act to increase hepatic HMGR activity
primarily by stabilizing HMGR mRNA and that deficiencies
in those hormones that act to increase hepatic HMGR gene
expression lead to elevated serum cholesterol levels [36]. On
the other hand, studies using the DLD1 cell line suggest that
estrogens induce an early increase in LDLr at both the mRNA
and the protein level and later cause decreases in HMGR
activity and protein expression [37].
Although the mechanisms that regulate cholesterol
homeostasis are well known, [22] the literature describ-
ing putative physiological sex dierences in cholesterol
homeostasis-related proteins is limited [3842]. Further-
more, most of these papers are fragmented and very old,
and none of them focus on the mechanisms underlying
these sex-related dierences. De Marinis and coworkers [43]
provided evidence of sex-related physiological dierences in
proteins involved in cholesterol homeostasis. In particular,
activity and expression levels of HMGR are lower in 3-
month-old female rats and in 17-β-estradiol-treated 3-
month-old male rats than in 3-month-old untreated male
rats. Moreover, 3-month-old female rats express lower levels
of SREBP-2 and higher levels of Insig than their male
counterparts. Sex-related variations in expression of these
regulatory proteins are functionally consistent with the well-
accepted classical model of HMGR behavior [22,32], and
no sex-related dierences have been observed in either LDLr
expression or cholesterol levels, excluding the involvement of
end-product feedback in presence of physiological content
of estradiol. The dierence in the expression pattern of
regulatory proteins in males and females seems to be related
to the presence of estrogen, and altered expression of these
regulatory proteins drives the sex-related dierences in
HMGR expression.
2.1. Sex-Related Dierences in HMGR Dysregulation during
Aging. Due to the serious health-related consequences of
aging, significant eorts have been made to provide a
more complete understanding of this particular stage of
life. Current research aiming to delineate the biological
mechanisms of aging has yielded valuable information about
the molecular basis of age-related physiological deteriora-
tion. One of the critical problems associated with aging
is the increased incidence of CAD and, more generally,
cardiovascular diseases (CVD). Many risk factors predispose
elderly people to develop pathologies related to failure of
the heart vasculature, including hypercholesterolemia. Thus,
understanding the mechanisms driving increased cellular
and plasma cholesterol content during aging is essential in
defining specific intervention points.
During aging, hepatic lipid modifications occur. In par-
ticular, studies of 24-month-old male rats showed increased
plasma cholesterol levels and increased hepatic cholesterol
synthesis accompanied by full activation of HMGR [44,
45], which was dependent on the well-known age-related
increaseinreactiveoxygenspecies(ROS)[46,47]. The age-
related increase in activation of HMGR has been associated
with an increase in ROS [48,49]. The current model
proposes that increased ROS levels result in activation
of both p38 and AMPKα. In turn, p38 activation may
result in an increase in association of PP2A with HMGR,
leading to dephosphorylation and increased activation of
HMGR. AMPKαkinase activity is impaired by the enhanced
association of PP2A with HMGR [50]. Moreover, findings in
H2O2-stimulated HepG2 cells demonstrate that the eect of
ROS on HMGR dephosphorylation is mediated by activation
of the p38/MAPK pathway [45].
In addition to the short-term regulation mediated by
phosphorylation and dephosphorylation, long-term regu-
lation of HMGR also appears to be aected by aging.
Age-related variations in hormone levels and hormone
4 Current Gerontology and Geriatrics Research
sensitivity induce a decreased ability to maintain homeostatic
potential, and these hormonal changes are always associated
with changes in the expression or functionality of some
molecules. In particular, it has been clearly demonstrated
that the age-related decrease in insulin sensitivity induces
changes in some factors involved in cholesterol metabolism,
such as Insig-1 protein. This age-related reduction in Insig
expression results in slower degradation of HMGR [51,52].
While many studies have established that susceptibility
to CAD increases with age, little is known about the
mechanisms underlying the increased incidence of CAD in
postmenopausal women as compared to men of the same
age.
Previous studies have shown that 12-month-old
estropausal rats, in which estrogen levels are decreased, have
higher levels of plasma cholesterol, increased activation
of HMGR, and decreased LDLr membrane exposure than
3-month-old female rats. These changes result in decreased
cholesterol uptake and increased cholesterol synthesis,
supporting the correlation between hypercholesterolemia,
aging, and estropause. Increased activation of HMGR does
not depend on an increase in ROS as seen in aged-matched
male rats [53]. Instead, HMGR activation seems to be
due to decreased activation of AMPK during the period
of 17-βestradiol deficiency that occurs at the beginning
of estropause; this decrease in AMPK activation results in
decreased phosphorylation of HMGR.
Treatment of older female rats with 17-βestradiol results
in restoration of normal cholesterol levels, decreased activa-
tion of HMGR, and increased LDLr exposure on the plasma
membrane. Furthermore, while 17-βestradiol treatment
does not fully restore AMPK activation, AMPK is suciently
activated in older 17-βestradiol-treated female rats to
phosphorylate HMGR, reestablishing HMGR activity [54].
This estradiol-induced enhancement in AMPK activation
is supported by studies by Schulz and coworkers, who
demonstrated that estradiol-mediated AMPK activation was
independent of estrogen receptor ligand engagement and
involved catechol metabolism of estradiol [55].
The decrease in estradiol levels that occurs at the onset of
estropause does not aect long-term regulation of HMGR,
but mediates short-term HMGR regulation by controlling
activation of AMPK. Thus, a relationship exists between
changes in estrogen levels and HMGR-related modulation
of cholesterolemia in older female rats. The protective
role played by estrogens in modulating the lipid profile
is mediated not only through increases in plasma HDL,
decreases in plasma LDL, and decreased oxidation, but
also through regulation of AMPK activation, which inhibits
HMGR and cholesterol synthesis.
3. Conclusion and Future Perspectives
In elderly men and women, HMGR is highly activated;
however, the mechanisms driving dysregulation of HMGR
appear to be gender-dependent. Studies of aged male rats
suggest that in males, HMGR dysregulation is due to
increased association between PP2A and HMGR, which
results in increased activation of HMGR. On the other hand,
studies of estropausal female rats, in which estrogen levels
are decreased, suggest that the menopause-related increase
in HMGR activity is caused by the decreased activation of
AMPK observed during estrogen deficiency.
The regulation of HMGR activity has been an attrac-
tive target for pharmacological treatment of hypercholes-
terolemia, the main risk factor for CAD. Consequently,
better understanding of the molecular mechanisms that drive
dysregulation of HMGR activity and hypercholesterolemia in
aged men and women could provide gender-specific targets
for treatments to lower plasma cholesterol content, resulting
in both prevention and reduction of one of the main risk
factors for cardiovascular diseases.
Decreased cellular cholesterol synthesis leads to a home-
ostatic response involving up-regulation of cell-surface
receptors that bind atherogenic lipoproteins such as LDL
and VLDL. These lipoproteins are taken up by the cell
anddegraded[56], resulting in a reduction in circulating
atherogenic lipoproteins. This process helps to explain the
clinical usefulness of HMGR inhibitors (statins).
HMGR inhibition results in not only reductions in
cellular and plasma cholesterol levels, but also reductions in
other products synthesized through the cholesterol biosyn-
thetic pathway, such as ubiquinone, prenylated proteins, and
dolichol. The restoration of HMGR to its normal activation
state regulates the physiological synthesis of cholesterol
within cells, and restored cellular cholesterol levels are in turn
reflected in proper membrane LDLr presence [22].
Statins are eective means of primary and secondary
prevention of ischemic heart disease (IHD) in middle-aged
men; however, proof of the ecacy of statins in preventing
development and progression of IHD in women and elderly
people is less convincing. In the PROSPER (Prospective
Study of Pravastatin in the Elderly at Risk of vascular disease)
trial, pravastatin not only had no eectinmenandwomen
aged 70–82 years, but also significantly increased the rate
of breast cancer in these patients. In the ALLHAT-LLT
(Antihypertensive and Lipid-Lowering Treatment to Prevent
Heart Attack Trial) trial, pravastatin lowered neither the total
number of nonfatal myocardial infarctions and IHD deaths
nor total mortality in patients aged 65 years and older and in
women [57].
Considering the undesirable side eects of statins and
the eects of these drugs on other important compounds
in addition to cholesterol, development of new pharma-
cologically active compounds capable of regulating plasma
cholesterol content is critical to eectively control this
important CVD risk factor.
Many recent studies have described new compounds able
to decrease plasma cholesterol content. Although the exact
mechanisms by which these compounds act are unknown,
some of these compounds aect expression levels and activity
of HMGR. These novel compounds could thus represent
the future of hypercholesterolemia therapy and should be
studied further.
Hypercholesterolemia can also be approached using
other therapeutic targets; for example, proprotein convertase
subtilisin/kexin type 9 (PCSK9) has been implicated as
Current Gerontology and Geriatrics Research 5
an important regulator of LDL metabolism. PCSK9 belongs
to the subtilisin family of serine proteases and is highly
expressed in the liver [58]. Secreted PCSK9 modulates LDL
levels through posttranslational down-regulation of hepatic
LDLr protein [34]. Down-regulation of PCSK9 could thus
be eective in decreasing plasma cholesterol content by
increasing LDLr levels without aecting activity of HMGR
and its end-products.
In addition, squalene synthase, an enzyme that is
downstream of HMGR in the cholesterol synthesis pathway,
modulates the first committed step of hepatic cholesterol
biosynthesis at the final branching point of the cholesterol
biosynthetic pathway. Pharmacologic inhibitors of squalene
epoxidase and oxidosqualene cyclase, two enzymes that act
downstream of squalene synthase, may thus be useful in
reducing plasma LDL content [59].
Moreover, the identification of Scap and Insig as sterol-
binding proteins in mammalian cells has added a new level
of molecular detail to the understanding of regulation of
the SREBP pathway and subsequent regulation of HMGR
levels, providing new potential targets for pharmacological
intervention.
Much work remains to define the relationship between
hormonal changes and their eects on transcription factors
and cholesterol metabolism in dierent physiological and
pathological conditions. For example, some studies suggest
that estrogens are able to regulate cholesterol homeosta-
sis without directly aecting HMGR [43]. Additionally,
more detailed studies are required to define the specific
roles of Insig proteins and to determine the metabolic
consequences of their reciprocal regulation. In fact, since
they are required for feedback regulation of SREBP pro-
cessing and HMGR degradation, Insigs may represent a
new target for pharmacological intervention to maintain
blood cholesterol levels within the optimal range. None of
the papers we have cited focus on the sex-related dier-
ences.
Thus, although HMGR plays a pivotal role in regulating
cholesterol metabolism, future studies should address sex-
related dierences in the cholesterol biosynthetic pathway
to identify new targets for customized pharmacological
treatment of hypercholesterolemia.
Acknowledgments
The authors wish to thank past and present members of their
laboratories who contributed with data and discussions to
the ideas presented here. In particular, the invaluable and
dedicated work of our mentor Professor Anna Trentalance
is warmly acknowledged.
References
[1] E. J. Masoro, “Overview of caloric restriction and ageing,”
Mechanisms of Ageing and Development, vol. 126, no. 9, pp.
913–922, 2005.
[2] R. G. Cutler, “The dysdierentiative hypothesis of mammalian
ageing and longevity,” in The Ageing Brain, E. Jacobini, et al.,
Ed., Raven Press, New York, NY, USA, 1982.
[3]B.L.Strehler,Time, Cells, and Ageing, Academic Press, New
York, NY, USA, 1977.
[4] J. Bjorksten, “Cross linkage and the ageing process,” in
Theoretical Aspects of Ageing, M. Rothstein, Ed., Academic
Press, New York, NY, USA, 1974.
[5] R. R. Kohn, “Ageing of animals: possible mechanisms,” in
Principles of Mammalian Ageing,R.R.Kohn,Ed.,Prentice-
Hall, Englewood Clis, NJ, USA, 1978.
[6]R.C.Adelman,G.W.Britton,S.Rotenberg,L.Ceci,and
K. Karoly, “Endocrine regulation of enzyme activity in aging
animals of dierent genotypes,” Birth Defects: Original Article
Series, vol. 14, no. 1, pp. 355–364, 1978.
[7] J. A. Brody and D. B. Brock, “Epidemiological and statistical
characteristics of the United States elderly population,” in
Handbook of the Biology of Ageing, C. E. Finch and E. L.
Schneider, Eds., Van Nostrand Reinhold, New York, NY, USA,
1985.
[8] W. H. Ettinger Jr., P. W. Wahl, L. H. Kuller, et al., “Lipoprotein
lipids in older people: results from the Cardiovascular Health
Study. The CHS Collaborative Research Group,Circulation,
vol. 86, no. 3, pp. 858–869, 1992.
[9] W. P. Castelli, “Epidemiology of coronary heart disease: the
Framingham study,American Journal of Medicine, vol. 76, no.
2A, pp. 4–12, 1984.
[10] A. M. Garber, B. Littenberg, H. C. Sox Jr., J. L. Wagner,
and M. Gluck, “Costs and health consequences of cholesterol
screening for asymptomatic older Americans,Archives of
Internal Medicine, vol. 151, no. 6, pp. 1089–1095, 1991.
[11] R. Benfante and D. Reed, “Is elevated serum cholesterol level a
risk factor for coronary heart disease in the elderly?” Journal of
the American Medical Association, vol. 263, no. 3, pp. 393–396,
1990.
[12] E. Barrett-Connor, L. Suarez, K.-T. Khaw, M. H. Criqui, and
D. L. Wingard, “Ischemic heart disease risk factors after age
50,Journal of Chronic Diseases, vol. 37, no. 12, pp. 903–908,
1984.
[13] W. P. Castelli, P. W. F. Wilson, D. Levy, and K. Anderson,
“Cardiovascular risk factors in the elderly,American Journal
of Cardiology, vol. 63, no. 16, pp. H12–H19, 1989.
[14] M. Matasconi, P. Parini, B. Angelin, and M. Rudling, “Pituitary
control of cholesterol metabolism in normal and LDL receptor
knock-out mice: eects of hypophysectomy and growth
hormone treatment,Biochimica et Biophysica Acta, vol. 1736,
no. 3, pp. 221–227, 2005.
[15] M. Rudling, P. Parini, and B. Angelin, “Growth hormone
and bile acid synthesis. Key role for the activity of hepatic
microsomal cholesterol 7alpha-hydroxylase in the rat,Journal
of Clinical Investigation, vol. 99, no. 9, pp. 2239–2245, 1997.
[16] D. F. Skafar, R. Xu, J. Morales, J. Ram, and J. R. Sowers,
“Female sex hormones and cardiovascular disease in women,
Journal of Clinical Endocrinology and Metabolism, vol. 82, no.
12, pp. 3913–3918, 1997.
[17] K. Moorthy, U. C. S. Yadav, A. K. Mantha, et al., “Estradiol and
progesterone treatments change the lipid profile in naturally
menopausal rats from dierent age groups, Biogerontology,
vol. 5, no. 6, pp. 411–419, 2004.
[18] S. R. J. Maxwell, “Women and heart disease,” Basic Research in
Cardiology, vol. 93, supplement 2, pp. 79–84, 1998.
[19] K. Moorthy, D. Sharma, S. F. Basir, and N. Z. Baquer,
Administration of estradiol and progesterone modulate the
activities of antioxidant enzyme and aminotransferases in
naturally menopausal rats,” Experimental Gerontology, vol. 40,
no. 4, pp. 295–302, 2005.
6 Current Gerontology and Geriatrics Research
[20] P. A. Heine, J. A. Taylor, G. A. Iwamoto, D. B. Lubahn,
and P. S. Cooke, “Increased adipose tissue in male and
female estrogen receptor—a knockout mice,Proceedings of
the National Academy of Sciences of the United States of
America, vol. 97, no. 23, pp. 12729–12734, 2000.
[21] C. Martini and V. Pallottini, “Cholesterol: from feeding to gene
regulation,Genes and Nutrition, vol. 2, no. 2, pp. 181–193,
2007.
[22] M. S. Brown and J. L. Goldstein, “A proteolytic pathway
that controls the cholesterol content of membranes, cells, and
blood,Proceedings of the National Academy of Sciences of the
United States of America, vol. 96, no. 20, pp. 11041–11048,
1999.
[23] P. J. Espenshade and A. L. Hughes, “Regulation of sterol
synthesis in eukaryotes,Annual Review of Genetic s, vol. 41,
pp. 401–427, 2007.
[24] J. L. Goldstein and M. S. Brown, “Regulation of the meval-
onate pathway,Nature, vol. 343, no. 6257, pp. 425–430, 1990.
[25] T. Ravid, R. Doolman, R. Avner, D. Harats, and J. Roitel-
man, “The ubiquitin-proteasome pathway mediates the regu-
lated degradation of mammalian 3-hydroxy-3-methylglutaryl-
coenzyme A reductase,Journal of Biological Chemistry, vol.
275, no. 46, pp. 35840–35847, 2000.
[26] Z. H. Beg, J. A. Stonik, and H. B. Brewer Jr., “Phosphorylation
of hepatic 3-hydroxy-3-methylglutaryl coenzyme A reductase
and modulation of its enzymic activity by calcium-activated
and phospholipid-dependent protein kinase,Journal of Bio-
logical Chemistry, vol. 260, no. 3, pp. 1682–1687, 1985.
[27] R. V. Omkumar, B. G. Darnay, and V. W. Rodwell, “Mod-
ulation of Syrian hamster 3-hydroxy-3-methylglutaryl-CoA
reductase activity by phosphorylation. Role of serine 871,”
Journal of Biological Chemistry, vol. 269, no. 9, pp. 6810–6814,
1994.
[28]D.G.Hardie,S.A.Hawley,andJ.W.Scott,“AMP-activated
protein kinase—development of the energy sensor concept,
Journal of Physiology, vol. 574, part 1, pp. 7–15, 2006.
[29] S. A. Hawley, M. Davison, A. Woods, et al., “Characterization
of the AMP-activated protein kinase kinase from rat liver and
identification of threonine 172 as the major site at which
it phosphorylates AMP-activated protein kinase,Journal of
Biological Chemistry, vol. 271, no. 44, pp. 27879–27887, 1996.
[30] V. Janssens and J. Goris, “Protein phosphatase 2A: a highly
regulated family of serine/threonine phosphatases implicated
in cell growth and signalling,Biochemical Journal, vol. 353,
part 3, pp. 417–439, 2001.
[31] F.Xu,S.D.Rychnovsky,J.D.Belani,H.H.Hobbs,J.C.Cohen,
and R. B. Rawson, “Dual roles for cholesterol in mammalian
cells,” Proceedings of the National Academy of Sciences of the
United States of America, vol. 102, no. 41, pp. 14551–14556,
2005.
[32] J. L. Goldstein, R. A. DeBose-Boyd, and M. S. Brown, “Protein
sensors for membrane sterols,Cell, vol. 124, no. 1, pp. 35–36,
2006.
[33] M. S. Brown and J. L. Goldstein, “The SREBP pathway:
regulation of cholesterol metabolism by proteolysis of a
membrane-bound transcription factor,Cell,vol.89,no.3,pp.
331–340, 1997.
[34]J.D.Horton,J.L.Goldstein,andM.S.Brown,“SREBPs:
activators of the complete program of cholesterol and fatty
acid synthesis in the liver,Journal of Clinical Investigation, vol.
109, no. 9, pp. 1125–1131, 2002.
[35] N. Sever, B.-L. Song, D. Yabe, J. L. Goldstein, M. S. Brown,
and R. A. DeBose-Boyd, “Insig-dependent ubiquitination and
degradation of mammalian 3-hydroxy-3-methylglutaryl-CoA
reductase stimulated by sterols and geranylgeraniol,Journal of
Biological Chemistry, vol. 278, no. 52, pp. 52479–52490, 2003.
[36] G. C. Ness and C. M. Chambers, “Feedback and hormonal
regulation of hepatic 3-hydroxyl-3- methylglutaryl coenzyme
A reductase: the concept of cholesterol buering capac-
ity,Proceedings of the Society for Experimental Biology and
Medicine, vol. 224, no. 1, pp. 8–9, 2000.
[37] C. Messa, M. Notarnicola, F. Russo, et al., “Estrogenic
regulation of cholesterol biosynthesis and cell growth in
DLD-1 human colon cancer cells,Scandinavian Journal of
Gastroenterology, vol. 40, no. 12, pp. 1454–1461, 2005.
[38] Y. J. Abul-Hajj, “Stimulation by estradiol benzoate of hepatic
beta-hydroxy-beta-methylglutaryl coenzyme A reductase in
normal and ovariectomized rats,Steroids,vol.31,no.6,pp.
841–847, 1978.
[39]S.E.Carlson,A.D.Mitchell,M.L.Carter,andS.Goldfarb,
“Evidence that physiologic levels of circulating estrogens
and neonatal sex-imprinting modify postpubertal hepatic
microsomal 3-hydroxy-3-methylglutaryl coenzyme A reduc-
tase activity,Biochimica et Biophysica Acta, vol. 633, no. 2, pp.
154–161, 1980.
[40] Y.-S. Choi, S. Goto, I. Ikeda, and M. Sugano, “Age-related
changes in lipid metabolism in rats: the consequence of
moderate food restriction,Biochimica et Biophysica Acta, vol.
963, no. 2, pp. 237–242, 1988.
[41] K. N. Hewitt, W. C. Boon, Y. Murata, M. E. Jones, and E.
R. Simpson, “The aromatase knockout mouse presents with
a sexually dimorphic disruption to cholesterol homeostasis,
Endocrinology, vol. 144, no. 9, pp. 3895–3903, 2003.
[42] M. Kojima and M. Degawa, “Gender-related dierence in
altered gene expression of a sterol regulatory element binding
protein, SREBP-2, by lead nitrate in rats: correlation with
development of hypercholesterolemia,Journal of Applied
Tox i c o lo g y , vol. 26, no. 4, pp. 381–384, 2006.
[43] E. De Marinis, C. Martini, A. Trentalance, and V. Pallottini,
“Sex dierences in hepatic regulation of cholesterol homeosta-
sis,Journal of Endocrinology, vol. 198, no. 3, pp. 635–643,
2008.
[44] M. Marino, V. Pallottini, C. D’Eramo, G. Cavallini, E.
Bergamini, and A. Trentalance, “Agerelated changes of choles-
terol and dolichol biosynthesis in rat liver,Mechanisms of
Ageing and Development, vol. 123, no. 8, pp. 1183–1189, 2002.
[45] V. Pallottini, C. Martini, G. Cavallini, et al., “Age-related
HMG-CoA reductase deregulation depends on ROS-induced
p38 activation,Mechanisms of Ageing and Development, vol.
128, no. 11-12, pp. 688–695, 2007.
[46] D. Harman, “Aging: a theory based on free radical and
radiation chemistry,Journal of gerontology, vol. 11, no. 3, pp.
298–300, 1956.
[47] E. R. Stadtman, “Protein oxidation and ageing,Free Radical
Research, vol. 40, no. 12, pp. 1250–1258, 2006.
[48] V. Pallottini, C. Martini, A. Pascolini, et al., “3-hydroxy-3-
methylglutaryl coenzyme A reductase deregulation and age-
related hypercholesterolemia: a new role for ROS,Mecha-
nisms of Ageing and Development, vol. 126, no. 8, pp. 845–851,
2005.
[49] V. Pallottini, C. Martini, A. M. Bassi, P. Romano, G. Nanni,
and A. Trentalance, “Rat HMG CoA reductase activation in
thioacetamide-induced liver injury is related to an increased
reactive oxygen species content,Journal of Hepatology, vol. 44,
no. 2, pp. 368–374, 2006.
Current Gerontology and Geriatrics Research 7
[50] C. Martini, V. Pallottini, G. Cavallini, A. Donati, E. Bergamini,
and A. Trentalance, “Caloric restrictions aect some factors
involved in age-related hypercholesterolemia,Journal of Cel-
lular Biochemistry, vol. 101, no. 1, pp. 235–243, 2007.
[51] V. Pallottini, L. Montanari, G. Cavallini, E. Bergamini, Z. Gori,
and A. Trentalance, “Mechanisms underlying the impaired
regulation of 3-hydroxy-3-methylglutaryl coenzyme A reduc-
tase in aged rat liver,Mechanisms of Ageing and Development,
vol. 125, no. 9, pp. 633–639, 2004.
[52] V. Pallottini, C. Martini, G. Cavallini, et al., “Modified HMG-
CoA reductase and LDLr regulation is deeply involved in age-
related hypercholesterolemia,Journal of Cellular Biochem-
istry, vol. 98, no. 5, pp. 1044–1053, 2006.
[53] S. Straniero, G. Cavallini, A. Donati, et al., “Stimulation of
autophagy by antilipolytic drugs may rescue rodents from age-
associated hypercholesterolemia,Rejuvenation Research, vol.
12, no. 2, pp. 77–84, 2009.
[54] L. Trapani, F. Violo, and V. Pallottini, “Hypercholesterolemia
and 3-hydroxy-3-methylglutaryl coenzyme A reductase regu-
lation in aged female rats,Experimental Gerontology, vol. 45,
no. 2, pp. 119–128, 2010.
[55] E. Schulz, E. Anter, M. H. Zou, and J. F. Keaney Jr., “Estradiol-
mediated endothelial nitric oxide synthase association with
heat shock protein 90 requires adenosine monophosphatede-
pendent protein kinase,Circulation, vol. 111, no. 25, pp. 473–
480, 2005.
[56] M. S. Brown and J. L. Goldstein, “Cholesterol feedback: from
Schoenheimers bottle to Scaps MELADL,Journal of Lipid
Research, vol. 50, pp. S15–S27, 2009.
[57] D. V. Preobrazhenski˘
ı, B. A. Sidorenko, S. A. Pataraia, I. D.
Vyshinskaia, and O. V. Borisenko, “Hypercholesterolemia in
men and women of various age—part II: the problem of
ecacy and safety of statins,Kardiologiia, vol. 47, no. 11, pp.
75–85, 2007.
[58]J.C.Y.Chan,D.E.Piper,Q.Cao,etal.,“Apropro-
tein convertase subtilisin/kexin type 9 neutralizing antibody
reduces serum cholesterol in mice and nonhuman primates,
Proceedings of the National Academy of Sciences of the United
States of America, vol. 106, no. 24, pp. 9820–9825, 2009.
[59] S. Seiki and W. H. Frishman, “Pharmacologic inhibition of
squalene synthase and other downstream enzymes of the
cholesterol synthesis pathway: a new therapeutic approach to
treatment of hypercholesterolemia,Cardiology in Review, vol.
17, no. 2, pp. 70–76, 2009.
... Such sex disparity was also observed in a study of Taiwanese CHD patients in which women taking stains were less likely to achieve < 160 mg/dL of TC levels compared with men taking equivalent dose of stains 35 . One potential explanation relates to estrogens, which have been suggested to protect women from CVD 36 . CVD is less prevalent in premenopausal women than men and women experience an increased rate of CVD after the onset of menopause, with estrogen replacement therapy resulting in improved blood lipid profiles in postmenopausal women 36 . ...
... One potential explanation relates to estrogens, which have been suggested to protect women from CVD 36 . CVD is less prevalent in premenopausal women than men and women experience an increased rate of CVD after the onset of menopause, with estrogen replacement therapy resulting in improved blood lipid profiles in postmenopausal women 36 . Although the mechanism underlying cholesterol-lowering effect of estrogen remains elusive, estrogens have been reported to increase cholesterol clearance via increasing LDL receptors and to decrease cholesterol synthesis via inhibiting HMGR 36 . ...
... CVD is less prevalent in premenopausal women than men and women experience an increased rate of CVD after the onset of menopause, with estrogen replacement therapy resulting in improved blood lipid profiles in postmenopausal women 36 . Although the mechanism underlying cholesterol-lowering effect of estrogen remains elusive, estrogens have been reported to increase cholesterol clearance via increasing LDL receptors and to decrease cholesterol synthesis via inhibiting HMGR 36 . In an experimental study, HMGR activity and expression were lower in female rats and in 17-β-estradiol treated male rats than in male rats 37 . ...
Article
Full-text available
Despite many diabetic patients having hypercholesterolemia, the association of total cholesterol (TC) levels with CVD risk in type 2 diabetes (T2D) patients is unclear. Diagnosis of type 2 diabetes often leads to changes in total cholesterol (TC) levels. Thus, we examined whether changes in TC levels from pre- to post-diagnosis of T2D were associated with CVD risk. From the National Health Insurance Service Cohort, 23,821 individuals diagnosed with T2D from 2003 to 2012 were followed-up for non-fatal CVD incidence through 2015. Two measurements of TC, 2 years before and after T2D diagnosis, were classified into 3 levels (low, middle, high) to define changes in cholesterol levels. Cox proportional hazards regression was performed to estimate adjusted hazards ratios (aHRs) and 95% confidence intervals (CIs) for the associations between changes in cholesterol levels and CVD risk. Subgroup analyses were performed by use of lipid-lowering drugs. Compared with low–low, aHR of CVD was 1.31 [1.10–1.56] for low–middle and 1.80 [1.15–2.83] for low–high. Compared with middle–middle, aHR of CVD was 1.10 [0.92–1.31] for middle–high but 0.83 [0.73–0.94] for middle–low. Compared with high–high, aHR of CVD was 0.68 [0.56–0.83] for high–middle and 0.65 [0.49–0.86] for high–low. The associations were observed regardless of use of lipid-lowering drugs. For diabetic patients, management of TC levels may be important to lower CVD risk.
... In the case of women with hypercholesterolemia, there are well-known factors such as age (85,86), pork rind (87,88), mayonnaise consumption (89,90), and BMI (91,92). Other lessknown predictors emerge from our study. ...
Article
Full-text available
Introduction Mexico ranks second in the global prevalence of obesity in the adult population, which increases the probability of developing dyslipidemia. Dyslipidemia is closely related to cardiovascular diseases, which are the leading cause of death in the country. Therefore, developing tools that facilitate the prediction of dyslipidemias is essential for prevention and early treatment. Methods In this study, we utilized a dataset from a Mexico City cohort consisting of 2,621 participants, men and women aged between 20 and 50 years, with and without some type of dyslipidemia. Our primary objective was to identify potential factors associated with different types of dyslipidemia in both men and women. Machine learning algorithms were employed to achieve this goal. To facilitate feature selection, we applied the Variable Importance Measures (VIM) of Random Forest (RF), XGBoost, and Gradient Boosting Machine (GBM). Additionally, to address class imbalance, we employed Synthetic Minority Over-sampling Technique (SMOTE) for dataset resampling. The dataset encompassed anthropometric measurements, biochemical tests, dietary intake, family health history, and other health parameters, including smoking habits, alcohol consumption, quality of sleep, and physical activity. Results Our results revealed that the VIM algorithm of RF yielded the most optimal subset of attributes, closely followed by GBM, achieving a balanced accuracy of up to 80%. The selection of the best subset of attributes was based on the comparative performance of classifiers, evaluated through balanced accuracy, sensitivity, and specificity metrics. Discussion The top five features contributing to an increased risk of various types of dyslipidemia were identified through the machine learning technique. These features include body mass index, elevated uric acid levels, age, sleep disorders, and anxiety. The findings of this study shed light on significant factors that play a role in dyslipidemia development, aiding in the early identification, prevention, and treatment of this condition.
... The level of bioavailable estrogen is therefore important for normal physiological functioning of the liver and disruption of this signaling axis can have profound effects in both men and women (summarized Figure 3A,B). For example, lower levels of circulating estrogens found in postmenopausal women and in men are associated with an increase in the levels of plasma cholesterol and low-density lipoprotein (LDL) promoting fat accumulation and altering lipid homeostasis in the liver [99][100][101]. It has been shown that GPER1 is more important in aging males [85,86], whereas plasma ERα is more important in female lipid regulation [102,103] (Table 1). The estrogen pathway also plays a role in liver glucose metabolism and homeostasis [104] (Table 1), regulating insulin release, expression of the glucose transporter (GLUT) gene and glycogen synthesis [104][105][106]. ...
Article
Full-text available
The liver performs a multitude of bodily functions, whilst retaining the ability to regenerate damaged tissue. In this review, we discuss sex steroid biology, regulation of mammalian liver physiology and the development of new model systems to improve our understanding of liver biology in health and disease. A major risk factor for the development of liver disease is hepatic fibrosis. Key drivers of this process are metabolic dysfunction and pathologic activation of the immune system. Although non-alcoholic fatty liver disease (NAFLD) is largely regarded as benign, it does progress to non-alcoholic steatohepatitis in a subset of patients, increasing their risk of developing cirrhosis and hepatocellular carcinoma. NAFLD susceptibility varies across the population, with obesity and insulin resistance playing a strong role in the disease development. Additionally, sex and age have been identified as important risk factors. In addition to the regulation of liver biochemistry, sex hormones also regulate the immune system, with sexual dimorphism described for both innate and adaptive immune responses. Therefore, sex differences in liver metabolism, immunity and their interplay are important factors to consider when designing, studying and developing therapeutic strategies to treat human liver disease. The purpose of this review is to provide the reader with a general overview of sex steroid biology and their regulation of mammalian liver physiology.
... Moreover, statins inhibition to HMGCR reduces androgen production and their use at the time of androgen deprivation is associated with delayed progression of PC [13]. Previous studies observed that long-term HMGCR regulation may be affected by aging; due to agerelated changes in hormone levels and sensitivity [28][29][30], justifying the current observed association with age. Heemers and colleagues proved that androgens activate SREBPs to promote de novo lipogenesis and lipid uptake [31]. ...
Article
Full-text available
Aim The role of surgical castration and rosuvastatin treatment on lipid profile and lipid metabolism related markers was evaluated for their prognostic significance in metastatic prostate cancer (mPC) patients. Methods A total of 84 newly diagnosed castrated mPC patients treated with castration were recruited and divided into two groups: Group I served as control (statin non-users) while group II treated with Rosuvastatin (20 mg/day) for 6 months and served as statin users. Prostate specific antigen (PSA), epidermal growth factor receptor (EGFR), Caveolin-1 (CAV1), lipid profile (LDL, HDL, triglycerides (TG) and total cholesterol (TC)) and lipid metabolism related markers (aldoketoreductase (AKR1C4), HMG-CoA reductase (HMGCR), ATP-binding cassette transporter A1 (ABCA1), and soluble low density lipoprotein receptor related protein 1 (SLDLRP1)) were measured at baseline, after 3 and 6 months. Overall survival (OS) was analyzed by Kaplan-Meier and COX regression for prognostic significance. Results Before castration, HMG-CoA reductase was elevated in patients 3385 pg/ml, P = 0.001), PSA (>40 ng/ml, P = 0.003) and CAV1 (>4955 pg/ml, P = 0.021). Conclusion Results of the current study suggest that the peripheral lipidogenic effects of rosuvastatin may have an impact on the treatment outcome and survival of castrated mPC patients. Trail registration This trial was registered at the Pan African Clinical Trial Registry with identification number PACTR202102664354163 and at ClinicalTrials.gov with identification number NCT04776889.
... HMG-COA reductase represents the rate limiting step of cholesterol and isoprenoid synthesis [33]. LYC is a polyisoprenoid, potent antioxidant as compared to other carotenoid members which is synthesized in plant from mevalonate via HMG-COA reductase [34]. ...
Article
Aim The present study aimed mainly to demonstrate the metabolic effects of lycopene (LYC) or atorvastatin (ATOR) in diabetic hyperlipidemic rat model. Main methods Rats were randomly classified into four groups; the first was fed normal chow diet (NC) while the other three groups received streptozotocin (STZ) along with CCT-diet. The second group received no treatment (diabetic hyperlipidemic control, DHC), the third one received ATOR (50 mg/kg/day) while the fourth one received LYC (20 mg/kg/day). Serum and tissue samples were collected for biochemical and histological evaluations. Key findings DHC rats demonstrated significant hyperglycemia, dyslipidemia, increased hepatic fatty acids synthetase (FAS), malondialdehyde (MDA), tumor necrosis factor- alpha (TNF-α), 3-hydroxy 3-methylglutaryl coenzyme A (HMG-CoA) reductase and ATP Citrate lyase (ACLY). However, hepatic reduced glutathione (GSH) and phosphorylated form of AMP-activated protein kinase (AMPK-P) activities showed significant decrease. ATOR or LYC administration induced hypoglycemic and hypolipidemic effects; decreased hepatic levels of MDA, TNF-α, HMG-CoA reductase, ACLY and FAS along with GSH and AMPK-P increases. Histopathological findings showed clear correlation with the biomarkers results. Significance LYC demonstrated favorable significant effects regarding the biomarkers studied as compared to ATOR and may be expressed as a potent therapeutic agent of natural origin for hyperlipidemia complications either alone or in combination with other hypolipidemic drugs.
... Wanita memiliki hormon estrogen yang berfungsi sebagai kardioprotektif atau mencegah terbentuknya plak di pembuluh darah arteri. Namun, dalam keadaan pre menopause atau menopause, hormon estrogen pada wanita akan berkurang dan dapat meningkatkan risiko terjadinya penyakit jantung dan pembuluh darah (Trapani et al, 2010 (Mawi, 2011). Terdapat juga faktor risiko yang tidak dapat dikendalikan meliputi usia dan jenis kelamin (Mahan & Escott-Stump, 2012). ...
Article
Cholesterol total level is a risk factor for cardiovascular disease. Mung beans contain soluble fiber and isoflavones that decrease cholesterol levels. The aim of this research is to find out the potency of mung beans juice to decrease total cholesterol total level of hypercholesterol woman. Research design was quasi experimental with pretest - posttest control group. The subjects were 34 women with hypercholesterolemia, and divided into two groups ie 17 womens in Penarukan Village which is treatment group and 17 womens in Ketapang Village which is control groups. All the subjects have met the inclusion criteria. The treatment group consumed juice of 300 ml/day for 14 days while the control group did not get juice. It was a single blind treatment. Cholesterol total level measurement used Electrode Based Biosensor and CHOD-PAP methods. The mean of cholesterol total level of control group before and after was increasing 5,7% ie 240,1 mg/dl and 253,9 mg/dl, but intervention group was decreasing 11% ie 220 mg/dl and 195,7 mg/dl. Paired t-test result shows that there is a significant difference between total cholesterol levels before and after in the treatment group and control group (p= 0.000). The conclusion of this research is the provision of mung beans juice 300 ml/day for 14 days reduce total cholesterol levels. Keywords : Hypercholesterolemia Women; Mung beans Juice; Total Cholesterol Level;
Article
Background: Lipoprotein cholesterol measurements fluctuated across the menstrual cycle, corresponding to rising and declining concentrations of gonadotropins and ovarian steroid hormones levels in young women, whereas premenopausal women and those who inside the menopausal transition characterized by a poor ovarian response to high secretion of gonadotropin leading to low levels of ovarian steroids. Aims: To investigate the lipoprotein cholesterol changes and their interactions with sex steroid hormones during different phases of the menstrual cycle in menstruated young and old (premenopausal) women. Methods: The sample included thirty healthy women (aged 20-45 years) divided into two groups with 15 women /group according to their ages )20-25 years( (40-45 years). Blood samples were collected on the 8th, 16th, and 24th days of the menstrual cycle from both young and old menstruating women to compare the previous parameters between these days for each group and compare these parameters between similar days for the first and second groups. Lipid profile (Total cholesterol (TC), Triglyceride (TG), High density lipoprotein (HDL), Low density lipoprotein (LDL), and Very low density lipoprotein (VLDL) changes be assayed by Bio-Systems Kit components. Statistical analysis was carried out using one-way Analysis of Variance (ANOVA), followed by Duncan's test, and t-tests were used to assess differences between groups. Results: TC and TG levels did not significantly increase on the 8th day of the menstrual cycle, which represents the follicular phase, in comparison with the other days (16th and 24th days). However, LDL level increased significantly (p ? 0.05) on the 8th day compared to the other days. On the other hand, HDL and VLDL levels increased on the 16th day of the menstrual cycle, which represents the ovulatory phase. Discussion: TC, TG, and LDL levels increased during the follicular phase and were accompanied by high levels of FSH and estradiol, leading to a favorable lipid profile, whereas FSH and estradiol are considered regulators of cholesterol biosynthesis and a buffering capacity. HDL and VLDL levels rise during ovulation to meet the requirements of ovulation and ensure successful ovulation. Conclusions: Studying and evaluating changes in the lipid profile according to the menstrual cycle phase of the menstrual cycle represents an important issue in knowing the physiological and healthy woman state and preventing cardiovascular disease incidence.
Article
The public and scientists remain skeptical about egg consumption, given that cardiovascular diseases (CVDs) are the leading causes of death in worldwide. This review mainly explained the recurrence of contradictory conclusions about relationships between egg consumption and CVD risk and discussed effects of egg cholesterol intake on cholesterol homeostasis. Factors including individual health status and cholesterol sensitivity, dietary pattern, region, and race should be distinguished when understanding generalized conclusions. Identified compensatory mechanisms in response to dietary cholesterol and the resulting balance in cholesterol biosynthesis, absorption, and efflux supported the view that moderate egg consumption had no substantial overall impacts on cholesterol homeostasis in healthy people. Excessive cholesterol intake is not recommended in individuals with distempered metabolism. More than cholesterol metabolism, impacts of egg consumption as a part of overall diet on CVD risk should be considered from aspects of nutrient intake, lipid metabolism, and energy supply in the future.
Article
Full-text available
Rotator Cuff Injuries (RCI) are prevalent cause of shoulder pain affecting over 20% of the population in the USA. Surgical repair of the torn rotator cuff helps in relieving the pressure on the rotator cuff tendon and from symptoms, however tendon-to-bone healing after rotator cuff surgery still has a high failure rate. Hyperlipidemia has been strongly associated with RCI although the cellular and molecular mechanisms are largely unknown. The focus of this critical review is to further explore the role of hyperlipidemia in RCI and rotator cuff tissue repair to determine its implication as a risk factor for tears, repair, and retears. A literature review was conducted to elucidate the role of hyperlipidemia as an inflammatory mediator and catalyst for structural instability within the shoulder. The results from various studies were critically reviewed to summarize the relationship between hyperlipidemia and rotator cuff pathology. Hyperlipidemia induces LDL-particle entrapment within the dense regular collagen of rotator cuff tendons resulting in foam cell aggregation and macrophage recruitment. Subsequent inflammatory pathways including the JAK2/STAT3 pathway and NLRP3 inflammasome pathway led to persistent inflammation and Extracellular Matrix (ECM) degradation within the rotator cuff. While arthroscopic repair remains the most common treatment modality, nonsurgical treatment including statins, vitamin D, and targeting miRNA are also of therapeutic benefit. Hyperlipidemia interferes with arthroscopic repairs by inducing inflammation and stiffness within tendons and increases the risk of retears. Most notably, targeting underlying mechanisms influencing inflammation has large therapeutic value as a novel treatment strategy for the management of rotator cuff pathology.
Article
Elevated low density lipoprotein cholesterol levels in circulation may get deposited in coronary arteries and result in atherosclerotic plaques at an early age and act as risk factor for cardiovascular diseases. More recent and detailed anthropometric indices in addition to traditional indices were assessed in this study along with lipid profile and lipoprotein indices in urban population (n = 100) of Amritsar, Punjab. n = 48 subjects comprised the case group and n = 52 subjects the control group. Study was approved by the Institutional Ethics Committee of Guru Nanak Dev University and individual voluntary written informed consent was provided by the study group. 46% of cases were obese and lipids and lipoprotein indices were significantly higher in obese group while low density lipoprotein cholesterol and low density lipoprotein cholesterol/high density lipoprotein cholesterol also in overweight category. Factor analysis extracted the 7 components out of 23 variables with a cummulative variance of 81.45%. Females had higher central obesity, waist hip ratio, waist height ratio and body adiposity index. Pearson’s correlation analysis revealed a positive significant association of age (r = 0.363, p = 0.027), socio-economic status (r = 0.328, p = 0.023), waist hip ratio (r = 0.298, p = 0.042), waist height ratio (r = 0.864, p = 0 000) and body mass index (r = 0.794, p = 0.000) with low density lipoprotein cholesterol levels and of gender with atherogenic index (r = 0.363, p = 0.011). Also, a significant correlation of total body fat mass with atherogenic index (r = 0.286, p = 0.048) and and body roundness index with low density lipoprotein (r = 0.345, p = 0.016) was observed. Multivariate regression analysis revealed age (B = 0.318, t = 2.380, p = 0.022) and gender (B = 0.370, t = 2.687, p = 0.010) to be significant predictors of body roundness index as well as age (B = 0.139, t = 2.307, p = 0.027) of low density lipoprotein cholesterol. The highest prediction accuracy was showed by waist circumference (AUC = 0.597) and the least by a body shape index (AUC = 0.458) as revealed by Receiver Operating Characteristics analysis. A positive association of low density lipoproteins with central and general obesity, signifies that lipid metabolism gets affected by adiposity and may further affect the lipoprotein indices which are reported to be associated with atherosclerosis and coronary heart diseases.
Article
Full-text available
Protein phosphatase 2A (PP2A) comprises a family of serine/threonine phosphatases, minimally containing a well conserved catalytic subunit, the activity of which is highly regulated. Regulation is accomplished mainly by members of a family of regulatory subunits, which determine the substrate specificity, (sub)cellular localization and catalytic activity of the PP2A holoenzymes. Moreover, the catalytic subunit is subject to two types of post-translational modification, phosphorylation and methylation, which are also thought to be important regulatory devices. The regulatory ability of PTPA (PTPase activator), originally identified as a protein stimulating the phosphotyrosine phosphatase activity of PP2A, will also be discussed, alongside the other regulatory inputs. The use of specific PP2A inhibitors and molecular genetics in yeast, Drosophila and mice has revealed roles for PP2A in cell cycle regulation, cell morphology and development. PP2A also plays a prominent role in the regulation of specific signal transduction cascades, as witnessed by its presence in a number of macromolecular signalling modules, where it is often found in association with other phosphatases and kinases. Additionally, PP2A interacts with a substantial number of other cellular and viral proteins, which are PP2A substrates, target PP2A to different subcellular compartments or affect enzyme activity. Finally, the de-regulation of PP2A in some specific pathologies will be touched upon.
Article
To predict the consequences of cholesterol screening among elderly Americans who do not have symptoms of heart disease, we explore the cost implications of a cholesterol screening program, evaluate evidence linking hypercholesterolemia to coronary heart disease and mortality in the elderly, and describe the likely effects of therapy of hypercholesterolemia. According to our calculations, if all Americans 65 years of age and older adhered to a cholesterol screening program similar to the one proposed by the National Cholesterol Education Program, minimum annual expenditures for screening and treatment would be between $1.6 billion and $16.8 billion, depending on the effectiveness of diet and the cost of the medications used to treat hypercholesterolemia. There is no direct evidence that this program would lessen overall morbidity or extend the lives of elderly Americans. (Arch Intern Med. 1991;151:1089-1095)
Data
Ageing has been defined as a progressive decrease in physiological capacity and a reduced ability to respond to environmental stresses. It has been observed that diet-restricted animals show a minor morbidity in age-related disease. Among these age-related diseases, hypercholesterolemia is the most recurring one and it is often associated with cardiac failure. Several studies have been published indicating age-dependent changes in circulating levels of cholesterol in both humans and in rodents; recently changes have also been reported in the proteins involved in cholesterol homeostasis, that is, 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoAR), Insig-induced gene (Insig) protein, SREBP cleavage activating protein (SCAP), sterol regulatory element binding protein (SREBP), and low density lipoprotein receptor (LDLr). Most age-related modifications of biochemical parameters are normalized or very improved in food-restricted animals, so the aim of this work is to examine whether or not alterations of the factors involved in cholesterol homeostasis which occur during ageing could be counteracted by caloric restriction (CR). The data show that the diet restrictions used attenuate the age-related effects on the factors involved in the synthesis and the degradation rate of HMG-CoAR; in spite of this, CRs have a good effect on the age-related hypercholesterolemia whose reduction seems to depend both on the correct membrane LDLr localization and on the proper restored HMG-CoAR activity.
Data
Background/Aims: In thioacetamide-induced liver injury a modification of isoprenoid content and an increase of reactive oxygen species has been described. We have examined how reactive oxygen species influence the 3-hydroxy-3-methylglutaryl coenzyme A reductase, the rate limiting enzyme of the isoprenoid biosynthetic pathway, to verify if changes of that enzyme activity are involved in the changed lipid composition of the liver. Methods: In chronic and acute thioacetamide-treated rat liver we measured the reactive oxygen species content, the activation state and K M , the level and degradation rate of the hepatic reductase, its short term regulatory enzymes and the liver lipid profile. Results: In thioacetamide-treated rat liver, the reactive oxygen species content is high and the reductase is fully activated with no modifications in its K M and its short term regulatory enzymes. The reductase level is reduced in chronic thioacetamide treated rats and its degradation rate is altered. Conclusions: The data show a relationship between reactive oxygen species production and altered 3-hydroxy-3-methylglutaryl coenzyme A reductase activity. It is suggested that reducing the levels of reactive oxygen species may improve the altered lipid profile found in liver injury. q 2005 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.
Data
As the main risk factor for cardiovascular disease, hypercholesterolemia is one of the most studied age-related metabolic alterations. In the liver, cholesterol homeostasis is strictly regulated through the modulation of the 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoA reductase), the key enzyme of cholesterol biosynthesis. With ageing, hepatic HMG-CoA reductase becomes completely activated and cholesterol content increases in the blood. The research reported in this paper uses the regulatory enzymes of reductase (i.e., the AMP-dependent kinase (AMPK) and the protein phosphatase 2A (PP2A)), the HMG-CoA reductase thermodependent activity and the ''in vitro'' enzyme degradation to elucidate the role played by the HMG-CoA reductase regulation and its membrane interaction. Related experiments were performed on 3 and 24 months ''ad libitum'' (AL) fed rats and 24 months caloric-restricted rats. The results show no changes in the PP2A level and the activation state of AMP dependent kinase in aged ''ad libitum'' fed rats. By contrast, the activation state of the kinase is enhanced in the aged caloric-restricted animals. With respect to the adult, the thermodependent activity of reductase remains unchanged, while the degradation rate of the HMG-CoA reductase is slower and independent on proteasome. These findings support the hypothesis that a different arrangement of the HMG-CoA reductase membrane domain in aged rats is a cause of reductase deregulation.