Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Free Radic Res ; 43(2): 156-64, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19160110

ABSTRACT

Oxidative modifications are a hallmark of oxidative imbalance in the brains of individuals with Alzheimer's, Parkinson's and prion diseases and their respective animal models. While the causes of oxidative stress are relatively well-documented, the effects of chronically reducing oxidative stress on cognition, pathology and biochemistry require further clarification. To address this, young and aged control and amyloid-beta protein precursor-over-expressing mice were fed a diet with added R-alpha lipoic acid for 10 months to determine the effect of chronic antioxidant administration on the cognition and neuropathology and biochemistry of the brain. Both wild type and transgenic mice treated with R-alpha lipoic acid displayed significant reductions in markers of oxidative modifications. On the other hand, R-alpha lipoic acid had little effect on Y-maze performance throughout the study and did not decrease end-point amyloid-beta load. These results suggest that, despite the clear role of oxidative stress in mediating amyloid pathology and cognitive decline in ageing and AbetaPP-transgenic mice, long-term antioxidant therapy, at levels within tolerable nutritional guidelines and which reduce oxidative modifications, have limited benefit.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Antioxidants/pharmacology , Oxidative Stress/drug effects , Thioctic Acid/pharmacology , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Cognition/drug effects , Disease Models, Animal , Immunohistochemistry , Maze Learning/drug effects , Mice , Mice, Transgenic
2.
Int J Clin Exp Pathol ; 1(2): 134-46, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18784806

ABSTRACT

The re-expression of multiple cell cycle markers representing various cell cycle phases in postmitotic pyramidal neurons suggests that neurons in Alzheimer disease (AD) attempt to re-enter the cell cycle. Entry into the cell cycle requires activation of G1 to S phase cell cycle proteins, among which retinoblastoma protein (pRb) is a key regulator. pRb inhibits the transcription of cell cycle proteins in the nucleus of healthy cells by interaction and consequent blocking of the active site of E2F, dependent upon the phosphate stoichiometry and combination of the locations of their 16 potential phosphorylation sites on pRb. Therefore, to determine whether pRb is involved in the aberrant cell cycle phenotype in AD neurons, a systematic immunocytochemical evaluation of the phosphorylation status of pRb protein using antibodies specific for multiple phosphorylation sites (i.e., pSpT249/252, pS612, pS795, pS807, pS811 and pT821) was carried out in the hippocampal regions of brains from AD patients. Increased levels of phospho-pRb (ppRb) for all these phosphorylation sites were noted in the brains of AD patients as compared to control cases. More importantly, redistribution of ppRb from the nucleus to the cytoplasm of susceptible neurons, with significant localization in neurofibrillary tangles and neuritic plaques, was observed. Additional studies revealed extensive co-localization between phospho-p38 and ppRb, implicating that p38 activation may contribute to cell cycle abnormalities through pRb phosphorylation. Taken together, these data supports the concept of neuronal cell cycle re-entry in AD and indicates a crucial role for pRb in this process.

3.
Adv Clin Chem ; 45: 139-53, 2008.
Article in English | MEDLINE | ID: mdl-18429496

ABSTRACT

For decades, Alzheimer's disease (AD) has been linked to aging, gender, and menopause. Not surprisingly, this led most investigators to focus on the role of estrogen. While undoubtedly important, estrogen is unlikely the key determinant of disease pathogenesis. Rather, it appears that estrogen may work in conjunction with a novel determinant of disease pathogenesis, namely gonadotropins. The fact that gonadotropins, specifically luteinizing hormone, play a pivotal role in disease is apparent from significant etiological, epidemiological, and pathological evidences. Moreover, targeting gonadotropins appears to have beneficial actions as a therapeutic regimen.


Subject(s)
Alzheimer Disease/physiopathology , Estrogens/physiology , Gonadotropins/physiology , Menopause , Female , Humans , Male , Sex Factors
4.
Clin Med Res ; 5(3): 177-83, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18056027

ABSTRACT

Several hypotheses have been proposed that attempt to explain the pathogenesis of Alzheimer Disease (AD) including theories involving senile plaque and neurofibrillary tangle formation, increased oxidative stress, and cell cycle abnormalities, since evidence for each of these pathological phenomena have been well documented in AD. Recent epidemiological and experimental data also support a role for the gonadotropin luteinizing hormone in AD. Paralleling the female predominance for developing AD, luteinizing hormone levels are significantly higher in females as compared to males, and furthermore, luteinizing hormone levels are higher still in individuals who succumb to AD. Luteinizing hormone, which is capable of modulating cognitive behavior, is not only present in the brain, but also has the highest receptor levels in the hippocampus, a key processor of cognition that is severely deteriorated in AD. Furthermore, we recently examined cognitive performance in a well-characterized transgenic mouse that over-expresses luteinizing hormone and found that these animals show decreased cognitive performance when compared to controls. We have also found that abolishing luteinizing hormone in amyloid-beta protein precursor transgenic mice (Tg2576) using a potent gonadotropin-lowering gonadotropin-releasing hormone agonist, leuprolide acetate, resulted in improved hippocampally-related cognitive performance and decreased amyloid-beta deposition. These findings, together with data indicating that luteinizing hormone modulates amyloid-beta protein precursor processing in vivo and in vitro, suggest that luteinizing hormone may contribute to AD pathology through an amyloid-dependent mechanism. These promising findings support the importance of luteinizing hormone in AD and bring to the forefront an alternative, and much needed, therapeutic avenue for the treatment of this insidious disease.


Subject(s)
Alzheimer Disease/drug therapy , Fertility Agents, Female/therapeutic use , Leuprolide/therapeutic use , Luteinizing Hormone , Alzheimer Disease/blood , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/metabolism , Animals , Disease Progression , Humans , Luteinizing Hormone/agonists , Luteinizing Hormone/blood , Prognosis
5.
Mol Cell Endocrinol ; 269(1-2): 107-11, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17376589

ABSTRACT

Questions surrounding estrogen therapy for post-menopausal cognitive decline and dementia led us to examine the role of luteinizing hormone that becomes elevated after menopause. We examined hippocampal-associated cognitive performance, as measured with the Y-maze task, in two strains of transgenic mice, one (Tg-LHbeta) which over-expresses luteinizing hormone and another (LHRKO), which has increased circulating luteinizing hormone levels, but its receptors are silenced. Our results demonstrate that Tg-LHbeta, but not LHRKO mice, show decreased Y-maze performance when compared to aged-matched wild-type animals. These findings indicate that increased luteinizing hormone levels, in the presence of functional receptors may, at least in part, be responsible for cognitive decline after menopause. As such, modulation of luteinizing hormone or its receptor levels may prove to be useful therapeutic strategies for cognitive decline associated with aging and age-related neurodegenerative diseases such as Alzheimer disease.


Subject(s)
Cognition Disorders/chemically induced , Cognition/drug effects , Luteinizing Hormone/blood , Luteinizing Hormone/pharmacology , Animals , Female , Luteinizing Hormone, beta Subunit/genetics , Male , Maze Learning/drug effects , Mice , Mice, Inbred Strains , Mice, Transgenic , Receptors, LH/genetics
6.
Mol Cell Endocrinol ; 260-262: 271-5, 2007 Jan 02.
Article in English | MEDLINE | ID: mdl-17052835

ABSTRACT

While there is ample experimental evidence supporting the role of estrogen in the pathogenesis of Alzheimer disease, recent inconclusive data regarding hormone replacement therapy (HRT), specifically, the unexpected results of the Women's Health Initiative (WHI) Memory Study has raised serious questions regarding the protective effects of estrogen. Because of this and other inconsistencies in the estrogen hypothesis, we propose that another hormone of the hypothalamic-pituitary-gonadal axis, luteinizing hormone, is a major factor in the pathogenesis of Alzheimer disease. Specifically, we suspect that the increase in gonadotropin concentrations, and not the decrease in steroid hormone (e.g., estrogen) production following menopause/andropause, is a primary causative factor for the development of Alzheimer disease. In this review, we examine how the gonadotropins may play a central and determining role in modulating the susceptibility to, and progression of, Alzheimer disease.


Subject(s)
Alzheimer Disease/etiology , Gonadotropins/metabolism , Sex Characteristics , Alzheimer Disease/therapy , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonads/pathology , Humans , Hypothalamus/pathology , Pituitary Gland/pathology
7.
Article in English | MEDLINE | ID: mdl-18220951

ABSTRACT

Epidemiological and experimental data supporting a role for luteinizing hormone in Alzheimer disease is accumulating. Paralleling the female predominance for developing Alzheimer disease, luteinizing hormone levels are significantly higher in females as compared to males and luteinizing hormone levels are higher still in individuals who succumb to Alzheimer disease. Importantly, luteinizing hormone, which is capable of modulating cognitive behavior, is not only present in the brain, but also has the highest receptor levels in the hippocampus, a key processor of cognition that is severely deteriorated in Alzheimer disease. These findings, together with data indicating that luteinizing hormone modulates amyloid-beta protein precursor processing in vivo and in vitro, suggests that luteinizing hormone may contribute to Alzheimer disease pathology through an amyloid-dependent mechanism. Indeed, abolishing luteinizing hormone, using a potent gonadotropin-lowering agent, leuprolide acetate, in the amyloid-beta protein precursor transgenic mice improved hippocampally-related cognitive performance and decreased amyloid-beta deposition. These promising findings support the importance of luteinizing hormone in Alzheimer disease and bring to the forefront an alternative, and much needed therapeutic avenue for the treatment of this insidious disease.


Subject(s)
Alzheimer Disease/metabolism , Luteinizing Hormone/metabolism , Animals , Clinical Trials as Topic , Female , Humans , Male
8.
J Biomed Biotechnol ; 2006(3): 39508, 2006.
Article in English | MEDLINE | ID: mdl-17047306

ABSTRACT

Recent evidence indicates that, alongside oxidative stress, dysregulation of the cell cycle in neurons susceptible to degeneration in Alzheimer disease may play a crucial role in the initiation of the disease. As such, the role of reproductive hormones, which are closely associated with the cell cycle both during development and after birth, may be of key import. While estrogen has been the primary focus, the protective effects of hormone replacement therapy on cognition and dementia only during a crucial period led us to expand the study of hormonal influences to other members of the hypothalamic pituitary axis. Specifically, in this review, we focus on luteinizing hormone, which is not only increased in the sera of patients with Alzheimer disease but, like estrogen, is modulated by hormone replacement therapy and also influences cognitive behavior and pathogenic processing in animal models of the disease. Targeting gonadotropins may be a useful treatment strategy for disease targeting multiple pleiotropic downstream consequences.

9.
Mol Neurodegener ; 1: 14, 2006 Oct 03.
Article in English | MEDLINE | ID: mdl-17018137

ABSTRACT

BACKGROUND: Alzheimer disease (AD) is clinically characterized by progressive memory loss, impairments in behavior, language and visual-spatial skills and ultimately, death. Epidemiological data reporting the predisposition of women to AD has led to a number of lines of evidence suggesting that age-related changes in hormones of the hypothalamic-pituitary-gonadal (HPG) axis following reproductive senescence, may contribute to the etiology of AD. Recent studies from our group and others have reported not only increases in circulating gonadotropins, namely luteinizing hormone (LH) in individuals with AD compared with control individuals, but also significant elevations of LH in vulnerable neuronal populations in individuals with AD compared to control cases as well as the highest density of gonadotropin receptors in the brain are found within the hippocampus, a region devastated in AD. However, while LH is higher in AD patients, the downstream consequences of this are incompletely understood. To begin to examine this issue, here, we examined the expression levels of steroidogenic acute regulatory (StAR) protein, which regulates the first key event in steroidogenesis, namely, the transport of cholesterol into the mitochondria, and is regulated by LH through the cyclic AMP second messenger pathway, in AD and control brain tissue. RESULTS: Our data revealed that StAR protein was markedly increased in both the cytoplasm of hippocampal pyramidal neurons as well as in the cytoplasm of other non-neuronal cell types from AD brains when compared with age-matched controls. Importantly, and suggestive of a direct mechanistic link, StAR protein expression in AD brains colocalized with LH receptor expression. CONCLUSION: Therefore, our findings suggest that LH is not only able to bind to its receptor and induce potentially pathogenic signaling in AD, but also that steroidogenic pathways regulated by LH may play a role in AD.

10.
Drugs R D ; 7(3): 187-93, 2006.
Article in English | MEDLINE | ID: mdl-16752944

ABSTRACT

Estrogen and other sex hormones have received a great deal of attention for their speculative role in Alzheimer's disease (AD), but at present a direct connection between estrogen and the pathogenesis of AD remains elusive and somewhat contradictory. For example, on one hand there is a large body of evidence suggesting that estrogen is neuroprotective and improves cognition, and that hormone replacement therapy (HRT) at the onset of menopause reduces the risk of developing AD decades later. However, on the other hand, studies such as the Women's Health Initiative demonstrate that HRT initiated in elderly women increases the risk of dementia. While estrogen continues to be investigated, the disparity of findings involving HRT has led many researchers to examine other hormones of the hypothalamic-pituitary-gonadal axis such as luteinising hormone (LH) and follicle-stimulating hormone. In this review, we propose that LH, rather than estrogen, is the paramount player in the pathogenesis of AD. Notably, both men and women experience a 3- to 4-fold increase in LH with aging, and LH receptors are found throughout the brain following a regional pattern remarkably similar to those neuron populations affected in AD. With respect to disease, serum LH level is increased in women with AD relative to non-diseased controls, and levels of LH in the brain are also elevated in AD. Mechanistically, we propose that elevated levels of LH may be a fundamental instigator responsible for the aberrant reactivation of the cell cycle that is seen in AD. Based on these aforementioned aspects, clinical trials underway with leuprolide acetate, a gonadotropin-releasing hormone agonist that ablates serum LH levels, hold great promise as a ready means of treatment in individuals afflicted with AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Estrogens/physiology , Gonadotropin-Releasing Hormone/agonists , Estrogen Replacement Therapy/adverse effects , Estrogen Replacement Therapy/methods , Estrogens/administration & dosage , Estrogens/adverse effects , Female , Gonadotropins/physiology , Humans , Male , Sex Factors
11.
Biochim Biophys Acta ; 1762(4): 447-52, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16503402

ABSTRACT

Until recently, the study of hormonal influences in Alzheimer disease was limited to the role of sex steroids. Despite numerous epidemiological studies supporting a protective role for estrogen in Alzheimer disease, recent studies show that estrogen administration in elderly women increases the risk of disease. Reconciling these contradictory reports, we previously hypothesized that other hormones of the hypothalamic-pituitary-gonadal axis, such as luteinizing hormone, may be involved in the onset and development of the disease. In this regard, luteinizing hormone is elevated in Alzheimer disease and is known to modulate amyloidogenic processing of amyloid-beta protein precursor. Therefore, in this study, to evaluate the therapeutic potential of luteinizing hormone ablation, we administered a gonadotropin-releasing hormone analogue, leuprolide acetate, to an aged transgenic mouse model of Alzheimer disease (Tg 2576) and measured cognitive Y-maze performance and amyloid-beta deposition after 3 months of treatment. Our data indicate that luteinizing hormone ablation significantly attenuated cognitive decline and decreased amyloid-beta deposition as compared to placebo-treated animals. Importantly, leuprolide acetate-mediated reduction of amyloid-beta correlated with improved cognition. Since both cognitive loss and amyloid-beta deposition are features of Alzheimer disease, leuprolide acetate treatment may prove to be a useful therapeutic strategy for this disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cognition , Hippocampus/pathology , Luteinizing Hormone/physiology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cognition/drug effects , Female , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hippocampus/metabolism , Leuprolide/pharmacology , Luteinizing Hormone/agonists , Maze Learning , Mice , Mice, Transgenic , Mutation , Protein Isoforms/genetics , Protein Isoforms/metabolism
12.
Curr Pharm Des ; 12(6): 691-7, 2006.
Article in English | MEDLINE | ID: mdl-16472159

ABSTRACT

Several hypotheses have been proposed attempting to explain the pathogenesis of Alzheimer disease (AD) including theories involving amyloid deposition, tau phosphorylation, oxidative stress, metal ion dysregulation and inflammation. Strong evidence suggests that each one contributes to disease pathogenesis, though none of these mechanisms result in all the downstream changes that occur during the course of AD. For this reason, we and others have begun the search for a causative factor that predates known features found in AD, and that might be a fundamental initiator of the pathophysiological cascade. In this regard, we propose that the dysregulation of the cell cycle that occurs in neurons susceptible to degeneration in the hippocampus during AD is a potential causative factor that would initiate all known pathological events. Neuronal changes supporting alterations in cell cycle control in the etiology of AD include the ectopic expression of markers of the cell cycle, organelle kinesis and cytoskeletal alterations including tau phosphorylation. Given the early and presumably devastating consequences of cell cycle re-entry, we have made a concerted effort to elucidate the initiating factor that drives aberrant mitotic re-entry in AD. As a result of the gender bias present in AD, we suspect that postmenopausal and andropausal hormones may be involved and, with this in mind, in this review we specifically focus on the gonadotropins. Therapeutic interventions targeted at gonadotropins, if they are indeed the driving mitogenic force, could both prevent disease in those patients currently asymptomatic or halt, and even reverse, disease in those currently afflicted.


Subject(s)
Alzheimer Disease/physiopathology , Cell Cycle/physiology , Hormones/physiology , Alzheimer Disease/drug therapy , Animals , Cell Cycle/drug effects , Drug Therapy/methods , Drug Therapy/trends , Gonadotropins/antagonists & inhibitors , Gonadotropins/physiology , Hormone Antagonists/therapeutic use , Humans , Models, Biological
13.
Ann N Y Acad Sci ; 1052: 201-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16024763

ABSTRACT

Epidemiological data showing a predisposition of women to develop Alzheimer disease (AD) led many researchers to investigate the role of sex steroids, namely estrogen, in disease pathogenesis. Although there is circumstantial support for the role of estrogen, the unexpected results of the Women's Health Initiative (WHI) Memory Study, which reported an increase in the risk for probable dementia and impaired cognitive performance in postmenopausal women treated with a combination of estrogen and progestin, have raised serious questions regarding the protective effects of estrogen. Although explanations for these surprising results vary greatly, the WHI Memory Study cannot be correctly interpreted without a complete investigation of the effects of the other hormones of the hypothalamic-pituitary-gonadal (HPG) axis on the aging brain. Certain hormones of the HPG axis, namely, the gonadotropins (luteinizing hormone and follicle-stimulating hormone), are not only involved in regulating reproductive function via a complex feedback loop but are also known to cross the blood-brain barrier. We propose that the increase in gonadotropin concentrations, and not the decrease in steroid hormone (e.g., estrogen) production following menopause/andropause, is a potentially primary causative factor for the development of AD. In this review, we examine how the gonadotropins may play a central and determining role in modulating the susceptibility to, and progression of, AD. On this basis, we suggest that the results of the WHI Memory Study are not only predictable but also avoidable by therapeutically targeting the gonadotropins instead of the sex steroids.


Subject(s)
Alzheimer Disease/etiology , Estrogens/physiology , Gonadotropins, Pituitary/physiology , Aged , Aging , Blood-Brain Barrier , Brain/physiopathology , Estrogen Replacement Therapy/adverse effects , Female , Follicle Stimulating Hormone/physiology , Humans , Hypothalamus/physiopathology , Luteinizing Hormone/physiology , Memory , Ovary/physiopathology , Pituitary Gland/physiopathology , Postmenopause , Premenopause , Women's Health
14.
Mech Ageing Dev ; 126(10): 1019-25, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15936057

ABSTRACT

Several hypotheses have been proposed attempting to explain the pathogenesis of Alzheimer disease including, among others, theories involving amyloid deposition, tau phosphorylation, oxidative stress, metal ion dysregulation and inflammation. While there is strong evidence suggesting that each one of these proposed mechanisms contributes to disease pathogenesis, none of these mechanisms are able to account for all the physiological changes that occur during the course of the disease. For this reason, we and others have begun the search for a causative factor that predates known features found in Alzheimer disease, and that might therefore be a fundamental initiator of the pathophysiological cascade. We propose that the dysregulation of the cell cycle that occurs in neurons susceptible to degeneration in the hippocampus during Alzheimer disease is a potential causative factor that, together with oxidative stress, would initiate all known pathological events. Neuronal changes supporting alterations in cell cycle control in the etiology of Alzheimer disease include the ectopic expression of markers of the cell cycle, organelle kinesis and cytoskeletal alterations including tau phosphorylation. Such mitotic alterations are not only one of the earliest neuronal abnormalities in the disease, but as discussed herein, are also intimately linked to all of the other pathological hallmarks of Alzheimer disease including tau protein, amyloid beta protein precursor and oxidative stress, and even risk factors such as mutations in the presenilin genes. Therefore, therapeutic interventions targeted toward ameliorating mitotic changes would be predicted to have a profound and positive impact on Alzheimer disease progression.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/metabolism , Mitosis , Neurons/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid/metabolism , Animals , Hippocampus/pathology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Metals/metabolism , Neurons/pathology , Neuropharmacology/methods , Oxidative Stress , tau Proteins/metabolism
15.
Alzheimer Dis Assoc Disord ; 19(2): 95-9, 2005.
Article in English | MEDLINE | ID: mdl-15942328

ABSTRACT

Epidemiological data reporting the predisposition of women to Alzheimer disease has provided researchers with an important clue as to the identity of the driving pathogenic force and lead many to question the potential role of sex steroids, namely estrogen, in disease pathogenesis. However, while estrogen has become the primary focus of research in the field, inconclusive data regarding estrogen replacement therapy has lead some researchers to begin investigating the effects of the other hormones of the hypothalamic-pituitary-gonadal (HPG) axis on the aging brain. Certain hormones of the HPG axis, namely the gonadotropins (luteinizing hormone and follicle-stimulating hormone), are not only involved in regulating reproductive function via a complex feedback loop but are also known to cross the blood-brain barrier. Recently, we proposed that an increase in gonadotropin concentrations, not the decrease in steroid hormone (eg, estrogen) production following menopause/andropause, is a potentially primary causative factor for the development of Alzheimer disease. In this review, we examine how the gonadotropins may play a central and determining role in modulating the susceptibility to, and progression of, Alzheimer disease. Based on this, we suggest that therapeutic interventions targeted at gonadotropins may both prevent disease in those patients currently asymptomatic or may halt, and even reverse, disease in those currently afflicted.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Luteinizing Hormone/physiology , Sex Characteristics , Alzheimer Disease/drug therapy , Animals , Hormone Antagonists/therapeutic use , Humans
16.
Curr Med Chem ; 12(10): 1137-47, 2005.
Article in English | MEDLINE | ID: mdl-15892629

ABSTRACT

In recent years, Alzheimer disease (AD) has received great attention as an incurable and fatal disease that threatens the lives of aging individuals. Debates regarding areas of research and treatment designs have made headlines as scientists in the field question ongoing work. Despite these academic quarrels, significant insights concerning the cellular and molecular basis of AD have illuminated the potential causes and consequences of AD pathogenesis in the human brain. Additionally, assigning relationships among scientific evidence is difficult due to the nature of the disease. It is crucial to note that all findings do not constitute causality as AD has many stages of progression, and therefore a particular finding may reflect disease epiphenomenon. Determining the primary causes of disease are even more problematic when considering that a succinct timeline in which a normal aging brain develops AD-like changes due to a single cause may not be appropriate, as increasing lines of evidence indicate that multiple factors likely contribute to the clinical manifestation of AD. Implications for therapeutic strategies are dramatically affected by viewing AD as a multi-factorial disease state, one specific treatment may not be able to prevent or reverse AD if this is indeed the case. In this regard, the current focus on individual therapeutic targets may prove to be ineffective in the successful treatment of AD; however, if taken in combination, these singular therapies may likely result in the global suppression of AD. In this review, the scientific basis for common AD therapeutics as well as the efficacy of these treatments will be discussed.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/therapeutic use , Cholinesterase Inhibitors/therapeutic use , Excitatory Amino Acid Antagonists/therapeutic use , Hormone Replacement Therapy , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Neuroprotective Agents/therapeutic use
17.
J Neurosci Res ; 79(4): 554-60, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15635602

ABSTRACT

Two of the earliest manifestations of the selective neurodegeneration that occurs in Alzheimer's disease (AD) involve the oxidative modification of various biomacromolecules and the reexpression of a multitude of cell cycle-related proteins. Taken together with the proximal and ectopic increases in activated components of the ERK and p38 pathways, involved in mitotic and cellular stress signaling, respectively, there is a clear and important role for mitotic and oxidative insults in the pathogenesis of AD. Despite the mounting evidence, however, for the causal role of mitogenic abnormalities and oxidative stress in AD pathogenesis, the effect of the converging relevant pathways due to chronic stimulation in AD remains largely unknown. To delineate further the mechanism by which mitogenic and stress signaling cascades converge, we focused on one of the downstream effectors of activated ERK and p38, mitogen- and stress-activated kinase 1 (MSK1). Activated MSK1, phosphorylated at residues Ser376 and Thr581, was upregulated in vulnerable neurons in AD when compared to that in age-matched controls, whereas MSK1 phosphorylated at residue Ser360 was not increased in AD. Furthermore, activated MSK1 phosphorylated at Thr581 colocalized strongly with activated p38 but only weakly with activated ERK, whereas MSK1 phosphorylated at Ser376 colocalized strongly with activated ERK but only weakly with activated p38, suggesting potential preferential phosphorylation sites for the two upstream effectors.


Subject(s)
Alzheimer Disease/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Aged , Aged, 80 and over , Case-Control Studies , Female , Hippocampus/cytology , Histones/metabolism , Humans , Immunohistochemistry/methods , Male , Neurons/metabolism , Phosphorylation , Postmortem Changes , Serine/metabolism , Threonine/metabolism
18.
Curr Drug Targets CNS Neurol Disord ; 3(4): 281-5, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15379604

ABSTRACT

Based on epidemiological and observational studies, estrogen and hormone-replacement therapy were until recently viewed as major factors in the prevention of Alzheimer's disease (AD). However, a recent randomized clinical trial revealed that hormone replacement therapy using estrogen plus progestin may actually exacerbate the incidence of dementia when administered to elderly women. These contradictory reports have cast grave doubt on the role of estrogen in disease pathogenesis and led us to consider an alternate hypothesis that would be consistent with both observations. Specifically, we suspect that hormones of the hypothalamic pituitary gonadal axis such as gonadotropins, that are regulated by estrogen (or in males by testosterone), are involved in the pathogenesis of Alzheimer's disease. One such gonadotropin, luteinizing hormone (LH), is significantly elevated in both the sera and brain tissue of patients with AD and leads to an increased production of amyloid-beta. Importantly, a key role in disease pathogenesis is further supported by the fact that the distribution of neuronal receptors for LH parallels those populations of neurons that degenerate during the course of the disease. That gonadotropins, not estrogen nor testosterone, mediate disease pathogenesis has led to a paradigm shift, not only for the treatment of AD but a wide variety of other age-related diseases. Therefore, the effects of agents that abolish LH, such as leuprolide acetate, which are currently being evaluated in Phase II clinical trials for the treatment of AD, are eagerly anticipated.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Gonadotropins, Pituitary/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Leuprolide/pharmacology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/biosynthesis , Animals , Estrogens/adverse effects , Female , Gonadotropins, Pituitary/antagonists & inhibitors , Hormone Replacement Therapy/adverse effects , Humans , Hypothalamo-Hypophyseal System/metabolism , Leuprolide/therapeutic use , Luteinizing Hormone/antagonists & inhibitors , Luteinizing Hormone/metabolism , Male , Receptors, LH/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
19.
Acta Neurobiol Exp (Wars) ; 64(1): 113-8, 2004.
Article in English | MEDLINE | ID: mdl-15190686

ABSTRACT

The search for a definitive gender bias in Alzheimer's disease has resulted in a multitude of epidemiological findings that point to a higher prevalence and incidence of Alzheimer's disease in women. Due to this reported predisposition of women to Alzheimer's disease, the sex steroid estrogen has become the primary focus of research in this field, however, inconclusive data regarding estrogen replacement therapy has lead some researchers to further investigate the role of the other hormones of the hypothalamic-pituitary-gonadal (HPG) axis that have been, for the most, part overlooked. The hormones of the HPG axis, such as the gonadotropin, (luteinizing hormone and follicle-stimulating hormone), are involved in regulating reproductive function via a complex feedback loop. We propose that it is in fact the increase in gonadotropin concentrations and not the decrease in steroid hormone (e.g., estrogen) production following menopause/andropause that results in an increased risk of Alzheimer's disease. Furthermore, when the role of gonadotropins is taken into account, the data obtained from recent epidemiological studies and randomized trials suggesting the ineffectiveness estrogen may indeed be misinterpreted. In this review, we examine how hormones of the hypothalamic-pituitary-gonadal axis, in particular the gonadotropins, play a central and determining role in modulating the susceptibility to and progression of Alzheimer's disease. Based on this, we suggest that therapeutic interventions targeted at gonadotropins could both prevent disease in those patients currently asymptomatic or halt, and even reverse, disease in those currently afflicted.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/prevention & control , Estrogen Replacement Therapy , Estrogens/administration & dosage , Gonadotropins/metabolism , Alzheimer Disease/epidemiology , Female , Humans , Neuroprotective Agents/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...