Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 373
Filtrar
2.
J Intern Med ; 269(1): 8-15, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21158973

RESUMO

The hurtful feelings associated with failing can be devastating especially if the failure occurs after the investment of a considerable effort. The reflection of a lifetime of work in translational medicine has revealed that the study of failures can give birth to new insights that can be explored with important consequences. This article discusses the analysis of two failures that have led to remarkable discoveries. The first led to the discovery of TNF as an important mediator of inflammation that can, if unchecked, cause severe damage in mammals. The second is the identification of erythropoietin as the natural inhibitor of the production and biological activity of TNF. I hope that this paper will help give students the courage to persist in looking for the insights that are the by-products of failure, and to understand the long time lines in the path of discoveries.


Assuntos
Eritropoetina/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Modelos Animais de Doenças , Eritropoetina/história , Eritropoetina/uso terapêutico , História do Século XX , Humanos , Achados Incidentais , Inflamação/tratamento farmacológico , Inflamação/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/história , Fator de Necrose Tumoral alfa/toxicidade
3.
J Intern Med ; 264(5): 405-32, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19017170

RESUMO

In its classic hormonal role, erythropoietin (EPO) is produced by the kidney and regulates the number of erythrocytes within the circulation to provide adequate tissue oxygenation. EPO also mediates other effects directed towards optimizing oxygen delivery to tissues, e.g. modulating regional blood flow and reducing blood loss by promoting thrombosis within damaged vessels. Over the past 15 years, many unexpected nonhaematopoietic functions of EPO have been identified. In these more recently appreciated nonhormonal roles, locally-produced EPO signals through a different receptor isoform and is a major molecular component of the injury response, in which it counteracts the effects of pro-inflammatory cytokines. Acutely, EPO prevents programmed cell death and reduces the development of secondary, pro-inflammatory cytokine-induced injury. Within a longer time frame, EPO provides trophic support to enable regeneration and healing. As the region immediately surrounding damage is typically relatively deficient in endogenous EPO, administration of recombinant EPO can provide increased tissue protection. However, effective use of EPO as therapy for tissue injury requires higher doses than for haematopoiesis, potentially triggering serious adverse effects. The identification of a tissue-protective receptor isoform has facilitated the engineering of nonhaematopoietic, tissue-protective EPO derivatives, e.g. carbamyl EPO, that avoid these complications. Recently, regions within the EPO molecule mediating tissue protection have been identified and this has enabled the development of potent tissue-protective peptides, including some mimicking EPO's tertiary structure but unrelated in primary sequence.


Assuntos
Eritropoetina/uso terapêutico , Cicatrização/efeitos dos fármacos , Ferimentos e Lesões/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Citocinas/antagonistas & inibidores , Citocinas/imunologia , Eritropoese/fisiologia , Eritropoetina/análogos & derivados , Humanos , Isoformas de Proteínas/metabolismo , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes , Ferimentos e Lesões/imunologia
5.
Kidney Int ; 70(2): 246-50, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16738535

RESUMO

A greatly expanded understanding of the biology of endogenous erythropoietin (EPO) has emerged since the early 1990s. Originally viewed as the renal hormone dedicated to erythrocyte production, it is now clear that EPO is produced locally by many other tissues in response to physical or metabolic stress. In its autocrine-paracrine roles, EPO mediates preconditioning (ischemic tolerance) and specifically limits the destructive potential of tumor necrosis factor alpha and other proinflammatory cytokines in the brain, heart, kidney, and other tissues. As local production of EPO is generally suppressed following injury, administration of exogenous EPO has been a successful therapeutic approach in preclinical and clinical studies, for example, following ischemia-reperfusion and toxin-induced renal injuries, and in human stroke. The therapeutic time window of tissue protection by EPO is typically very wide in experimental models, showing effectiveness when administered before, during, or after an insult and raising optimism for a high clinical potential. Although there is progress in understanding the signaling pathways responsible for EPO's tissue-protective actions that are similar to, but not as redundant as, those employed for erythrocyte maturation, much work remains to be carried out. Experimental observations also suggest the existence of EPO receptor (EPOR) isoforms mediating EPO's diverse biological activities and have identified a tissue-protective receptor complex consisting of the EPOR and the beta common receptor (CD131) subunit that is also employed by granulocyte-macrophage colony-stimulating factor, interleukin-3 and interleukin-5. Successfully engineered analogues of EPO that selectively activate tissue protection without stimulating hematopoiesis confirm the concept of a tissue-protective receptor and have significant potential utility in the investigational and therapeutic arenas.


Assuntos
Eritropoetina/fisiologia , Eritropoetina/uso terapêutico , Nefropatias/tratamento farmacológico , Nefropatias/fisiopatologia , Rim/fisiologia , Animais , Endotélio Vascular/fisiologia , Humanos , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/fisiopatologia
7.
Mol Psychiatry ; 9(1): 42-54, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14581931

RESUMO

Erythropoietin (EPO) is a candidate compound for neuroprotection in human brain disease capable of combating a spectrum of pathophysiological processes operational during the progression of schizophrenic psychosis. The purpose of the present study was to prepare the ground for its application in a first neuroprotective add-on strategy in schizophrenia, aiming at improvement of cognitive brain function as well as prevention/slowing of degenerative processes. Using rodent studies, primary hippocampal neurons in culture, immunohistochemical analysis of human post-mortem brain tissue and nuclear imaging technology in man, we demonstrate that: (1) peripherally applied recombinant human (rh) EPO penetrates into the brain efficiently both in rat and humans, (2) rhEPO is enriched intracranially in healthy men and more distinctly in schizophrenic patients, (3) EPO receptors are densely expressed in hippocampus and cortex of schizophrenic subjects but distinctly less in controls, (4) rhEPO attenuates the haloperidol-induced neuronal death in vitro, and (4) peripherally administered rhEPO enhances cognitive functioning in mice in the context of an aversion task involving cortical and subcortical pathways presumably affected in schizophrenia. These observations, together with the known safety of rhEPO, render it an interesting compound for neuroprotective add-on strategies in schizophrenia and other human diseases characterized by a progressive decline in cognitive performance.


Assuntos
Eritropoetina/farmacocinética , Fármacos Neuroprotetores/farmacocinética , Esquizofrenia Paranoide/diagnóstico por imagem , Esquizofrenia Paranoide/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antipsicóticos/farmacologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Cognição/efeitos dos fármacos , Feminino , Haloperidol/farmacologia , Humanos , Radioisótopos de Índio , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/citologia , Neurônios/efeitos dos fármacos , Ratos , Proteínas Recombinantes , Tomografia Computadorizada de Emissão de Fóton Único
8.
Semin Hematol ; 38(3 Suppl 7): 33-9, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11523026

RESUMO

Erythropoietin (EPO) primarily is produced in the kidney and acts as a principal mediator of the physiologic response to hypoxia by increasing red blood cell production. Astrocytes and neurons in the central nervous system (CNS) also are known to produce EPO in response to hypoxia/ischemia. EPO appears to play a neuroprotective role based on preclinical data demonstrating the ability of recombinant human erythropoietin (r-HuEPO) to shield neurons from hypoxic/ischemic stress when administered intracerebraventricularly. In CNS models, systemically administered r-HuEPO has not been intensely investigated because large glycosylated molecules generally were deemed incapable of crossing the blood-brain barrier (BBB). A collaborative research effort identified expression of EPO receptors on human brain capillaries and a specific receptor-mediated transport of r-HuEPO across the BBB after a single intraperitoneal (IP) injection in rodents, with subsequent protection against various types of neuronal damage. For example, administration of r-HuEPO 24 hours before or up to 6 hours after focal ischemic stroke significantly reduced the extent of infarction. r-HuEPO also attenuated concussive brain injury, kainate-induced seizure activity, and autoimmune encephalomyelitis. These preclinical findings suggest that r-HuEPO may have therapeutic potential for stroke, head trauma, and epilepsy; additional studies are needed to confirm and extend these encouraging observations in animal models.


Assuntos
Eritropoese/efeitos dos fármacos , Animais , Doenças do Sistema Nervoso Central/líquido cefalorraquidiano , Doenças do Sistema Nervoso Central/tratamento farmacológico , Doenças do Sistema Nervoso Central/fisiopatologia , Modelos Animais de Doenças , Eritropoetina/líquido cefalorraquidiano , Eritropoetina/uso terapêutico , Humanos , Proteínas Recombinantes
9.
Semin Oncol ; 28(2 Suppl 8): 66-70, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11395856

RESUMO

Erythropoietin (EPO) is a glycoprotein that has been shown to mediate response to hypoxia, and is most notably recognized for its central role in erythropoiesis. In a series of experiments using rodent models, the ability of systemically administered recombinant human erythropoietin (r-HuEPO, epoetin alfa) to cross the blood-brain barrier and affect the outcome of neuronal injury or cognitive function was evaluated. It was shown that EPO and EPO receptors are expressed at capillaries of the brain-periphery interface, and that systemically administered epoetin alfa crossed the blood-brain barrier. Compared with control animals, epoetin alfa significantly reduced tissue damage in an ischemic stroke model when administered 24 hours before inducing stroke, with significant protection still evident when epoetin alfa was administered 6 hours poststroke. Epoetin alfa reduced injury by blunt trauma when administered 24 hours before trauma, with a significantly smaller volume of tissue necrosis noted when compared with controls. The observation that epoetin alfa may reduce nervous system inflammation was confirmed when an experimental autoimmune encephalomyelitis model in which rats were shown to have significantly delayed onset and reduced severity of experimental autoimmune encephalomyelitis symptoms after treatment with epoetin alfa. Epoetin alfa also was shown to ameliorate the latency and severity of seizures, and significantly increase survival versus controls when exposed to kainate. These findings suggest future potential therapeutic uses for epoetin alfa beyond its current use to increase erythropoiesis.


Assuntos
Encéfalo/metabolismo , Sistema Nervoso Central/efeitos dos fármacos , Eritropoetina/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptores da Eritropoetina/metabolismo , Animais , Barreira Hematoencefálica , Encéfalo/efeitos dos fármacos , Isquemia Encefálica/prevenção & controle , Encefalomielite/prevenção & controle , Epoetina alfa , Eritropoetina/metabolismo , Humanos , Ácido Caínico , Camundongos , Modelos Animais , Fármacos Neuroprotetores/metabolismo , Proteínas Recombinantes , Convulsões/induzido quimicamente , Convulsões/prevenção & controle
10.
Proc Natl Acad Sci U S A ; 98(7): 4044-9, 2001 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-11259643

RESUMO

Erythropoietin (EPO) promotes neuronal survival after hypoxia and other metabolic insults by largely unknown mechanisms. Apoptosis and necrosis have been proposed as mechanisms of cellular demise, and either could be the target of actions of EPO. This study evaluates whether antiapoptotic mechanisms can account for the neuroprotective actions of EPO. Systemic administration of EPO (5,000 units/kg of body weight, i.p.) after middle-cerebral artery occlusion in rats dramatically reduces the volume of infarction 24 h later, in concert with an almost complete reduction in the number of terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling of neurons within the ischemic penumbra. In both pure and mixed neuronal cultures, EPO (0.1--10 units/ml) also inhibits apoptosis induced by serum deprivation or kainic acid exposure. Protection requires pretreatment, consistent with the induction of a gene expression program, and is sustained for 3 days without the continued presence of EPO. EPO (0.3 units/ml) also protects hippocampal neurons against hypoxia-induced neuronal death through activation of extracellular signal-regulated kinases and protein kinase Akt-1/protein kinase B. The action of EPO is not limited to directly promoting cell survival, as EPO is trophic but not mitogenic in cultured neuronal cells. These data suggest that inhibition of neuronal apoptosis underlies short latency protective effects of EPO after cerebral ischemia and other brain injuries. The neurotrophic actions suggest there may be longer-latency effects as well. Evaluation of EPO, a compound established as clinically safe, as neuroprotective therapy in acute brain injury is further supported.


Assuntos
Apoptose , Isquemia Encefálica/patologia , Eritropoetina/farmacologia , Neurônios Motores/efeitos dos fármacos , Estresse Fisiológico/patologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Neurônios Motores/citologia , Fatores de Crescimento Neural/farmacologia , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley
11.
Novartis Found Symp ; 235: 202-12; discussion 212-6, 217-20, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11280026

RESUMO

Recent studies have revealed that reducing sugars, such as glucose, react with proteins through non-enzymatic glycosylation to form irreversible, covalently cross-linked proteins known as advanced glycation endproducts (AGEs). Furthermore, it has been demonstrated that this naturally occurring process, accelerated in diabetics due to hyperglycaemia, impairs biological functions leading to cardiovascular disorders, as well as diabetic and age-related complications. Pharmaceutical intervention to prevent or reverse these complications have focused on inhibiting the formation of AGEs by compounds such as dimethyl-3-phenacylthiazolium chloride or breaking the glucose derived cross-links by selective cleavage. Intervention targeted at AGE cross-links in vivo offers a way to interfere with age-related changes of tissues.


Assuntos
Produtos Finais de Glicação Avançada/metabolismo , Tiazóis/metabolismo , Animais , Humanos , Reação de Maillard , Tiazóis/farmacologia , Tiazóis/uso terapêutico
12.
Recent Prog Horm Res ; 56: 1-21, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11237208

RESUMO

Biological amines react with reducing sugars to form a complex family of rearranged and dehydrated covalent adducts that are often yellow-brown and/or fluorescent and include many cross-linked structures. Food chemists have long studied this process as a source of flavor, color, and texture changes in cooked, processed, and stored foods. During the 1970s and 1980s, it was realized that this process, called the Maillard reaction or advanced glycation, also occurs slowly in vivo. Advanced glycation endproducts (AGEs) that form are implicated, causing the complications of diabetes and aging, primarily via adventitious and crosslinking of proteins. Long-lived proteins such as structural collagen and lens crystallins particularly are implicated as pathogenic targets of AGE processes. AGE formation in vascular wall collagen appears to be an especially deleterious event, causing crosslinking of collagen molecules to each other and to circulating proteins. This leads to plaque formation, basement membrane thickening, and loss of vascular elasticity. The chemistry of these later-stage, glycation-derived crosslinks is still incompletely understood but, based on the hypothesis that AGE formation involves reactive carbonyl groups, the authors introduced the carbonyl reagent aminoguanidine hydrochloride as an inhibitor of AGE formation in vivo in the mid 1980s. Subsequent studies by many researchers have shown the effectiveness of aminoguanidine in slowing or preventing a wide range of complications of diabetes and aging in animals and, recently, in humans. Since, the authors have developed a new class of agents, exemplified by 4,5-dimethyl-3-phenacylthiazolium chloride (DPTC), which can chemically break already-formed AGE protein-protein crosslinks. These agents are based on a new theory of AGE crosslinking that postulates that alpha-dicarbonyl structures are present in AGE protein-protein crosslinks. In studies in aged animals, DPTC has been shown to be capable of reverting indices of vascular compliance to levels seen in younger animals. Human clinical trials are underway.


Assuntos
Envelhecimento , Diabetes Mellitus/metabolismo , Produtos Finais de Glicação Avançada/fisiologia , Animais , Humanos , Modelos Químicos , Tiazóis/farmacologia
13.
Proc Natl Acad Sci U S A ; 98(3): 1171-5, 2001 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-11158613

RESUMO

Nonenzymatic glycosylation and cross-linking of proteins by glucose contributes to an age-associated increase in vascular and myocardial stiffness. Some recently sythesized thiazolium compounds selectively break these protein cross-links, reducing collagen stiffness. We investigated the effects of 3-phenacyl-4,5-dimethylthiazolium chloride (ALT-711) on arterial and left ventricular (LV) properties and their coupling in old, healthy, nondiabetic Macaca mulatta primates (age 21 +/- 3.6 years). Serial measurements of arterial stiffness indices [i.e., aortic pulse wave velocity (PWV) and augmentation (AGI) of carotid arterial pressure waveform] as well as echocardiographic determinations of LV structure and function were made before and for 39 weeks after 11 intramuscular injections of ALT-711 at 1.0 mg/kg body weight every other day. Heart rate, brachial blood pressure, and body weight were unchanged by the drug. PWV and AGI decreased to a nadir at 6 weeks [PWV to 74.2 +/- 4.4% of baseline (B), P = 0.007; AGI to 41 +/- 7.3% of B, P = 0.046], and thereafter gradually returned to baseline. Concomitant increases in LV end diastolic diameter to 116.7 +/- 2.7% of B, P = 0.02; stroke volume index (SV(index)) to 173.1 +/- 40.1% of B, P = 0.01; and systolic fractional shortening to 180 +/- 29.7% of B, P = 0.01 occurred after drug treatment. The LV end systolic pressure/SV(index), an estimate of total LV vascular load, decreased to 60 +/- 12.1% of B (P = 0.02). The LV end systolic diameter/SV(index), an estimate of arterio-ventricular coupling, was improved (decreased to 54.3 +/- 11% of B, P < 0.002). Thus, in healthy older primates without diabetes, ALT-711 improved both arterial and ventricular function and optimized ventriculo-vascular coupling. This previously unidentified cross-link breaker may be an effective pharmacological therapy to improve impaired cardiovascular function that occurs in the context of heart failure associated with aging, diabetes, or hypertension, conditions in which arterial and ventricular stiffness are increased.


Assuntos
Artérias/fisiologia , Ecocardiografia/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Tiazóis/farmacologia , Função Ventricular Esquerda/fisiologia , Envelhecimento , Animais , Artérias/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Produtos Finais de Glicação Avançada , Frequência Cardíaca/efeitos dos fármacos , Hemodinâmica/fisiologia , Injeções Intramusculares , Macaca mulatta , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Pulso Arterial , Volume Sistólico/efeitos dos fármacos , Tiazóis/administração & dosagem , Fatores de Tempo , Função Ventricular Esquerda/efeitos dos fármacos
14.
Proc Natl Acad Sci U S A ; 97(19): 10526-31, 2000 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-10984541

RESUMO

Erythropoietin (EPO), recognized for its central role in erythropoiesis, also mediates neuroprotection when the recombinant form (r-Hu-EPO) is directly injected into ischemic rodent brain. We observed abundant expression of the EPO receptor at brain capillaries, which could provide a route for circulating EPO to enter the brain. In confirmation of this hypothesis, systemic administration of r-Hu-EPO before or up to 6 h after focal brain ischemia reduced injury by approximately 50-75%. R-Hu-EPO also ameliorates the extent of concussive brain injury, the immune damage in experimental autoimmune encephalomyelitis, and the toxicity of kainate. Given r-Hu-EPO's excellent safety profile, clinical trials evaluating systemically administered r-Hu-EPO as a general neuroprotective treatment are warranted.


Assuntos
Lesões Encefálicas/prevenção & controle , Eritropoetina/farmacocinética , Fármacos Neuroprotetores/farmacocinética , Animais , Biotina/metabolismo , Barreira Hematoencefálica , Eritropoetina/metabolismo , Eritropoetina/uso terapêutico , Feminino , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes , Convulsões/induzido quimicamente , Convulsões/fisiopatologia , Convulsões/prevenção & controle
15.
Proc Natl Acad Sci U S A ; 97(6): 2809-13, 2000 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-10706607

RESUMO

Decreased elasticity of the cardiovascular system is one of the hallmarks of the normal aging process of mammals. A potential explanation for this decreased elasticity is that glucose can react nonenzymatically with long-lived proteins, such as collagen and lens crystallin, and link them together, producing advanced glycation endproducts (AGEs). Previous studies have shown that aminoguanidine, an AGE inhibitor, can prevent glucose cross-linking of proteins and the loss of elasticity associated with aging and diabetes. Recently, an AGE cross-link breaker (ALT-711) has been described, which we have evaluated in aged dogs. After 1 month of administration of ALT-711, a significant reduction ( approximately 40%) in age-related left ventricular stiffness was observed [(57.1 +/- 6.8 mmHg x m(2)/ml pretreatment and 33.1 +/- 4.6 mmHg x m(2)/ml posttreatment (1 mmHg = 133 Pa)]. This decrease was accompanied by improvement in cardiac function.


Assuntos
Envelhecimento/fisiologia , Produtos Finais de Glicação Avançada/antagonistas & inibidores , Miocárdio/metabolismo , Tiazóis/farmacologia , Animais , Diástole/fisiologia , Cães , Elasticidade , Frequência Cardíaca/fisiologia , Hemodinâmica , Masculino , Volume Sistólico , Sístole/fisiologia , Função Ventricular
16.
Proc Natl Acad Sci U S A ; 96(5): 2385-90, 1999 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-10051651

RESUMO

Epidemiological studies suggest that there is a beneficial effect of moderate ethanol consumption on the incidence of cardiovascular disease. Ethanol is metabolized to acetaldehyde, a two-carbon carbonyl compound that can react with nucleophiles to form covalent addition products. We have identified a biochemical modification produced by the reaction of acetaldehyde with protein-bound Amadori products. Amadori products typically arise from the nonenzymatic addition of reducing sugars (such as glucose) to protein amino groups and are the precursors to irreversibly bound, crosslinking moieties called advanced glycation endproducts, or AGEs. AGEs accumulate over time on plasma lipoproteins and vascular wall components and play an important role in the development of diabetes- and age-related cardiovascular disease. The attachment of acetaldehyde to a model Amadori product produces a chemically stabilized complex that cannot rearrange and progress to AGE formation. We tested the role of this reaction in preventing AGE formation in vivo by administering ethanol to diabetic rats, which normally exhibit increased AGE formation and high circulating levels of the hemoglobin Amadori product, HbA1c, and the hemoglobin AGE product, Hb-AGE. In this model study, diabetic rats fed an ethanol diet for 4 weeks showed a 52% decrease in Hb-AGE when compared with diabetic controls (P < 0.001). Circulating levels of HbA1c were unaffected by ethanol, pointing to the specificity of the acetaldehyde reaction for the post-Amadori, advanced glycation process. These data suggest a possible mechanism for the so-called "French paradox," (the cardioprotection conferred by moderate ethanol ingestion) and may offer new strategies for inhibiting advanced glycation.


Assuntos
Acetaldeído/sangue , Consumo de Bebidas Alcoólicas , Diabetes Mellitus Experimental/sangue , Etanol/farmacologia , Produtos Finais de Glicação Avançada/sangue , Hemoglobinas/metabolismo , Acetaldeído/química , Acetaldeído/farmacologia , Animais , Cardiotônicos , Etanol/metabolismo , Hemoglobinas Glicadas/metabolismo , Produtos Finais de Glicação Avançada/antagonistas & inibidores , Produtos Finais de Glicação Avançada/metabolismo , Masculino , Ratos , Ratos Wistar , Espectrometria de Massa de Íon Secundário , Fatores de Tempo
17.
Novartis Found Symp ; 226: 265-77; discussion 277-80, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10645551

RESUMO

The biochemical mechanism of chloroquine resistance in Plasmodium falciparum remains unknown. We postulated that chloroquine-resistant strains could alter ion fluxes that then indirectly control drug accumulation within the parasite by affecting pH and/or membrane potential ('altered partitioning mechanism'). Two principal intracellular pH-regulating systems in many cell types are the amiloride-sensitive Na+/H+ exchanger (NHE), and the sodium-independent, stilbene-sensitive Cl-/HCO3- antiporter (AE). We report that under physiological conditions (balanced CO2 and HCO3-) chloroquine uptake and susceptibility are not altered by amiloride analogues. We also do not detect a significant difference in NHE activity between chloroquine-sensitive and chloroquine-resistant strains via single cell photometry methods. AE activity is dependent on the intracellular and extracellular concentrations of Cl- and HCO3- ions. Chloroquine-resistant strains differentially respond to experimental modifications in chloride-dependent homeostasis, including growth, cytoplasmic pH and pH regulation. Chloroquine susceptibility is altered by stilbene DIDS only on chloroquine-resistant strains. Our results suggest that a Cl(-)-dependent system (perhaps AE) has a significant effect on the uptake of chloroquine by the infected erythrocyte, and that alterations of this biophysical parameter may be part of the mechanism of chloroquine resistance in P. falciparum.


Assuntos
Antimaláricos/farmacocinética , Cloretos/metabolismo , Cloroquina/farmacocinética , Plasmodium falciparum/metabolismo , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Amilorida/farmacologia , Animais , Antimaláricos/uso terapêutico , Diuréticos/farmacologia , Resistência a Medicamentos , Trocadores de Sódio-Hidrogênio/metabolismo
18.
Mol Med ; 4(10): 648-57, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9848081

RESUMO

The macrophage occupies a central role in the host response to invasion, exerting its control over the developing inflammatory response largely through the elaboration of an assortment of endogenous mediators including many cytokines. The beta chemokine peptides, macrophage inflammatory protein [MIP]-1 alpha and MIP-1 beta, are two such effectors markedly up-regulated in macrophages following exposure to bacterial lipopolysaccharide (LPS). These highly homologous peptides, like the other members of the beta chemokine family, exhibit diverse but partially overlapping biological activity profiles, suggesting that the cellular participants and intensity of an inflammatory response may in part be regulated by selective expression of these chemokines. Studies reported here demonstrate that, in contrast to the "balanced" MIP-1 alpha/MIP-1 beta chemokine responses of LPS-stimulated macrophage cultures in vitro, circulating levels of MIP-1 beta are significantly higher than those of MIP-1 alpha following LPS administration in vivo. Further studies have revealed that several immunomodulatory cytokines known to be up-regulated in vivo as a consequence of exposure to an invasive stimulus (gamma-IFN, IL-10, IL-4, and transforming growth factor [TGF]-beta) down-regulated the LPS-induced release of MIP-1 alpha by macrophages in vitro, but spared the MIP-1 beta response. This altered pattern of secretion may explain, at least in part, the high circulating levels of MIP-1 beta relative to MIP-1 alpha observed in vivo in response to LPS challenge.


Assuntos
Interferon gama/imunologia , Interleucina-10/imunologia , Interleucina-4/imunologia , Proteínas Inflamatórias de Macrófagos/biossíntese , Macrófagos/imunologia , Fator de Crescimento Transformador beta/imunologia , Animais , Quimiocina CCL4 , Regulação para Baixo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-10/farmacologia , Interleucina-4/farmacologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Células Th2/imunologia , Fator de Crescimento Transformador beta/farmacologia
19.
J Immunol ; 160(11): 5588-95, 1998 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9605164

RESUMO

Multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), are characterized by episodic neurologic dysfunction, perivascular mononuclear cell inflammation occurring mainly in white matter, and demyelination. Strong circumstantial evidence supports the conclusion that macrophage activation and local production of proinflammatory cytokines are necessary for disease induction and lesion formation. We now report that CNI-1493, a small m.w. compound, which inhibits macrophage activation and subsequent proinflammatory cytokine production, suppresses EAE induced in the genetically susceptible SJL/J mouse. Treatment with 5 mg/kg/day completely suppressed mild disease (clinical index of 1.6 +/- 0.5 in the untreated group as compared with 0.0 +/- 0.0 for the treated group) and significantly reduced acute disease (clinical index of 4.3 +/- 0.7 in the untreated group as compared with 0.5 +/- 0.3 for the treated group). Suppression of clinical manifestations of the disease correlated with a significant decrease in histopathology and proinflammatory cytokine expression at the lesion site. Moreover, drug treatment during the chronic phase resulted in amelioration of clinical signs. The data presented here should prove useful in developing novel chemotherapeutic approaches for the treatment of MS.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/prevenção & controle , Hidrazonas/uso terapêutico , Imunossupressores/uso terapêutico , Ativação de Macrófagos/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Doença Crônica , Citocinas/antagonistas & inibidores , Citocinas/genética , Regulação para Baixo/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Injeções Intraperitoneais , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , RNA Mensageiro/antagonistas & inibidores , Recidiva , Medula Espinal/metabolismo , Linfócitos T/imunologia
20.
Antimicrob Agents Chemother ; 42(5): 1133-8, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9593140

RESUMO

Active nuclear importation of the human immunodeficiency virus (HIV) type 1 (HIV-1) preintegration complex (PIC) is required for the productive infection of nondividing cells, but it is believed to be dispensable for the infection of proliferating cells, such as activated T lymphocytes. To investigate this question, we exploited the properties of the small arylene bis (methyl ketone) compound CNI-H0294. We have previously shown that this compound associated with the HIV-1 matrix protein nuclear localization sequence and blocked binding of the HIV-1 PIC to yeast karyopherin alpha. CNI-H0294 abrogated nuclear importation of the HIV-1 genome in macrophages and effectively inhibited infection of nondividing cells. In this study we demonstrate that CNI-H0294 inhibits binding of the HIV-1 PIC to human karyopherin alpha and reduces nuclear importation of the viral genome in primary peripheral blood mononuclear cells (PBMCs). We also demonstrate that CNI-H0294 inhibits acute infection of PBMC cultures in vitro with a primary isolate of HIV-1 and reduces virus replication and virus load in cultures of endogenously infected PBMCs from seropositive individuals. Thus, as for infection of nondividing, terminally differentiated macrophages, HIV-1 uses active nuclear importation of the virus genome to infect activated CD4+ T cells. These results support nuclear importation as a novel target and CNI-H0294 and its derivatives as novel compounds for therapeutic intervention in HIV infection and AIDS.


Assuntos
Fármacos Anti-HIV/farmacologia , HIV-1/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Pirimidinas/farmacologia , Replicação Viral/efeitos dos fármacos , Sítios de Ligação/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/virologia , Soropositividade para HIV/virologia , HIV-1/crescimento & desenvolvimento , HIV-1/metabolismo , Humanos , Leucócitos Mononucleares/virologia , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/fisiologia , alfa Carioferinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...