Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
J Alzheimers Dis ; 93(2): 683-704, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37066912

RESUMO

BACKGROUND: Prior experience in early life has been shown to improve performance in aging and mice with Alzheimer's disease (AD) pathology. However, whether cognitive training at a later life stage would benefit subsequent cognition and reduce pathology in AD mice needs to be better understood. OBJECTIVE: This study aimed to verify if behavioral training in mid-adulthood would improve subsequent cognition and reduce AD pathology and astrogliosis. METHODS: Mixed-sex APP/PS1 and wildtype littermate mice received a battery of behavioral training, composed of spontaneous alternation in the Y-maze, novel object recognition and location tasks, and spatial training in the water maze, or handling only at 7 months of age. The impact of AD genotype and prior training on subsequent learning and memory of aforementioned tasks were assessed at 9 months. RESULTS: APP/PS1 mice made more errors than wildtype littermates in the radial-arm water maze (RAWM) task. Prior training prevented this impairment in APP/PS1 mice. Prior training also contributed to better efficiency in finding the escape platform in both APP/PS1 mice and wildtype littermates. Short-term and long-term memory of this RAWM task, of a reversal task, and of a transfer task were comparable among APP/PS1 and wildtype mice, with or without prior training. Amyloid pathology and astrogliosis in the hippocampus were also comparable between the APP/PS1 groups. CONCLUSION: These data suggest that cognitive training in mid-adulthood improves subsequent accuracy in AD mice and efficiency in all mice in the spatial task. Cognitive training in mid-adulthood provides no clear benefit on memory or on amyloid pathology in midlife.


Assuntos
Doença de Alzheimer , Humanos , Camundongos , Animais , Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Camundongos Transgênicos , Gliose/terapia , Gliose/patologia , Treino Cognitivo , Hipocampo/patologia , Modelos Animais de Doenças , Aprendizagem em Labirinto , Presenilina-1/genética , Presenilina-1/metabolismo , Peptídeos beta-Amiloides/metabolismo , Camundongos Endogâmicos C57BL
2.
Metab Brain Dis ; 37(7): 2315-2329, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35778625

RESUMO

Therapeutic hypothermia (TH) is the standard treatment for neonatal hypoxia-ischemia (HI) with a time window limited up to 6 h post injury. However, influence of sexual dimorphism in the therapeutic window for TH has not yet been elucidated in animal models of HI. Therefore, the aim of this study was to investigate the most effective time window to start TH in male and female rats submitted to neonatal HI. Wistar rats (P7) were divided into the following groups: NAÏVE and SHAM (control groups), HI (submitted to HI) and TH (submitted to HI and TH; 32ºC for 5 h). TH was started at 2 h (TH-2 h group), 4 h (TH-4 h group), or 6 h (TH-6 h group) after HI. At P14, animals were subjected to behavioural tests, volume of lesion and reactive astrogliosis assessments. Male and female rats from the TH-2 h group showed reduction in the latency of behavioral tests, and decrease in volume of lesion and intensity of GFAP immunofluorescence. TH-2 h females also showed reduction of degenerative cells and morphological changes in astrocytes. Interestingly, females from the TH-6 h group showed an increase in volume of lesion and in number of degenerative hippocampal cells, associated with worse behavioral performance. Together, these results indicate that TH neuroprotection is time- and sex-dependent. Moreover, TH started later (6 h) can worsen volume of brain lesion in females. These data indicate the need to develop specific therapeutic protocols for each sex and reinforce the importance of early onset of the hypothermic treatment.


Assuntos
Hipotermia Induzida , Hipóxia-Isquemia Encefálica , Animais , Masculino , Feminino , Ratos , Hipóxia-Isquemia Encefálica/terapia , Hipóxia-Isquemia Encefálica/patologia , Gliose/terapia , Gliose/patologia , Ratos Wistar , Animais Recém-Nascidos , Encéfalo , Isquemia/patologia , Isquemia/terapia , Modelos Animais de Doenças
3.
J Neuroinflammation ; 19(1): 139, 2022 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-35690757

RESUMO

BACKGROUND: Therapeutic hypothermia significantly improves outcomes after moderate-severe hypoxic-ischemic encephalopathy (HIE), but it is partially effective. Although hypothermia is consistently associated with reduced microgliosis, it is still unclear whether it normalizes microglial morphology and phenotype. METHODS: Near-term fetal sheep (n = 24) were randomized to sham control, ischemia-normothermia, or ischemia-hypothermia. Brain sections were immunohistochemically labeled to assess neurons, microglia and their interactions with neurons, astrocytes, myelination, and gitter cells (microglia with cytoplasmic lipid granules) 7 days after cerebral ischemia. Lesions were defined as areas with complete loss of cells. RNAscope® was used to assess microglial phenotype markers CD86 and CD206. RESULTS: Ischemia-normothermia was associated with severe loss of neurons and myelin (p < 0.05), with extensive lesions, astrogliosis and microgliosis with a high proportion of gitter cells (p < 0.05). Microglial wrapping of neurons was present in both the ischemia groups. Hypothermia improved neuronal survival, suppressed lesions, gitter cells and gliosis (p < 0.05), and attenuated the reduction of myelin area fraction. The "M1" marker CD86 and "M2" marker CD206 were upregulated after ischemia. Hypothermia partially suppressed CD86 in the cortex only (p < 0.05), but did not affect CD206. CONCLUSIONS: Hypothermia prevented lesions after cerebral ischemia, but only partially suppressed microglial wrapping and M1 marker expression. These data support the hypothesis that persistent upregulation of injurious microglial activity may contribute to partial neuroprotection after hypothermia, and that immunomodulation after rewarming may be an important therapeutic target.


Assuntos
Hipotermia Induzida , Hipotermia , Hipóxia-Isquemia Encefálica , Substância Branca , Animais , Gliose/terapia , Hipóxia-Isquemia Encefálica/metabolismo , Inflamação/terapia , Isquemia , Ovinos , Substância Branca/patologia
4.
Mol Cell Neurosci ; 118: 103694, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34954382

RESUMO

Rupture and stretching of spinal roots are common incidents that take place in high-energy accidents. The proximal axotomy of motoneurons by crushing of ventral roots is directly related to the degeneration of half of the lesioned population within the first two weeks. Moreover, only a small percentage of surviving motoneurons can successfully achieve regeneration after such a proximal lesion, and new treatments are necessary to improve this scenario. In this sense, mesenchymal stem cells (MSC) are of great interest once they secrete a broad spectrum of bioactive molecules that are immunomodulatory and can restore the environment after a lesion. The present work aimed at studying the effects of human mesenchymal stem cells (hMSC) therapy after ventral root crush (VRC) in mice. We evaluated motoneuron survival, glial reaction, and synapse preservation at the ventral horn. For this purpose, C57BL/6 J were submitted to a crush procedure of L4 to L6 ventral roots and treated with a single intravenous injection of adipose-derived hMSC. Evaluation of the results was carried out at 7, 14, and 28 days after injury. Analysis of motoneuron survival and astrogliosis showed that hMSC treatment resulted in higher motoneuron preservation (motoneuron survival ipsi/contralateral ratio: VRC group = 53%, VRC + hMSC group = 66%; p < 0.01), combined with reduction of astrogliosis (ipsi/contralateral GFAP immunolabeling: VRC group = 470%, VRC + hMSC group = 250%; p < 0.001). The morphological classification and Sholl analysis of microglial activation revealed that hMSC treatment reduced type V and increased type II profiles, indicating an enhancement of surveying over activated microglial cells. The glial reactivity modulation directly influenced synaptic inputs in apposition to axotomized motoneurons. In the hMSC-treated group, synaptic maintenance was increased (ipsi/contralateral synaptophysin immunolabeling: VRC group = 53%, VRC + hMSC group = 64%; p < 0.05). Overall, the present data show that intravenous injection of hMSC has neuroprotective and anti-inflammatory effects, decreasing reactive astrogliosis, and microglial reaction. Also, such cell therapy results in motoneuron preservation, combined with significant maintenance of spinal cord circuits, in particular those related to the ventral horn.


Assuntos
Gliose , Células-Tronco Mesenquimais , Animais , Gliose/terapia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neuroproteção , Medula Espinal , Raízes Nervosas Espinhais/lesões , Raízes Nervosas Espinhais/patologia
5.
Brain Res Bull ; 175: 196-204, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34339780

RESUMO

Reactive gliosis is a key feature and an important pathophysiological mechanism underlying chronic neurodegeneration following traumatic brain injury (TBI). In this study, we have explored the effects of intramuscular IGF-1 gene therapy on reactive gliosis and functional outcome after an injury of the cerebral cortex. Young adult male rats were intramuscularly injected with a recombinant adenoviral construct harboring the cDNA of human IGF-1 (RAd-IGF1), with a control vector expressing green fluorescent protein (RAd-GFP) or PBS as control. Three weeks after the intramuscular injections of adenoviral vectors, animals were subjected to a unilateral penetrating brain injury. The data revealed that RAd-IGF1 gene therapy significantly increased serum IGF1 levels and improved working memory performance after one week of TBI as compared to PBS or RAd-GFP lesioned animals. At the same time, when we analyzed the effects of therapy on glial scar formation, the treatment with RAd-IGF1 did not modify the number of glial fibrillary acidic protein (GFAP) positive cells, but we observed a decrease in vimentin immunoreactive astrocytes at 7 days post-lesion in the injured hemisphere compared to RAd-GFP group. Moreover, IGF-1 gene therapy reduced the number of Iba1+ cells with reactive phenotype and the number of MHCII + cells in the injured hemisphere. These results suggest that intramuscular IGF-1 gene therapy may represent a new approach to prevent traumatic brain injury outcomes in rats.


Assuntos
Lesões Encefálicas Traumáticas/terapia , Terapia Genética/métodos , Gliose/genética , Gliose/terapia , Fator de Crescimento Insulin-Like I/genética , Microglia , Animais , Lesões Encefálicas Traumáticas/psicologia , Proteínas de Ligação ao Cálcio/metabolismo , Vetores Genéticos/administração & dosagem , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Injeções Intramusculares , Masculino , Memória de Curto Prazo , Proteínas dos Microfilamentos/metabolismo , Neuroglia/imunologia , Neuroproteção , Desempenho Psicomotor , Ratos , Resultado do Tratamento , Vimentina/metabolismo
6.
Brain Res ; 1766: 147535, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34043998

RESUMO

We showed previously that voluntary long-term running improved cognition and motor skills, but in an age-dependent manner, in the Ts65Dn mouse model for Down syndrome (DS). Presently, we investigated the effect of running on the levels of some key proteins of the excitatory/inhibitory system, which is impaired in the trisomic brain, and on astroglia, a vital component of this system. Ts65Dn mice had free access to a running wheel for 9-13 months either from weaning or from the age of 7 months. Sedentary Ts65Dn mice served as controls. We found that running modified the levels of four of the seven proteins we tested that are associated with the glutamatergic/GABA-ergic system. Thus, Ts65Dn runners demonstrated increased levels of glutamine synthetase and metabotropic glutamate receptor 1 and decreased levels of glutamate transporter 1 and glutamic acid decarboxylase 65 (GAD65) versus sedentary mice, but of metabotropic glutamate receptor 1 and GAD65 only in the post-weaning cohort. GAD67, ionotropic N-methyl-D-aspartate type receptor subunit 1, and GABAAα5 receptors' levels were similar in runners and sedentary animals. The number of glial fibrillary acidic protein (GFAP)-positive astrocytes and the levels of GFAP were significantly reduced in runners relative to sedentary mice. Our study provides new insight into the mechanisms underlying the beneficial effect of voluntary, sustained running on function of the trisomic brain by identifying the involvement of proteins associated with glutamatergic and GABAergic systems and reduction in reactive astrogliosis.


Assuntos
Encéfalo/metabolismo , Síndrome de Down/metabolismo , Gliose/metabolismo , Gliose/terapia , Condicionamento Físico Animal/fisiologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Síndrome de Down/genética , Síndrome de Down/patologia , Transportador 2 de Aminoácido Excitatório/metabolismo , Feminino , Gliose/patologia , Glutamato Descarboxilase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Condicionamento Físico Animal/tendências , Receptores de GABA/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Fatores de Tempo
7.
J Mol Endocrinol ; 66(4): 259-272, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33729996

RESUMO

Earlier, it was shown that reversing the downregulation of neuritin expression in the brain improves central neuropathy in diabetic rats. We investigated the protective mechanism of neuritin in diabetic cognitive dysfunction via astrocytes. Further, the impact of the overexpression of neuritin in the cortex and the hippocampus on diabetic cognitive dysfunction and astrogliosis in type 2 diabetic (db/db) mice was assessed. Antagonists were used to inhibit the JAK2/STAT3 signaling pathway in U-118MG, an astrocyte cell line. Immunofluorescence, Western blotting, and real-time PCR were performed. Neuritin overexpression in the hippocampus of db/db mice significantly ameliorated cognitive dysfunction, hippocampal neuronal impairment, and synaptic plasticity deterioration, and inhibited astrogliosis and the JAK2/STAT3 signaling pathway in the hippocampus. Neuritin suppressed the JAK2/STAT3 signaling pathway to inhibit lipopolysaccharide-induced gliosis in U-118MG cells. It was observed that neuritin regulates the JAK2/STAT3 signaling pathway in astrocytes to inhibit astrogliosis and improve diabetic cognitive dysfunction.


Assuntos
Disfunção Cognitiva/genética , Gliose/genética , Janus Quinase 2/genética , Neuropeptídeos/genética , Fator de Transcrição STAT3/genética , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Córtex Cerebelar/metabolismo , Córtex Cerebelar/patologia , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Complicações do Diabetes/complicações , Complicações do Diabetes/genética , Complicações do Diabetes/patologia , Complicações do Diabetes/terapia , Neuropatias Diabéticas/genética , Neuropatias Diabéticas/patologia , Neuropatias Diabéticas/terapia , Modelos Animais de Doenças , Proteínas Ligadas por GPI/genética , Regulação da Expressão Gênica/genética , Gliose/induzido quimicamente , Gliose/patologia , Gliose/terapia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Lipopolissacarídeos/toxicidade , Camundongos , Plasticidade Neuronal/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ratos
8.
Cerebellum ; 20(3): 420-429, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33394333

RESUMO

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an abnormal expansion of CAG repeats in the Ataxin1 (ATXN1) gene. SCA1 is characterized by motor deficits, cerebellar neurodegeneration, and gliosis and gene expression changes. Expression of brain-derived neurotrophic factor (BDNF), growth factor important for the survival and function of cerebellar neurons, is decreased in ATXN1[82Q] mice, the Purkinje neuron specific transgenic mouse model of SCA1. As this decrease in BDNF expression may contribute to cerebellar neurodegeneration, we tested whether delivery of extrinsic human BDNF via osmotic ALZET pumps has a beneficial effect on disease severity in this mouse model of SCA1. Additionally, to test the effects of BDNF on established and progressing cerebellar pathogenesis and motor deficits, we delivered BDNF post-symptomatically. We have found that post-symptomatic delivery of extrinsic BDNF ameliorated motor deficits and cerebellar pathology (i.e., dendritic atrophy of Purkinje cells, and astrogliosis) indicating therapeutic potential of BDNF even after the onset of symptoms in SCA1. However, BDNF did not alter Purkinje cell gene expression changes indicating that certain aspects of disease pathogenesis cannot be ameliorated/slowed down with BDNF and that combinational therapies may be needed.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Ataxias Espinocerebelares/terapia , Animais , Cerebelo/patologia , Dendritos/patologia , Feminino , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Gliose/patologia , Gliose/terapia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Células de Purkinje/patologia , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia
9.
Int J Neurosci ; 131(6): 571-579, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32241216

RESUMO

PURPOSE: Clinical research suggests that transcranial direct current stimulation (tDCS) at bilateral supraorbital foramen and inferior orbital rim and nose intersections may facilitate rehabilitation after stroke. However, the underlying neurobiological mechanisms of tDCS remain poorly understood, impeding its clinical application. Here, we investigated the effect of tDCS applied after stroke on neural cells. MATERIALS AND METHODS: Middle cerebral arterial occlusion (MCAO) reperfusion was induced in rats. Animals with comparable infarcts were randomly divided into MCAO group and MCAO + tDCS group. Recovery of neurological function was assessed behaviorally by modified neurological severity score (mNSS). Ischemic tissue damage verified histologically by TTC and HE staining. Immunohistochemical staining, real-time qPCR, and western blot were applied to determine the changes of neural cells in ischemic brains. RESULTS: The results reveal that tDCS treated by multilead brain reflex instrument can promote the recovery of neurological function, remarkably reduce cerebral infarct volume, promote brain tissue rehabilitation, and can effectively inhibit astrocytosis and enhance neuronal survival and synaptic function in ischemic brains. CONCULSIONS: Our study suggests that tDCS treated by multilead brain reflex instrument could be prospectively developed into a clinical treatment modality.


Assuntos
Gliose/terapia , Infarto da Artéria Cerebral Média/reabilitação , AVC Isquêmico/reabilitação , Neurônios , Recuperação de Função Fisiológica , Reabilitação do Acidente Vascular Cerebral , Estimulação Transcraniana por Corrente Contínua , Animais , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/fisiopatologia , AVC Isquêmico/etiologia , AVC Isquêmico/patologia , AVC Isquêmico/fisiopatologia , Masculino , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , Índice de Gravidade de Doença
10.
Stem Cells Transl Med ; 10(3): 427-440, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33103374

RESUMO

There is increasing evidence that administration of many types of stem cells, including human amnion epithelial cells (hAECs), can reduce hypoxic-ischemic injury, including in the perinatal brain. However, the therapeutic window for single dose treatment is not known. We compared the effects of early and delayed intracerebroventricular administration of hAECs in fetal sheep at 0.7 gestation on brain injury induced by 25 minutes of complete umbilical cord occlusion (UCO) or sham occlusion. Fetuses received either 1 × 106 hAECs or vehicle alone, as an infusion over 1 hour, either 2 or 24 hours after UCO. Fetuses were killed for brain histology at 7 days post-UCO. hAEC infusion at both 2 and 24 hours had dramatic anti-inflammatory and anti-gliotic effects, including significantly attenuating the increase in microglia after UCO in the white and gray matter and the number of astrocytes in the white matter. Both protocols partially improved myelination, but had no effect on total or immature/mature numbers of oligodendrocytes. Neuronal survival in the hippocampus was increased by hAEC infusion at either 2 or 24 hours, whereas only hAECs at 24 hours were associated with improved neuronal survival in the striatum and thalamus. Neither protocol improved recovery of electroencephalographic (EEG) power. These data suggest that a single infusion of hAECs is anti-inflammatory, anti-gliotic, and neuroprotective in preterm fetal sheep when given up to 24 hours after hypoxia-ischemia, but was associated with limited white matter protection after 7 days recovery and no improvement in the recovery of EEG power.


Assuntos
Âmnio , Gliose , Hipóxia Encefálica/terapia , Transplante de Células-Tronco , Âmnio/citologia , Animais , Células Epiteliais/transplante , Feminino , Feto , Gliose/terapia , Humanos , Gravidez , Ovinos , Células-Tronco , Cordão Umbilical
11.
J Neurotrauma ; 38(6): 777-788, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33107383

RESUMO

Chronic spinal cord injury (SCI) is a devastating medical condition. In the acute phase after injury, there is cell loss resulting in chronic axonal damage and loss of sensory and motor function including loss of oligodendrocytes that results in demyelination of axons and further dysfunction. In the chronic phase, the inhibitory environment within the lesion including the glial scar can arrest axonal growth and regeneration and can also potentially affect transplanted cells. We hypothesized that glial scar ablation (GSA) along with cell transplantation may be required as a combinatorial therapy to achieve functional recovery, and therefore we proposed to examine the survival and fate of human induced pluripotent stem cell (iPSC) derived pre-oligodendrocyte progenitor cells (pre-OPCs) transplanted in a model of chronic SCI, whether this was affected by GSA, and whether this combination of treatments would result in functional recovery. In this study, chronically injured athymic nude (ATN) rats were allocated to one of three treatment groups: GSA only, pre-OPCs only, or GSA+pre-OPCs. We found that human iPSC derived pre-OPCs were multi-potent and retained the ability to differentiate into mainly oligodendrocytes or neurons when transplanted into the chronically injured spinal cords of rats. Twelve weeks after cell transplantation, we observed that more of the transplanted cells differentiated into oligodendrocytes when the glial scar was ablated compared with no GSA. Further, we also observed that a higher percentage of transplanted cells differentiated into V2a interneurons and motor neurons in the pre-OPCs only group when compared with GSA+pre-OPCs. This suggests that the local environment created by ablation of the glial scar may have a significant effect on the fate of cells transplanted into the injury site.


Assuntos
Gliose/terapia , Neurônios Motores/fisiologia , Células Precursoras de Oligodendrócitos/fisiologia , Oligodendroglia/fisiologia , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco/métodos , Animais , Células Cultivadas , Feminino , Corantes Fluorescentes/administração & dosagem , Gliose/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/química , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Pluripotentes Induzidas/transplante , Neurônios Motores/química , Células Precursoras de Oligodendrócitos/química , Células Precursoras de Oligodendrócitos/transplante , Oligodendroglia/química , Ratos , Rosa Bengala/administração & dosagem , Traumatismos da Medula Espinal/patologia , Vértebras Torácicas/lesões
13.
Int J Mol Sci ; 21(24)2020 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-33339379

RESUMO

A systemic inflammatory response induces multiple organ dysfunction and results in poor long-term neurological outcomes in neonatal sepsis. However, there is no effective therapy for treating or preventing neonatal sepsis besides antibiotics and supportive care. Therefore, a novel strategy to improve neonatal sepsis-related morbidity and mortality is desirable. Recently, we reported that prophylactic therapy with human amniotic stem cells (hAFSCs) improved survival in a rat model of lipopolysaccharide (LPS)-induced neonatal sepsis through immunomodulation. Besides improving the mortality, increasing survival without major morbidities is an important goal of neonatal intensive care for neonatal sepsis. This study investigated long-term neurological outcomes in neonatal sepsis survivors treated with hAFSCs using the LPS-induced neonatal sepsis model in rats. We found that prophylactic therapy with hAFSCs improved spatial awareness and memory-based behavior in neonatal sepsis survivors at adolescence in rats. The treatment suppressed acute reactive gliosis and subsequently reduced astrogliosis in the hippocampal region over a long period of assessment. To the best of our knowledge, this is the first report that proves the concept that hAFSC treatment improves cognitive impairment in neonatal sepsis survivors. We demonstrate the efficacy of hAFSC therapy in improving the mortality and morbidity associated with neonatal sepsis.


Assuntos
Disfunção Cognitiva/prevenção & controle , Gliose/prevenção & controle , Transplante de Células-Tronco Mesenquimais/métodos , Sepse Neonatal/complicações , Líquido Amniótico/citologia , Animais , Células Cultivadas , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/terapia , Gliose/etiologia , Gliose/terapia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Ratos , Ratos Sprague-Dawley
14.
Crit Care Med ; 48(4): e290-e298, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32205619

RESUMO

OBJECTIVES: Survivors of sepsis are frequently left with significant cognitive and behavioral impairments. These complications derive from nonresolving inflammation that persists following hospital discharge. To date, no study has investigated the effects of mesenchymal stromal cell therapy on the blood-brain barrier, astrocyte activation, neuroinflammation, and cognitive and behavioral alterations in experimental sepsis. DESIGN: Prospective, randomized, controlled experimental study. SETTING: Government-affiliated research laboratory. SUBJECTS: Male Swiss Webster mice (n = 309). INTERVENTIONS: Sepsis was induced by cecal ligation and puncture; sham-operated animals were used as control. All animals received volume resuscitation (1 mL saline/mouse subcutaneously) and antibiotics (meropenem 10 mg/kg intraperitoneally at 6, 24, and 48 hours). Six hours after surgery, mice were treated with mesenchymal stromal cells IV (1 × 10 cells in 0.05 mL of saline/mouse) or saline (0.05 mL IV). MEASUREMENTS AND MAIN RESULTS: At day 1, clinical score and plasma levels of inflammatory mediators were increased in cecal ligation and puncture mice. Mesenchymal stromal cells did not alter clinical score or survival rate, but reduced levels of systemic interleukin-1ß, interleukin-6, and monocyte chemoattractant protein-1. At day 15, survivor mice completed a battery of cognitive and behavioral tasks. Cecal ligation and puncture mice exhibited spatial and aversive memory deficits and anxiety-like behavior. These effects may be related to increased blood-brain barrier permeability, with altered tight-junction messenger RNA expression, increased brain levels of inflammatory mediators, and astrogliosis (induced at day 3). Mesenchymal stromal cells mitigated these cognitive and behavioral alterations, as well as reduced blood-brain barrier dysfunction, astrocyte activation, and interleukin-1ß, interleukin-6, tumor necrosis factor-α, and interleukin-10 levels in vivo. In cultured primary astrocytes stimulated with lipopolysaccharide, conditioned media from mesenchymal stromal cells reduced astrogliosis, interleukin-1ß, and monocyte chemoattractant protein-1, suggesting a paracrine mechanism of action. CONCLUSIONS: In mice who survived experimental sepsis, mesenchymal stromal cell therapy protected blood-brain barrier integrity, reduced astrogliosis and neuroinflammation, as well as improved cognition and behavior.


Assuntos
Barreira Hematoencefálica , Transtornos Cognitivos , Gliose , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Sepse , Animais , Masculino , Camundongos , Comportamento Animal , Barreira Hematoencefálica/metabolismo , Transtornos Cognitivos/prevenção & controle , Modelos Animais de Doenças , Gliose/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Estudos Prospectivos , Sepse/terapia
15.
Mol Ther ; 27(7): 1313-1326, 2019 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31043342

RESUMO

Mutations within over 250 known genes are associated with inherited retinal degeneration. Clinical success following gene-replacement therapy for congenital blindness due to RPE65 mutations establishes a platform for the development of downstream treatments targeting other forms of inherited ocular disease. Unfortunately, several challenges relevant to complex disease pathology and limitations of current gene-transfer technologies impede the development of related strategies for each specific form of inherited retinal degeneration. Here, we describe a gene-augmentation strategy that delays retinal degeneration by stimulating features of anabolic metabolism necessary for survival and structural maintenance of photoreceptors. We targeted two critical points of regulation in the canonical insulin/AKT/mammalian target of rapamycin (mTOR) pathway with AAV-mediated gene augmentation in a mouse model of retinitis pigmentosa. AAV vectors expressing the serine/threonine kinase, AKT3, promote dramatic preservation of photoreceptor numbers, structure, and partial visual function. This protective effect was associated with successful reprogramming of photoreceptor metabolism toward pathways associated with cell growth and survival. Collectively, these findings underscore the importance of AKT activity and downstream pathways associated with anabolic metabolism in photoreceptor survival and maintenance.


Assuntos
Terapia Genética/métodos , Neuroproteção/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Retinite Pigmentosa/terapia , Transdução de Sinais/genética , Transdução Genética , Animais , Sobrevivência Celular/genética , Células Cultivadas , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Modelos Animais de Doenças , Vetores Genéticos , Gliose/genética , Gliose/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação Puntual , Degeneração Retiniana/terapia , Retinite Pigmentosa/genética , Serina-Treonina Quinases TOR/metabolismo , Acuidade Visual/genética
16.
J Neurol Sci ; 402: 16-29, 2019 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-31100652

RESUMO

Spinal cord injury (SCI) is an incurable disorder with an unmet need of an effective treatment. Recently, autologous human bone marrow-derived stem cells have shown to promote functional improvement, due to their anti-inflammatory and regenerative/apocrine properties. In this study, the primary objective was to test whether a single intrathecal injection with a 100 µL suspension of 400,000 fresh human bone marrow-derived CD34+ and an equal number of CD105+ stem cells (Neuro-Cells (NC)), one day after balloon-compression of the spinal cord, improves motor function and reduces secondary damage in immunodeficient rats. During the first 5 weeks after this intervention, NC significantly improved locomotor recovery and induced less injury-associated adverse events compared to vehicle-treated rats. Histological analysis showed that NC reduced astrogliosis, and apoptosis early after administration (day 4), but not at a later stage (day 56) after SCI. Proteomic studies (at day 56) pointed to the release of paracrine factors and identified proteins involved in regenerative processes. As stem cells seem to reach their effects in acute lesions by mainly suppressing (secondary) inflammation, it is thus realistic to expect a lower magnitude of their eventual beneficial effect in T-cell deficient rats, a fact reinforcing the robustness of Neuro-Cells efficacy. Taken together, this study indicates that an intrathecal instillation of Neuro-Cells holds great promise as a neuro-regenerative intervention in a clinical setting with acute SCI patients.


Assuntos
Apoptose/fisiologia , Transplante de Medula Óssea/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia , Animais , Gliose/complicações , Gliose/terapia , Humanos , Interleucina-1beta/sangue , Interleucina-6/sangue , Locomoção/fisiologia , Masculino , Ratos , Ratos Nus , Traumatismos da Medula Espinal/sangue , Traumatismos da Medula Espinal/complicações , Fatores de Tempo , Fator de Necrose Tumoral alfa/sangue
17.
Methods Mol Biol ; 1940: 181-191, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30788826

RESUMO

Retinal explant culture systems have the potential to mimic the functional dynamics of the organ beyond those of the dissociated cells, thus making this technique a very powerful intermediate model system between in vitro cell cultures and in vivo animal models. The different retinal layers made of highly specialized cell types remain intact, while glia cell reactions and/or intercellular interactions can be evaluated under well-defined conditions in the lab.In retinal disorders neurodegeneration of mature retinal cells takes place. Therefore, we investigated the adult murine neuroretina in organ culture to test its suitability for use in preclinical therapeutic applications. Here we describe a method for the organ culture of adult murine retina (>12 weeks) used to establish survival, cellular changes and early degeneration patterns of neuronal and glial cells. After enucleation of the eyeball and careful dissection of the retina from the sclera and retinal pigment epithelium, the detached retina is cultured with photoreceptor facing down on a supporting track-etched polycarbonate membrane in a 6-well culture plate maintained in a humidified atmosphere of 5% CO2 and 95% air at 37 °C. After 1, 2, 3, 4, 6, 8, or 10 days retinal explants can be harvested and immediately processed for RNA isolation or fixed in paraformaldehyde for histological analysis.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Técnicas de Cultura de Órgãos/métodos , Retina/crescimento & desenvolvimento , Retina/metabolismo , Doenças Retinianas/terapia , Animais , Técnicas de Cultura de Células , Células Cultivadas , Modelos Animais de Doenças , Gliose/terapia , Camundongos , Neuroglia/citologia , Neuroglia/patologia , Neurônios/citologia , Neurônios/patologia , Retina/transplante , Degeneração Retiniana/terapia , Descolamento Retiniano/terapia , Doenças Retinianas/patologia
18.
J Tissue Eng Regen Med ; 13(3): 509-521, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30726582

RESUMO

Spinal cord injuries (SCIs) result in the loss of sensory and motor function with massive cell death and axon degeneration. We have previously shown that transplantation of spinal cord-derived ependymal progenitor cells (epSPC) significantly improves functional recovery after acute and chronic SCI in experimental models, via neuronal differentiation and trophic glial cell support. Here, we propose an improved procedure based on transplantation of epSPC in a tubular conduit of hyaluronic acid containing poly (lactic acid) fibres creating a biohybrid scaffold. In vitro analysis showed that the poly (lactic acid) fibres included in the conduit induce a preferential neuronal fate of the epSPC rather than glial differentiation, favouring elongation of cellular processes. The safety and efficacy of the biohybrid implantation was evaluated in a complete SCI rat model. The conduits allowed efficient epSPC transfer into the spinal cord, improving the preservation of the neuronal tissue by increasing the presence of neuronal fibres at the injury site and by reducing cavities and cyst formation. The biohybrid-implanted animals presented diminished astrocytic reactivity surrounding the scar area, an increased number of preserved neuronal fibres with a horizontal directional pattern, and enhanced coexpression of the growth cone marker GAP43. The biohybrids offer an improved method for cell transplantation with potential capabilities for neuronal tissue regeneration, opening a promising avenue for cell therapies and SCI treatment.


Assuntos
Materiais Biocompatíveis/farmacologia , Traumatismos da Medula Espinal/terapia , Regeneração da Medula Espinal , Animais , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Epêndima/citologia , Feminino , Gliose/terapia , Ácido Hialurônico/farmacologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Poliésteres/farmacologia , Porosidade , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/patologia , Regeneração da Medula Espinal/efeitos dos fármacos
19.
CNS Neurosci Ther ; 25(1): 36-46, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29804326

RESUMO

INTRODUCTION: A possible target for stroke management is modulation of neuroinflammation. Evidence suggests that food components may exert anti-inflammatory properties and thus may reduce stroke-induced brain damage. AIM: To investigate the efficacy of a diet, containing anti-inflammatory ingredients, as treatment for focal ischemic brain damage induced by photothrombotic stroke in the somatosensory cortex of rats. RESULTS: Brain lesions were surrounded by strong astrogliosis on both day 7 and day 21 after stroke and were accompanied by a trend toward globally decreased glucose metabolism on day 7. The investigational diet applied 2 weeks before the ischemia did not affect astrocyte activation on day 7, but reduced it at day 21. The investigational diet applied immediately after the ischemia, increased astrocyte activation on day 7 and completely reversed this effect on day 21. Moreover, postischemic intervention increased glucose metabolism in somatosensory cortex ipsilateral to the lesion on day 7. CONCLUSION: This study reveals potentially beneficial effects of a diet containing elevated amounts of anti-inflammatory nutrients on the recovery from ischemic brain damage. Therefore, dietary intervention can be considered as an adjuvant therapy for recovery from this brain pathology.


Assuntos
Encéfalo/metabolismo , Inflamação/dietoterapia , Inflamação/metabolismo , Acidente Vascular Cerebral/dietoterapia , Acidente Vascular Cerebral/metabolismo , Animais , Animais não Endogâmicos , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/patologia , Isquemia Encefálica/dietoterapia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Gliose/dietoterapia , Gliose/metabolismo , Gliose/terapia , Glucose/metabolismo , Inflamação/terapia , Masculino , Atividade Motora , Distribuição Aleatória , Ratos Sprague-Dawley , Acidente Vascular Cerebral/patologia
20.
PLoS One ; 13(11): e0208105, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30485360

RESUMO

Cerebral palsy (CP) encompasses a group of non-progressive brain disorders that are often acquired through perinatal hypoxic-ischemic (HI) brain injury. Injury leads to a cascade of cell death events, resulting in lifetime motor and cognitive deficits. There are currently no treatments that can repair the resulting brain damage and improve functional outcomes. To date, preclinical research using neural precursor cell (NPC) transplantation as a therapy for HI brain injury has shown promise. To translate this treatment to the clinic, it is essential that human-derived NPCs also be tested in animal models, however, a major limitation is the high risk of xenograft rejection. A solution is to transplant the cells into immune-deficient rodents, but there are currently no models of HI brain injury established in such a cohort of animals. Here, we demonstrate that a model of HI brain injury can be generated in immune-deficient Prkdc knockout (KO) rats. Long-term deficits in sensorimotor function were similar between KO and wildtype (WT) rats. Interestingly, some aspects of the injury were more severe in KO rats. Additionally, human induced pluripotent stem cell derived (hiPSC)-NPCs had higher survival at 10 weeks post-transplant in KO rats when compared to their WT counterparts. This work establishes a reliable model of neonatal HI brain injury in Prkdc KO rats that will allow for future transplantation, survival, and long-term evaluation of the safety and efficacy of hiPSC-NPCs for neonatal brain damage. This model will enable critical preclinical translational research using human NPCs.


Assuntos
Modelos Animais de Doenças , Hipóxia-Isquemia Encefálica/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Células-Tronco Neurais/transplante , Animais , Animais Recém-Nascidos , Encéfalo/patologia , Sobrevivência Celular , Proteína Quinase Ativada por DNA/genética , Gliose/patologia , Gliose/terapia , Humanos , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Neurais/patologia , Proteínas Nucleares/genética , Distribuição Aleatória , Ratos Long-Evans , Ratos Transgênicos , Imunodeficiência Combinada Severa/genética , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...