Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Semin Perinatol ; 46(5): 151598, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35589461

RESUMO

In this report, we summarize evidence on mechanisms of injury after intraventricular hemorrhage resulting in post-hemorrhagic white matter injury and hydrocephalus and correlate that with the possibility of cellular therapy. We describe how two stem cell lines (MSC & USSC) acting in a paracrine fashion offer promise for attenuating the magnitude of injury in animal models and for improved functional recovery by: lowering the magnitude of apoptosis and neuronal cell death, reducing inflammation, and thus, mitigating white matter injury that culminates in improved motor and neurocognitive outcomes. Animal models of IVH are analyzed for their similarity to the human condition and we discuss merits of each approach. Studies on stem cell therapy for IVH in human neonates is described. Lastly, we offer suggestions on what future studies are needed to better understand mechanisms of injury and recovery and argue that human trials need to be expanded in parallel to animal research.


Assuntos
Células-Tronco Adultas , Células-Tronco Mesenquimais , Animais , Encéfalo , Sangue Fetal , Humanos , Recém-Nascido , Células-Tronco Mesenquimais/metabolismo , Transplante de Células-Tronco
2.
Stem Cells Transl Med ; 8(11): 1157-1169, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31322326

RESUMO

Intraventricular hemorrhage (IVH) is a severe complication of preterm birth, which leads to hydrocephalus, cerebral palsy, and mental retardation. There are no available therapies to cure IVH, and standard treatment is supportive care. Unrestricted somatic stem cells (USSCs) from human cord blood have reparative effects in animal models of brain and spinal cord injuries. USSCs were administered to premature rabbit pups with IVH and their effects on white matter integrity and neurobehavioral performance were evaluated. USSCs were injected either via intracerebroventricular (ICV) or via intravenous (IV) routes in 3 days premature (term 32d) rabbit pups, 24 hours after glycerol-induced IVH. The pups were sacrificed at postnatal days 3, 7, and 14 and effects were compared to glycerol-treated but unaffected or nontreated control. Using in vivo live bioluminescence imaging and immunohistochemical analysis, injected cells were found in the injured parenchyma on day 3 when using the IV route compared to ICV where cells were found adjacent to the ventricle wall forming aggregates; we did not observe any adverse events from either route of administration. The injected USSCs were functionally associated with attenuated microglial infiltration, less apoptotic cell death, fewer reactive astrocytes, and diminished levels of key inflammatory cytokines (TNFα and IL1ß). In addition, we observed better preservation of myelin fibers, increased myelin gene expression, and altered reactive astrocyte distribution in treated animals, and this was associated with improved locomotor function. Overall, our findings support the possibility that USSCs exert anti-inflammatory effects in the injured brain mitigating many detrimental consequences associated with IVH. Stem Cells Translational Medicine 2019;8:1157-1169.


Assuntos
Células-Tronco Adultas/citologia , Comportamento Animal , Hemorragia Cerebral/complicações , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Modelos Animais de Doenças , Sangue Fetal/citologia , Transtornos Neurocognitivos/prevenção & controle , Animais , Humanos , Transtornos Neurocognitivos/etiologia , Testes Neuropsicológicos , Coelhos
3.
J Neurosci ; 36(3): 872-89, 2016 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-26791217

RESUMO

Intraventricular hemorrhage (IVH) in premature infants results in inflammation, arrested oligodendrocyte progenitor cell (OPC) maturation, and reduced myelination of the white matter. Hyaluronan (HA) inhibits OPC maturation and complexes with the heavy chain (HC) of glycoprotein inter-α-inhibitor to form pathological HA (HC-HA complex), which exacerbates inflammation. Therefore, we hypothesized that IVH would result in accumulation of HA, and that either degradation of HA by hyaluronidase treatment or elimination of HCs from pathological HA by HA oligosaccharide administration would restore OPC maturation, myelination, and neurological function in survivors with IVH. To test these hypotheses, we used the preterm rabbit model of glycerol-induced IVH and analyzed autopsy samples from premature infants. We found that total HA levels were comparable in both preterm rabbit pups and human infants with and without IVH, but HA receptors--CD44, TLR2, TLR4--were elevated in the forebrain of both humans and rabbits with IVH. Hyaluronidase treatment of rabbits with IVH reduced CD44 and TLR4 expression, proinflammatory cytokine levels, and microglia infiltration. It also promoted OPC maturation, myelination, and neurological recovery. HC-HA and tumor necrosis factor-stimulated gene-6 were elevated in newborns with IVH; and depletion of HC-HA levels by HA oligosaccharide treatment reduced inflammation and enhanced myelination and neurological recovery in rabbits with IVH. Hence, hyaluronidase or HA oligosaccharide treatment represses inflammation, promotes OPC maturation, and restores myelination and neurological function in rabbits with IVH. These therapeutic strategies might improve the neurological outcome of premature infants with IVH. Significance statement: Approximately 12,000 premature infants develop IVH every year in the United States, and a large number of survivors with IVH develop cerebral palsy and cognitive deficits. The onset of IVH induces inflammation of the periventricular white matter, which results in arrested maturation of OPCs and myelination failure. HA is a major component of the extracellular matrix of the brain, which regulates inflammation through CD44 and TLR2/4 receptors. Here, we show two mechanism-based strategies that effectively enhanced myelination and neurological recovery in preterm rabbit model of IVH. First, degrading HA by hyaluronidase treatment reduced CD44 and TLR4 expression, proinflammatory cytokines, and microglial infiltration, as well as promoted oligodendrocyte maturation and myelination. Second, intraventricular injection of HA oligosaccharide reduced inflammation and enhanced myelination, conceivably by depleting HC-HA levels.


Assuntos
Hemorragia Cerebral/metabolismo , Ventrículos Cerebrais/metabolismo , Ácido Hialurônico/biossíntese , Hialuronoglucosaminidase/biossíntese , Oligossacarídeos/biossíntese , Recuperação de Função Fisiológica/fisiologia , Animais , Animais Recém-Nascidos , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/patologia , Ventrículos Cerebrais/efeitos dos fármacos , Ventrículos Cerebrais/patologia , Feminino , Humanos , Ácido Hialurônico/administração & dosagem , Recém-Nascido , Injeções Intraventriculares , Masculino , Oligossacarídeos/administração & dosagem , Gravidez , Coelhos , Recuperação de Função Fisiológica/efeitos dos fármacos
4.
J Neurosci ; 33(2): 411-23, 2013 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-23303921

RESUMO

Premature infants exhibit neurodevelopmental delay and reduced growth of the cerebral cortex. However, the underlying mechanisms have remained elusive. Therefore, we hypothesized that neurogenesis in the ventricular and subventricular zones of the cerebral cortex would continue in the third trimester of pregnancy and that preterm birth would suppress neurogenesis. To test our hypotheses, we evaluated autopsy materials from human fetuses and preterm infants of 16-35 gestational weeks (gw). We noted that both cycling and noncycling Sox2(+) radial glial cells and Tbr2(+) intermediate progenitors were abundant in human preterm infants until 28 gw. However, their densities consistently decreased from 16 through 28 gw. To determine the effect of premature birth on neurogenesis, we used a rabbit model and compared preterm [embryonic day 29 (E29), 3 d old] and term (E32, <2 h old) pups at an equivalent postconceptional age. Glutamatergic neurogenesis was suppressed in preterm rabbits, as indicated by the reduced number of Tbr2(+) intermediate progenitors and the increased number of Sox2(+) radial glia. Additionally, hypoxia-inducible factor-1α, vascular endothelial growth factor, and erythropoietin were higher in term than preterm pups, reflecting the hypoxic intrauterine environment of just-born term pups. Proneural genes, including Pax6 and Neurogenin-1 and -2, were higher in preterm rabbit pups compared with term pups. Importantly, neurogenesis and associated factors were restored in preterm pups by treatment with dimethyloxallyl glycine, a hypoxia mimetic agent. Hence, glutamatergic neurogenesis continues in the premature infants, preterm birth suppresses neurogenesis, and hypoxia-mimetic agents might restore neurogenesis, enhance cortical growth, and improve neurodevelopmental outcome of premature infants.


Assuntos
Neurogênese/fisiologia , Terceiro Trimestre da Gravidez/fisiologia , Nascimento Prematuro/fisiopatologia , Adulto , Animais , Contagem de Células , Ventrículos Cerebrais/crescimento & desenvolvimento , Eritropoetina/fisiologia , Feminino , Idade Gestacional , Glicina/farmacologia , Humanos , Hipóxia/fisiopatologia , Fator 1 Induzível por Hipóxia/biossíntese , Fator 1 Induzível por Hipóxia/fisiologia , Imuno-Histoquímica , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Proteínas do Tecido Nervoso/biossíntese , Células-Tronco Neurais/fisiologia , Gravidez , Coelhos , Transdução de Sinais/fisiologia , Telencéfalo/crescimento & desenvolvimento , Fator A de Crescimento do Endotélio Vascular/fisiologia , Proteínas Wnt/fisiologia , beta Catenina/fisiologia
5.
Stroke ; 41(8): 1766-73, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20616316

RESUMO

BACKGROUND AND PURPOSE: Prenatal glucocorticoids prevent germinal matrix hemorrhage in premature infants. The underlying mechanism, however, is elusive. Germinal matrix is enriched with angiogenic vessels exhibiting paucity of pericytes and glial fibrillary acidic protein-positive astrocyte end feet. Therefore, we asked whether glucocorticoid treatment would suppress angiogenesis and enhance periendothelial coverage by pericytes and glial fibrillary acidic protein-positive end feet in the germinal matrix microvasculature. METHODS: We treated pregnant rabbits with intramuscular betamethasone and delivered pups prematurely by cesarean section at E29 (term=32 days). Endothelial turnover, vascular density, pericyte coverage, glial fibrillary acidic protein-positive end feet, cell death, and growth factors orchestrating angiogenesis, including vascular endothelial growth factor, angiopoietins, transforming growth factor-beta, and platelet-derived growth factor-B, were compared between betamethasone-treated and untreated pups. Similar comparisons were done between autopsy materials from premature infants exposed and unexposed to prenatal glucocorticoids. RESULTS: Antenatal glucocorticoid treatment reduced endothelial proliferation, vascular density, and vascular endothelial growth factor expression in the germinal matrix of both rabbits and humans. The pericyte coverage was greater in glucocorticoid-treated rabbit pups and human infants than in controls, but not the glial fibrillary acidic protein-positive end feet coverage. Transforming growth factor-beta, but not angiopoietins and platelet-derived growth factor-B, were elevated in glucocorticoid-treated rabbit pups compared with controls. Betamethasone treatment induced apoptosis, neuronal degeneration, and gliosis in rabbit pups. However, there was no evidence of increased cell death in glucocorticoid-exposed human infants. CONCLUSIONS: Prenatal glucocorticoid suppresses vascular endothelial growth factor and elevates transforming growth factor-beta levels, which results in angiogenic inhibition, trimming of neovasculature, and enhanced pericyte coverage. These changes contribute to stabilizing the germinal matrix vasculature, thereby reducing its propensity to hemorrhage. Prenatal glucocorticoid exposure does not induce neural cell death in humans, unlike rabbits.


Assuntos
Betametasona/farmacologia , Circulação Cerebrovascular/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Proliferação de Células/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Gliose/metabolismo , Glucocorticoides/farmacologia , Humanos , Imuno-Histoquímica , Recém-Nascido , Microdissecção , Degeneração Neural/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Brain ; 133(Pt 8): 2264-80, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20488889

RESUMO

Intraventricular haemorrhage is a major complication of prematurity that results in neurological dysfunctions, including cerebral palsy and cognitive deficits. No therapeutic options are currently available to limit the catastrophic brain damage initiated by the development of intraventricular haemorrhage. As intraventricular haemorrhage leads to an inflammatory response, we asked whether cyclooxygenase-2, its derivative prostaglandin E2, prostanoid receptors and pro-inflammatory cytokines were elevated in intraventricular haemorrhage; whether their suppression would confer neuroprotection; and determined how cyclooxygenase-2 and cytokines were mechanistically-linked. To this end, we used our rabbit model of intraventricular haemorrhage where premature pups, delivered by Caesarian section, were treated with intraperitoneal glycerol at 2 h of age to induce haemorrhage. Intraventricular haemorrhage was diagnosed by head ultrasound at 6 h of age. The pups with intraventricular haemorrhage were treated with inhibitors of cyclooxygenase-2, prostanoid receptor-1 or tumour necrosis factor-α; and cell-infiltration, cell-death and gliosis were compared between treated-pups and vehicle-treated controls during the first 3 days of life. Neurobehavioural performance, myelination and gliosis were assessed in pups treated with cyclooxygenase-2 inhibitor compared to controls at Day 14. We found that both protein and messenger RNA expression of cyclooxygenase-2, prostaglandin E2, prostanoid receptor-1, tumour necrosis factor-α and interleukin-1ß were consistently higher in the forebrain of pups with intraventricular haemorrhage relative to pups without intraventricular haemorrhage. However, cyclooxygenase-1 and prostanoid receptor 2-4 levels were comparable in pups with and without intraventricular haemorrhage. Cyclooxygenase-2, prostanoid receptor-1 or tumour necrosis factor-α inhibition reduced inflammatory cell infiltration, apoptosis, neuronal degeneration and gliosis around the ventricles of pups with intraventricular haemorrhage. Importantly, cyclooxygenase-2 inhibition alleviated neurological impairment, improved myelination and reduced gliosis at 2 weeks of age. Cyclooxygenase-2 or prostanoid receptor-1 inhibition reduced tumour necrosis factor-α level, but not interleukin-1ß. Conversely, tumour necrosis factor-α antagonism did not affect cyclooxygenase-2 expression. Hence, prostanoid receptor-1 and tumour necrosis factor-α are downstream to cyclooxygenase-2 in the inflammatory cascade induced by intraventricular haemorrhage, and cyclooxygenase-2-inhibition or suppression of downstream molecules--prostanoid receptor-1 or tumour necrosis factor-α--might be a viable neuroprotective strategy for minimizing brain damage in premature infants with intraventricular haemorrhage.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , Hemorragias Intracranianas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Receptores de Prostaglandina E/antagonistas & inibidores , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Ventrículos Cerebrais , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Modelos Animais de Doenças , Gliose/tratamento farmacológico , Gliose/metabolismo , Gliose/patologia , Interleucina-1beta/metabolismo , Hemorragias Intracranianas/metabolismo , Hemorragias Intracranianas/patologia , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/metabolismo , Coelhos , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP1 , Fator de Necrose Tumoral alfa/metabolismo
7.
Clin Chim Acta ; 405(1-2): 127-31, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19394322

RESUMO

BACKGROUND: Hyperhomocysteinemia, a documented risk factor for CAD is highly prevalent in Indians. The rationale behind the current study is to explore the genetic and environmental causes for such high prevalence as there are limited studies in this context. METHODS: A total of 108 CAD cases and 108 controls were analyzed for tHcy and 4 folate pathway genetic polymorphisms [methylene tetrahydrofolate reductase (MTHFR) C677T, 5-methyltetrahydrofolate homocysteine methyl transferase (MTR) A2756G, methionine synthase reductase (MTRR) A66G and glutamate carboxypeptidase II (GCPII) C1561T] using reverse phase HPLC and PCR-RFLP methods respectively. RESULTS: MTHFR 677T, MTRR 66A, GCPII 1561T, male gender, alcohol intake, smoking, diabetes, creatinine and hypertension were found to influence tHcy. After controlling for confounding factors, Hyperhomocysteinemia and two of its genetic determinants i.e. MTHFR C677T [OR: 1.96, 95% CI: 1.06-3.61] and GCP II C1561T [OR: 2.09, 95% CI: 1.09-3.97] were found to be associated with risk for CAD. Significant epistatic interactions were observed between MTHFR 677T/MTR 2756G and GCP II 1561T/MTRR 66G. Alcohol intake in subjects with MTR 2756G allele was found to inflate the risk for CAD [OR: 4.15, 95% CI: 1.35-12.69]. CONCLUSION: Hyperhomocysteinemia, C677T MTHFR and C1561T GCPII are risk factors for CAD. Potential gene--gene and gene--environment interactions indicate the need for multi-variate analyses for risk prediction.


Assuntos
Doença das Coronárias/sangue , Doença das Coronárias/genética , Homocisteína/sangue , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Adulto , Idoso , Alelos , Doença das Coronárias/epidemiologia , Feminino , Ferredoxina-NADP Redutase/genética , Ferredoxina-NADP Redutase/metabolismo , Genótipo , Glutamato Carboxipeptidase II/genética , Glutamato Carboxipeptidase II/metabolismo , Humanos , Índia/epidemiologia , Masculino , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco
8.
Biochim Biophys Acta ; 1759(7): 340-7, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16949687

RESUMO

Mechanisms to maintain blood pressure in the face of infection are critical to survival. The angiotensinogen (AGT) gene locus is an important component of this response. Thus the AGT gene, expressed predominantly by liver cells, is known to be a positive acute phase reactant. We have previously demonstrated activation of the AGT promoter in hepatocytes through the IL6/STAT3 signaling mechanism. We have now investigated whether IFN-gamma, a cytokine also induced in response to diverse infections, can regulate AGT gene expression, and have elucidated the molecular mechanism involved. IFN gamma treatment up-regulated AGT mRNA level and promoter activity in Hep3B hepatocytes. Sequential deletion of the promoter from the 5' side suggested the major IFN gamma responsive DNA element to be between -303 and -103. This region contained a candidate STAT1-binding site between -271 and -279. EMSA and chromatin immuno-precipitation (ChIP) assays confirmed that IFN-gamma treatment induced the binding of STAT1 to this element. Reporter constructs containing this AGT promoter derived element in a multimerized context but not a mutant version were responsive to IFN gamma. Moreover mutating this STAT1 element in the context of the wild-type AGT holo promoter reduced responsiveness to IFN gamma. In contrast to the clear synergism between dexamethasone and IL 6 in the upregulation of the AGT promoter (through interaction between GR and STAT3), the combination of IFN gamma with IL 6 or with dexamethasone did not further increase AGT promoter activity suggesting that the IFN gamma/STAT1 pathway represents a separate signaling mechanism. These data highlight the redundancy in cytokine-mediated host response pathways aimed at the maintenance of blood pressure during infection.


Assuntos
Angiotensinogênio/genética , Regulação da Expressão Gênica , Interferon gama/farmacologia , Fator de Transcrição STAT1/metabolismo , Transcrição Gênica/efeitos dos fármacos , Sítios de Ligação , Células Cultivadas , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Hepatócitos/metabolismo , Humanos , Interleucina-6/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Elementos de Resposta/efeitos dos fármacos , Deleção de Sequência , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA