Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Metabolomics ; 18(1): 8, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34989922

RESUMEN

INTRODUCTION: A rapidly growing body of data documents associations between disease of the brain and small molecules generated by gut-microbiota (GMB). While such metabolites can affect brain function through a variety of mechanisms, the most direct action would be on the central nervous system (CNS) itself. OBJECTIVE: Identify indolic and phenolic GMB-dependent small molecules that reach bioactive concentrations in primate CNS. METHODS: We conducted a PubMed search for metabolomic studies of the primate CNS [brain tissue or cerebrospinal fluid (CSF)] and then selected for phenolic or indolic metabolites that (i) had been quantified, (ii) were GMB-dependent. For each chemical we then conducted a search for studies of bioactivity conducted in vitro in human cells of any kind or in CNS cells from the mouse or rat. RESULTS: 36 metabolites of interests were identified in primate CNS through targeted metabolomics. Quantification was available for 31/36 and in vitro bioactivity for 23/36. The reported CNS range for 8 metabolites 2-(3-hydroxyphenyl)acetic acid, 2-(4-hydroxyphenyl)acetic acid, 3-(3-hydroxyphenyl)propanoic acid, (E)-3-(3,4-dihydroxyphenyl)prop-2-enoic acid [caffeic acid], 3-hydroxybenzoic acid, 4-hydroxybenzoic acid, 2-acetamido-3-(1H-indol-3-yl)propanoic acid [N-acetyltryptophan], 1H-indol-3-yl hydrogen sulfate [indoxyl-3-sulfate] overlapped with a bioactive concentration. However, the number and quality of relevant studies of CNS neurochemistry as well as of bioactivity were highly limited. Structural isomers, multiple metabolites and potential confounders were inadequately considered. CONCLUSION: The potential direct bioactivity of GMB-derived indolic and phenolic molecules on primate CNS remains largely unknown. The field requires additional strategies to identify and prioritize screening of the most promising small molecules that enter the CNS.


Asunto(s)
Microbioma Gastrointestinal , Metabolómica , Animales , Sistema Nervioso Central/metabolismo , Ratones , Fenoles/metabolismo , Primates/metabolismo , Ratas
2.
Artículo en Inglés | MEDLINE | ID: mdl-28584146

RESUMEN

Antibiotics excreted into the intestinal tract may disrupt the microbiota that provide colonization resistance against enteric pathogens and alter normal metabolic functions of the microbiota. Many of the bacterial metabolites produced in the intestinal tract are absorbed systemically and excreted in urine. Here, we used a mouse model to test the hypothesis that alterations in levels of targeted bacterial metabolites in urine specimens could provide useful biomarkers indicating disrupted or intact colonization resistance. To assess in vivo colonization resistance, mice were challenged with Clostridium difficile spores orally 3, 6, and 11 days after the completion of 2 days of treatment with piperacillin-tazobactam, aztreonam, or saline. For concurrent groups of antibiotic-treated mice, urine samples were analyzed by using liquid chromatography-tandem mass spectrometry (LC-MS/MS) to quantify the concentrations of 11 compounds targeted as potential biomarkers of colonization resistance. Aztreonam did not affect colonization resistance, whereas piperacillin-tazobactam disrupted colonization resistance 3 days after piperacillin-tazobactam treatment, with complete recovery by 11 days after treatment. Three of the 11 compounds exhibited a statistically significant and >10-fold increase (the tryptophan metabolite N-acetyltryptophan) or decrease (the plant polyphenyl derivatives cinnamoylglycine and enterodiol) in concentrations in urine 3 days after piperacillin-tazobactam treatment, followed by recovery to baseline that coincided with the restoration of in vivo colonization resistance. These urinary metabolites could provide useful and easily accessible biomarkers indicating intact or disrupted colonization resistance during and after antibiotic treatment.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Glicina/análogos & derivados , Intestinos/microbiología , Lignanos/orina , Triptófano/análogos & derivados , Animales , Antibacterianos/farmacología , Aztreonam/metabolismo , Aztreonam/farmacología , Biomarcadores/orina , Cromatografía Liquida , Clostridioides difficile/efectos de los fármacos , Farmacorresistencia Bacteriana/fisiología , Glicina/orina , Intestinos/efectos de los fármacos , Metaboloma/efectos de los fármacos , Metabolómica , Ratones , Ácido Penicilánico/análogos & derivados , Ácido Penicilánico/metabolismo , Ácido Penicilánico/farmacología , Piperacilina/metabolismo , Piperacilina/farmacología , Combinación Piperacilina y Tazobactam , Espectrometría de Masas en Tándem , Triptófano/orina
3.
Microorganisms ; 10(12)2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36557660

RESUMEN

Since influenza and coronaviruses are currently deadly and emerging threats worldwide, better treatment, remediation and prevention options are needed. In that regard, a basic understanding of severe acute respiratory syndrome (SARS)-CoV-2/COVID-19 (Betacoronaviridae) and other viral pathogen mechanisms of transmission are expected. Unfortunately, unprecedented, and growing distrust of vaccines and even masks or personal protective equipment (PPE) in the United States and elsewhere presents itself as an added challenge. We postulate that development of improved and highly effective prophylactic measures, together with new life-saving therapies that do inhibit or otherwise treat infection of SARS-CoV-2, influenza and other viral pathogens, could be an adjunct measure to globally protect vulnerable individuals from pandemic threats. In this review, we share what we learned from the past COVID experience to offer a multifactorial and improved approach to current and future pandemic infections or threats using low-cost means.

4.
J Biol Chem ; 285(40): 30489-95, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20670940

RESUMEN

The presence of the prion protein (PrP) in normal human urine is controversial and currently inconclusive. This issue has taken a special relevance because prion infectivity has been demonstrated in urine of animals carrying experimental or naturally occurring prion diseases, but the actual presence and tissue origin of the infectious prion have not been determined. We used immunoprecipitation, one- and two-dimensional electrophoresis, and mass spectrometry to prove definitely the presence of PrP in human urine and its post-translational modifications. We show that urinary PrP (uPrP) is truncated mainly at residue 112 but also at other residues up to 122. This truncation makes uPrP undetectable with some commonly used antibodies to PrP. uPrP is glycosylated and carries an anchor which, at variance with that of cellular PrP, lacks the inositol-associated phospholipid moiety, indicating that uPrP is probably shed from the cell surface. The detailed characterization of uPrP reported here definitely proves the presence of PrP in human urine and will help determine the origin of prion infectivity in urine.


Asunto(s)
Enfermedades por Prión/orina , Priones/orina , Procesamiento Proteico-Postraduccional , Humanos , Priones/patogenicidad
5.
Pathog Immun ; 5(1): 382-418, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33474520

RESUMEN

BACKGROUND: The gut microbiome (GMB) generates numerous small chemicals that can be absorbed by the host and variously biotransformed, incorporated, or excreted. The resulting metabolome can provide information about the state of the GMB, of the host, and of their relationship. Exploiting this information in the service of biomarker development is contingent on knowing the GMB-sensitivity of the individual chemicals comprising the metabolome. In this regard, human studies have lagged far behind animal studies. Accordingly, we tested the hypothesis that serum levels of chemicals unequivocally demonstrated to be GMB-sensitive in rodent models would also be affected in a clinical patient sample treated with broad spectrum antibiotics. METHODS: We collected serum samples from 20 hospitalized patients before, during, and after treatment with broad-spectrum antibiotics. We also collected samples from 5 control patients admitted to the hospital but not prescribed antibiotics. We submitted the samples for a non-targeted metabolomic analysis and then focused on chemicals known to be affected both by germ-free status and by antibiotic treatment in the mouse and/or rat. RESULTS: Putative identification was obtained for 499 chemicals in human serum. An aggregate analysis did not show any time x treatment interactions. However, our literature search identified 10 serum chemicals affected both by germ-free status and antibiotic treatment in the mouse or rat. Six of those chemicals were measured in our patient samples and additionally met criteria for inclusion in a focused analysis. Serum levels of 5 chemicals (p-cresol sulfate, phenol sulfate, hippurate, indole propionate, and indoxyl sulfate) declined significantly in our group of antibiotic-treated patients but did not change in our patient control group. CONCLUSIONS: Broad-spectrum antibiotic treatment in patients lowered serum levels of selected chemicals previously demonstrated to be GMB-sensitive in rodent models. Interestingly, all those chemicals are known to be uremic solutes that can be derived from aromatic amino acids (L-phenylalanine, L-tyrosine, or L-tryptophan) by anaerobic bacteria, particularly Clostridial species. We conclude that judiciously selected serum chemicals can reliably detect antibiotic-induced suppression of the GMB in man and thus facilitate further metabolome-based biomarker development.

6.
J Cell Mol Med ; 13(5): 853-65, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19292735

RESUMEN

Alzheimer disease (AD) and stroke are two leading causes of age-associated dementia. Increasing evidence points to vascular damage as an early contributor to the development of AD and AD-like pathology. In this review, we discuss the role of G protein-coupled receptor kinase 2 (GRK2) as it relates to individuals affected by AD and how the cardiovasculature plays a role in AD pathogenesis. The possible involvement of GRKs in AD pathogenesis is an interesting notion, which may help bridge the gap in our understanding of the heartbrain connection in relation to neurovisceral damage and vascular complications in AD, since kinases of this family are known to regulate numerous receptor functions both in the brain, myocardium, and elsewhere. The aim of this review is to discuss our findings of overexpression of GRK2 in the context of the early pathogenesis of AD, because increased levels of GRK2 immunoreactivity were found in vulnerable neurons of AD patients as well as in a two-vessel occlusion (2-VO) mammalian model of ischaemia. Also, we consider the consequences for this overexpression as a loss of G-protein coupled receptor (GPCR) regulation, as well as suggest a potential role for GPCRs and GRKs in a unifying theory of AD pathogenesis, particularly in the context of cerebrovascular disease. We synthesize this newer information and attempt to put it into context with GRKs as regulators of diverse physiological cellular functions that could be appropriate targets for future pharmacological intervention.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Trastornos Cerebrovasculares/enzimología , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Neuronas/enzimología , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/patología , Animales , Trastornos Cerebrovasculares/complicaciones , Trastornos Cerebrovasculares/patología , Humanos , Neuronas/patología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/patología
7.
J Cell Mol Med ; 13(2): 320-33, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18373733

RESUMEN

Brain function declines with age and is associated with diminishing mitochondrial integrity. The neuronal mitochondrial ultrastructural changes of young (4 months) and old (21 months) F344 rats supplemented with two mitochondrial metabolites, acetyl-L-carnitine (ALCAR, 0.2%[wt/vol] in the drinking water) and R-alpha-lipoic acid (LA, 0.1%[wt/wt] in the chow), were analysed using qualitative and quantitative electron microscopy techniques. Two independent morphologists blinded to sample identity examined and scored all electron micrographs. Mitochondria were examined in each micrograph, and each structure was scored according to the degree of injury. Controls displayed an age-associated significant decrease in the number of intact mitochondria (P = 0.026) as well as an increase in mitochondria with broken cristae (P < 0.001) in the hippocampus as demonstrated by electron microscopic observations. Neuronal mitochondrial damage was associated with damage in vessel wall cells, especially vascular endothelial cells. Dietary supplementation of young and aged animals increased the proliferation of intact mitochondria and reduced the density of mitochondria associated with vacuoles and lipofuscin. Feeding old rats ALCAR and LA significantly reduced the number of severely damaged mitochondria (P = 0.02) and increased the number of intact mitochondria (P < 0.001) in the hippocampus. These results suggest that feeding ALCAR with LA may ameliorate age-associated mitochondrial ultrastructural decay and are consistent with previous studies showing improved brain function.


Asunto(s)
Acetilcarnitina/farmacología , Envejecimiento/fisiología , Mitocondrias , Neuronas , Ácido Tióctico/farmacología , Acetilcarnitina/administración & dosificación , Animales , Suplementos Dietéticos , Hipocampo/citología , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Mitocondrias/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Ácido Tióctico/administración & dosificación
8.
Psychopharmacology (Berl) ; 236(5): 1471-1489, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31197432

RESUMEN

Schizophrenia and autism spectrum disorder have long been associated with elevated levels of various small phenolic molecules (SPMs). In turn, the gut microbiota (GMB) has been implicated in the kinetics of many of these analytes. Unfortunately, research into the possible relevance of GMB-mediated SPMs to neuropsychiatry continues to be limited by heterogeneous study design, numerous sources of variance and technical challenges. Some SPMs have multiple structural isomers and most have conjugates. Without specialized approaches, SPMs can be incorrectly assigned or inaccurately quantified. In addition, SPM levels can be affected by dietary polyphenol or protein consumption and by various medications and diseases. Nonetheless, heterotypical excretion of various SPMs in association with schizophrenia or autism continues to be reported in independent samples. Recent studies in human cerebrospinal fluid demonstrate the presence of many SPMs A large number of these are bioactive in experimental models. Whether such mechanisms are relevant to the human brain in health or disease is not known. Systematic metabolomic and microbiome studies of well-characterized populations, an appreciation of multiple confounds, and implementation of standardized approaches across platforms and sites are needed to delineate the potential utility of the phenolic interactome in neuropsychiatry.


Asunto(s)
Trastorno del Espectro Autista/metabolismo , Encéfalo/metabolismo , Microbioma Gastrointestinal/fisiología , Fenoles/metabolismo , Esquizofrenia/metabolismo , Trastorno del Espectro Autista/psicología , Encéfalo/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Polifenoles/administración & dosificación , Polifenoles/metabolismo , Psicología del Esquizofrénico
9.
Pathog Immun ; 4(2): 271-293, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31773068

RESUMEN

BACKGROUND: The gut microbiome (GMB) generates numerous chemicals that are absorbed systemically and excreted in urine. Antibiotics can disrupt the GMB ecosystem and weaken its resistance to colonization by enteric pathogens such as Clostridium difficile. If the changes in GMB composition and metabolism are sufficiently large, they can be reflected in the urinary metabo-lome. Characterizing these changes could provide a potentially valuable biomarker of the status of the GMB. While preliminary studies suggest such a possibility, the high level of data variance presents a challenge to translational applications. Since many GMB-generated chemicals are derived from the biotransformation of plant-derived dietary polyphenols, administering an oral precursor challenge should amplify GMB-dependent changes in urinary metabolites. METHODS: A course of antibiotics (clindamycin, piperacillin/tazobactam, or aztreonam) was administered SC daily (days 1 and 2) to mice receiving polyphenol-rich green tea in drinking water. Urine was collected at baseline as well as days 3, 7, and 11. Levels of pyrogallol and pyrocatechol, two phenolic molecules unequivocally GMB-dependent in humans but that had not been similarly examined in mice, were quantified. RESULTS: In confirmation of our hypothesis, differential changes in murine urinary pyrogallol levels identified the treatments (clindamycin, piperacillin/tazobactam) previously associated with a weakening of colonization resistance to Clostridium difficile. The changes in pyrocatechol levels did not withstand corrections for multiple comparisons. CONCLUSIONS: In the mouse, urinary pyrogallol and, in all likelihood, pyrocatechol levels, are GMB-dependent and, in combination with precursor challenge, deserve further consideration as potential metabolomic biomarkers for the health and dysbiotic vulnerability of the GMB.

10.
Mini Rev Med Chem ; 8(13): 1395-406, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18991755

RESUMEN

Alzheimer disease treatment has yet to yield a successful therapy that addresses the source of the damage found in brains. Of the varied proposed theories of AD etiology, reactive oxygen species (ROS) generation is cited as a common factor. Efforts to reduce the pathology associated with ROS via antioxidants therefore offer new hope to patients suffering from this devastative disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antioxidantes/uso terapéutico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Antioxidantes/farmacología , Encéfalo/patología , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Humanos , Estrés Oxidativo/efectos de los fármacos
11.
Vasc Health Risk Manag ; 4(3): 721-30, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18827923

RESUMEN

The pathogenesis that is primarily responsible for Alzheimer's disease (AD) and cerebrovascular accidents (CVA) appears to involve chronic hypoperfusion. We studied the ultrastructural features of vascular lesions and mitochondria in brain vascular wall cells from human AD biopsy samples and two transgenic mouse models of AD, yeast artificial chromosome (YAC) and C57B6/SJL Tg (+), which overexpress human amyloid beta precursor protein (AbetaPP). In situ hybridization using probes for normal and 5 kb deleted human and mouse mitochondrial DNA (mtDNA) was performed along with immunocytochemistry using antibodies against the Abeta peptide processed from AbetaPP, 8-hydroxy-2'-guanosine (8OHG), and cytochrome c oxidase (COX). More amyloid deposition, oxidative stress markers as well as mitochondrial DNA deletions and structural abnormalities were present in the vascular walls of the human AD samples and the AbetaPP-YAC and C57B6/SJL Tg (+) transgenic mice compared to age-matched controls. Ultrastructural damage in perivascular cells highly correlated with endothelial lesions in all samples. Therefore, pharmacological interventions, directed at correcting the chronic hypoperfusion state, may change the natural course of the development of dementing neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/irrigación sanguínea , Encéfalo/ultraestructura , ADN Mitocondrial/ultraestructura , Animales , Aterosclerosis , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microcirculación , Neuronas/ultraestructura
12.
Microorganisms ; 6(4)2018 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-30340384

RESUMEN

'Leaky gut' syndrome, long-associated with celiac disease, has attracted much attention in recent years and for decades, was widely known in complementary/alternative medicine circles. It is often described as an increase in the permeability of the intestinal mucosa, which could allow bacteria, toxic digestive metabolites, bacterial toxins, and small molecules to 'leak' into the bloodstream. Nervous system involvement with celiac disease is know to occur even at subclinical levels. Gluten and gluten sensitivity are considered to trigger this syndrome in individuals genetically predisposed to celiac disease. However, the incidence of celiac disease in the general population is quite low. Nevertheless, increased public interest in gluten sensitivity has contributed to expanded food labels stating 'gluten-free' and the proliferation of gluten-free products, which further drives gluten-free lifestyle changes by individuals without frank celiac disease. Moreover, systemic inflammation is associated with celiac disease, depression, and psychiatric comorbidities. This mini-review focuses on the possible neurophysiological basis of leaky gut; leaky brain disease; and the microbiota's contribution to inflammation, gastrointestinal, and blood-brain barrier integrity, in order to build a case for possible mechanisms that could foster further 'leaky' syndromes. We ask whether a gluten-free diet is important for anyone or only those with celiac disease.

13.
Bioanalysis ; 10(19): 1591-1608, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30295550

RESUMEN

AIM: Co-metabolism between a human host and the gastrointestinal microbiota generates many small phenolic molecules such as 3-hydroxy-3-(3-hydroxyphenyl)propanoic acid (3,3-HPHPA), which are reported to be elevated in schizophrenia and autism. Characterization of these chemicals, however, has been limited by analytic challenges. METHODOLOGY/RESULTS: We applied HPLC to separate and quantify over 50 analytes, including multiple structural isomers of 3,3-HPHPA in human cerebrospinal fluid, serum and urine. Confirmation of identity was provided by NMR, by MS and other detection methods. The highly selective methods support rapid quantification of multiple metabolites and exhibit superior chromatographic behavior. CONCLUSION: An improved ultra-HPLC-MS/MS and structural approaches can accurately quantify 3,3-HPHPA and related analytes in human biological matrices.


Asunto(s)
Hidroxibenzoatos/metabolismo , Metabolómica/métodos , Cromatografía Líquida de Alta Presión , Humanos , Hidroxibenzoatos/sangre , Hidroxibenzoatos/líquido cefalorraquídeo , Hidroxibenzoatos/orina , Isomerismo , Espectrometría de Masas en Tándem
14.
Curr Pharm Des ; 12(6): 691-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16472159

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Ciclo Celular/fisiología , Hormonas/fisiología , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Ciclo Celular/efectos de los fármacos , Quimioterapia/métodos , Quimioterapia/tendencias , Gonadotropinas/antagonistas & inhibidores , Gonadotropinas/fisiología , Antagonistas de Hormonas/uso terapéutico , Humanos , Modelos Biológicos
15.
Neurotox Res ; 10(1): 43-56, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17000469

RESUMEN

Heterotrimeric guanine nucleotide-binding (G) protein-coupled receptor kinases (GRKs) are cytosolic proteins that are known to contribute to the adaptation of the heptahelical G protein-coupled receptors (GPCRs) and to regulate downstream signals through these receptors. GPCRs mediate the action of messengers that are key modulators of cardiac and vascular cell function, such as growth and differentiation. GRKs are members of a multigene family, which are classified into three subfamilies and are found in cardiac, vascular and cerebral tissues. Increasing evidence strongly supports the hypothesis that vascular damage is an early contributor to the development of Alzheimer disease (AD) and/or other pathology that can mimic human AD. Based on this hypothesis, and since kinases of this family are known to regulate numerous receptor functions both in the brain, myocardium and elsewhere, we explored cellular and subcellular localization by immunoreactivity of G protein-coupled receptor kinase 2 (GRK2), also known as beta-adrenergic receptor kinase-1(betaARK1), in the early pathogenesis of AD and in ischemia reperfusion injury models of brain hypoperfusion. In the present study, we used the two-vessel carotid artery occlusion model, namely the 2-VO system that results in chronic brain hypoperfusion (CBH) and mimics mild cognitive impairment (MCI) and vascular changes in AD pathology. Our findings demonstrate the early overexpression of GRK2 member kinase in the cerebrovasculature, especially endothelial cells (EC) following CBH, as well as in select cells from human AD tissue. We found a significant increase in GRK2 immunoreactivity in the EC of AD patients and after CBH, which preceded any amyloid deposition. Since GRK2 activity is associated with certain compensatory changes in brain cellular compartments and in ischemic cardiac tissue, our findings suggest that chronic hypoperfusion initiates oxidative stress in these conditions and appears to be the main initiating injury stimulus for disruption of brain and cerebrovascular homeostasis and metabolism.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/patología , Expresión Génica/fisiología , Infarto de la Arteria Cerebral Media/patología , Mitocondrias/metabolismo , Quinasas de Receptores Adrenérgicos beta/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/genética , Animales , Biomarcadores/metabolismo , Encéfalo/ultraestructura , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Quinasa 2 del Receptor Acoplado a Proteína-G , Humanos , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Microscopía Inmunoelectrónica/métodos , Persona de Mediana Edad , Mitocondrias/patología , Mitocondrias/ultraestructura , Ratas , Ratas Sprague-Dawley , Quinasas de Receptores Adrenérgicos beta/genética
16.
Sci Aging Knowledge Environ ; 2005(4): pe3, 2005 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-15673817

RESUMEN

The extracellular matrix accumulates biologically active advanced glycation endproducts such as carboxymethyl-lysine (CML). Alikhani et al. recently reported that CML-rich collagen, representing an artifically aged matrix protein, induces apoptosis in vivo and in fibroblasts. This observation may have profound implications for the biology of cell-matrix interactions during aging.


Asunto(s)
Apoptosis , Matriz Extracelular/fisiología , Fibroblastos/citología , Productos Finales de Glicación Avanzada/fisiología , Lisina/análogos & derivados , Envejecimiento/patología , Animales , Ciclo Celular/fisiología , Lisina/fisiología , Reacción de Maillard , Modelos Biológicos , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/fisiología , Transducción de Señal
17.
Front Biosci (Elite Ed) ; 7(2): 229-41, 2015 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-25553376

RESUMEN

Although there has been much research into autism or autistic spectrum disorder (ASD), there is room for considerable conjecture regarding the etiology of these developmental brain disorders. ASD is marked by a complex interaction between environmental factors and genetic predisposition, including epistasis. This manuscript argues that changes in oxidative metabolism, thiamine homeostasis, heavy metal deposition and cellular immunity have a role in the etiopathogenesis of autism and ASD. Recent findings from our group and others provide evidence for abnormal thiol metabolism, marked by significant alteration in the deposition of several trace heavy metal species. Together with these, we find differences in thiamine homeostasis in ASD patients, which can be corrected by supplementation. We hypothesize that altered thiol metabolism from heavy metal toxicity, one of the key mechanisms for oxidative stress production, may be responsible for the biochemical alterations in transketolase, dysautonomia and abnormal thiamine homeostasis. Although it is unknown why these particular metals accumulate, we suspect that children with ASD and forms of autism may have particular trouble excreting thiol-toxic heavy metal species, many of which exist as divalent cations. We maintain mercury accumulation is evidence of altered clearance. Together with concomitant oxidative stress, these findings may offer an intriguing component or possible mechanism for oxidative stress-mediated neurodegeneration in ASD patients. Regardless of the exact cause, these factors may be more important to the etiology of this symptomatically diverse disease spectrum. Here, we offer insight into new avenues of exploration as well as the development of novel treatment approaches for these growing and devastating diseases.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/etiología , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Tiamina/metabolismo , Transcetolasa/metabolismo , Trastornos Generalizados del Desarrollo Infantil/tratamiento farmacológico , Homeostasis , Humanos , Mutación , Estrés Oxidativo , Tiamina/análogos & derivados , Tiamina/uso terapéutico
18.
J Gerontol A Biol Sci Med Sci ; 58(6): 508-16, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12807921

RESUMEN

The accumulation of Maillard reaction products increases with age in long-lived proteins and can be retarded by caloric restriction. Here we determined whether caloric restriction inhibits formation of glycation and glycoxidation products in skin collagen of squirrel and rhesus monkeys between 1990-1997. Restricted monkeys (n = 11, n = 30, respectively) were maintained at 70% of caloric intake of controls (n = 25, n = 32, respectively). Glycation was assessed by furosine and glycoxidation by pentosidine and carboxymethyl-lysine. With age, the rate of furosine formation moderately but nonsignificantly (p >.05) increased in both control monkey groups. It significantly (p =.011) decreased in the caloric-restricted rhesus, but not squirrel monkeys. Caloric restriction did not significantly decrease the pentosidine or carboxymethyl-lysine rates in either species of monkeys. These results suggest that caloric restriction, when maintained long-term in nonhuman primates, tends to decrease glycation, but not glycoxidation.


Asunto(s)
Envejecimiento/metabolismo , Arginina/análogos & derivados , Colágeno/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Lisina/análogos & derivados , Piel/metabolismo , Análisis de Varianza , Animales , Arginina/metabolismo , Biomarcadores , Peso Corporal , Cromatografía Líquida de Alta Presión , Privación de Alimentos , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Longevidad , Lisina/metabolismo , Macaca mulatta , Oxidación-Reducción , Análisis de Regresión , Saimiri
19.
Neurotox Res ; 5(7): 491-504, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14715433

RESUMEN

Chronic vascular hypoperfusion induces oxidative stress and brain energy failure, and leads to neuronal death, which manifests as cognitive impairment and the development of brain pathology as in Alzheimer disease (AD). It is becoming more widely accepted that AD is characterized by impairments in energy metabolism. We hypothesize that hypoperfusion-induced mitochondrial failure plays a central role in the generation of reactive oxygen species, resulting in oxidative damage to brain cellular compartments, especially in the vascular endothelium and neuronal cell bodies in AD. All of these changes have been found to occur before pathology and coexist during the progression of AD. In this review we have summarized recent evidence and our own knowledge regarding the relationship between the hypoperfusion-induced vascular damage that initiates oxidative stress and mitochondrial abnormalities that appear to be a key target for the development of AD pathology. Future investigations into both the mechanisms behind amyloid beta (Abeta) deposition and the possible accelerating effects of environmental factors, such as chronic hypoxia/reperfusion may open the door for effective pharmacological treatments of AD. We hypothesize that an imbalance between endothelium derived vasoconstrictors and vasodilators, along with an antioxidant system deficiency and mitochondria lesions are prominent in AD. Future studies examining the importance of mitochondrial pathophysiology in different brain cellular compartments may provide insight not only into neurodegenerative and/or cerebrovascular disease pathobiology but may also provide targets for treating these conditions.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Trastornos Cerebrovasculares/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Animales , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiopatología , Trastornos Cerebrovasculares/genética , Trastornos Cerebrovasculares/patología , Trastornos Cerebrovasculares/fisiopatología , Humanos , Ratones , Mitocondrias/patología
20.
Neurol Res ; 26(5): 547-53, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15265272

RESUMEN

Nitric oxide (NO) is a short-life key bioregulatory active molecule in the cardiovascular, immune and nervous systems. NO is synthesized by converting L-arginine to L-citrulline by enzymes called NO synthase (NOS). The growing body of evidence strongly supports the theory that this molecule appears to be one of the key targets for the disruption of normal brain homeostasis, which causes the development of brain lesions and pathology such as in Alzheimer's disease (AD) or other related dementia. The vascular content of NO activity appears especially to be a main contributor to this pathology before the over-expression of other NOS isoforms activity in a different brain cellular compartment. We speculate that pharmacological intervention using NO donors and/or NO suppressors will be able to delay or minimize the development of brain pathology and further progression of mental retardation.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Encéfalo/enzimología , Arterias Cerebrales/enzimología , Trastornos Cerebrovasculares/enzimología , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Arterias Cerebrales/patología , Arterias Cerebrales/fisiopatología , Trastornos Cerebrovasculares/complicaciones , Trastornos Cerebrovasculares/fisiopatología , Demencia Vascular/complicaciones , Demencia Vascular/enzimología , Demencia Vascular/fisiopatología , Humanos , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA