Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Stem Cell Reports ; 2(5): 620-32, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24936450

RESUMO

The microenvironment is a critical mediator of stem cell survival, proliferation, migration, and differentiation. The majority of preclinical studies involving transplantation of neural stem cells (NSCs) into the CNS have focused on injured or degenerating microenvironments, leaving a dearth of information as to how NSCs differentially respond to intact versus damaged CNS. Furthermore, single, terminal histological endpoints predominate, providing limited insight into the spatiotemporal dynamics of NSC engraftment and migration. We investigated the early and long-term engraftment dynamics of human CNS stem cells propagated as neurospheres (hCNS-SCns) following transplantation into uninjured versus subacutely injured spinal cords of immunodeficient NOD-scid mice. We stereologically quantified engraftment, survival, proliferation, migration, and differentiation at 1, 7, 14, 28, and 98 days posttransplantation, and identified injury-dependent alterations. Notably, the injured microenvironment decreased hCNS-SCns survival, delayed and altered the location of proliferation, influenced both total and fate-specific migration, and promoted oligodendrocyte maturation.


Assuntos
Células-Tronco Neurais/transplante , Traumatismos da Medula Espinal/terapia , Animais , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Proliferação de Células , Humanos , Imageamento Tridimensional , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neurais/citologia , Oligodendroglia/citologia , Traumatismos da Medula Espinal/patologia , Fatores de Tempo , Transplante Heterólogo
2.
Stem Cells Transl Med ; 2(10): 731-44, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23981724

RESUMO

Clinical immunosuppression protocols use calcineurin inhibitors, such as cyclosporine A (CsA) or tacrolimus (FK506), or mammalian target of rapamycin (mTOR) inhibitors, such as sirolimus (rapamycin). These compounds alter immunophilin ligand signaling pathways, which are known to interact downstream with mediators for human neural stem cell (hNSC) differentiation and proliferation, suggesting that immunosuppressants may directly alter hNSC properties. We investigated whether immunosuppressants can exert direct effects on the differentiation, proliferation, survival, and migration of human central nervous system-derived stem cells propagated as neurospheres (hCNS-SCns) in vitro and in an in vivo model of spinal cord injury. We identified unique, immunosuppressant-dependent effects on hCNS-SCns differentiation and proliferation in vitro. All immunosuppressants tested increased neuronal differentiation, and CsA and rapamycin inhibited proliferation in vitro. No immunosuppressant-mediated effects on hCNS-SCns survival or migration in vitro were detected. These data suggested that immunosuppressant administration could alter hCNS-SCns properties in vivo. We tested this hypothesis by administering immunosuppressants to constitutively immunodeficient spinal cord injured mice and assessed survival, proliferation, differentiation, and migration of hCNS-SCns after 14 weeks. In parallel, we administered immunosuppressants to immunocompetent spinal cord injury (SCI) mice and also evaluated hCNS-SCns engraftment and fate. We identified no effect of immunosuppressants on the overall hCNS-SCns fate profile in either xenotransplantation model. Despite a lower level of human cell engraftment in immunocompetent SCI mice, functional locomotor recovery was observed in animals receiving hCNS-SCns transplantation with no evidence of allodynia. These data suggest that local cues in the microenvironment could exert a stronger influence on hCNS-SCns than circulating levels of immunosuppressants; however, differences between human and rodent metabolism/pharmokinetics and xenograft versus allograft paradigms could be determining factors.


Assuntos
Imunossupressores/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Recuperação de Função Fisiológica/efeitos dos fármacos , Transplante Heterólogo
3.
J Huntingtons Dis ; 1(1): 119-32, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-24086178

RESUMO

The Huntington's disease (HD) mutation leads to a complex process of Huntingtin (Htt) aggregation into multimeric species that eventually form visible inclusions in cytoplasm, nuclei and neuronal processes. One hypothesis is that smaller, soluble forms of amyloid proteins confer toxic effects and contribute to early cell dysfunction. However, analysis of mutant Htt aggregation intermediates to identify conformers that may represent toxic forms of the protein and represent potential drug targets remains difficult. We performed a detailed analysis of aggregation conformers in multiple in vitro, cell and ex vivo models of HD. Conformation-specific antibodies were used to identify and characterize aggregation species, allowing assessment of multiple conformers present during the aggregation process. Using a series of assays together with these antibodies, several forms could be identified. Fibrillar oligomers, defined as having a ß-sheet rich conformation, are observed in vitro using recombinant protein and in protein extracts from cells in culture or mouse brain and shown to be globular, soluble and non-sedimentable structures. Compounds previously described to modulate visible inclusion body formation and reduce toxicity in HD models were also tested and consistently found to alter the formation of fibrillar oligomers. Interestingly, these compounds did not alter the rate of visible inclusion formation, indicating that fibrillar oligomers are not necessarily the rate limiting step of inclusion body formation. Taken together, we provide insights into the structure and formation of mutant Htt fibrillar oligomers that are modulated by small molecules with protective potential in HD models.


Assuntos
Amiloide/química , Amiloide/genética , Doença de Huntington/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Animais , Química Encefálica , Linhagem Celular Tumoral , Humanos , Proteína Huntingtina , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Mutação/genética
4.
Regen Med ; 6(3): 367-406, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21548741

RESUMO

There is potential for a variety of stem cell populations to mediate repair in the diseased or injured CNS; in some cases, this theoretical possibility has already transitioned to clinical safety testing. However, careful consideration of preclinical animal models is essential to provide an appropriate assessment of stem cell safety and efficacy, as well as the basic biological mechanisms of stem cell action. This article examines the lessons learned from early tissue, organ and hematopoietic grafting, the early assumptions of the stem cell and CNS fields with regard to immunoprivilege, and the history of success in stem cell transplantation into the CNS. Finally, we discuss strategies in the selection of animal models to maximize the predictive validity of preclinical safety and efficacy studies.


Assuntos
Sistema Nervoso Central/patologia , Sobrevivência de Enxerto/imunologia , Medicina Regenerativa/métodos , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/imunologia , Animais , Sobrevivência Celular/imunologia , Modelos Animais de Doenças , Rejeição de Enxerto/imunologia , Hematopoese , Humanos , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/terapia , Células-Tronco/metabolismo , Transplante Heterólogo
5.
PLoS One ; 4(6): e5871, 2009 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-19517014

RESUMO

BACKGROUND: Human central nervous system-stem cells grown as neurospheres (hCNS-SCns) self-renew, are multipotent, and have potential therapeutic applications following trauma to the spinal cord. We have previously shown locomotor recovery in immunodeficient mice that received a moderate contusion spinal cord injury (SCI) and hCNS-SCns transplantation 9 days post-injury (dpi). Engrafted hCNS-SCns exhibited terminal differentiation to myelinating oligodendrocytes and synapse-forming neurons. Further, selective ablation of human cells using Diphtheria toxin (DT) abolished locomotor recovery in this paradigm, suggesting integration of human cells within the mouse host as a possible mechanism for the locomotor improvement. However, the hypothesis that hCNS-SCns could alter the host microenvironment as an additional or alternative mechanism of recovery remained unexplored; we tested that hypothesis in the present study. METHODS AND FINDINGS: Stereological quantification of human cells using a human-specific cytoplasmic marker demonstrated successful cell engraftment, survival, migration and limited proliferation in all hCNS-SCns transplanted animals. DT administration at 16 weeks post-transplant ablated 80.5% of hCNS-SCns. Stereological quantification for lesion volume, tissue sparing, descending serotonergic host fiber sprouting, chondroitin sulfate proteoglycan deposition, glial scarring, and angiogenesis demonstrated no evidence of host modification within the mouse spinal cord as a result of hCNS-SCns transplantation. Biochemical analyses supplemented stereological data supporting the absence of neural stem-cell mediated host repair. However, linear regression analysis of the number of engrafted hCNS-SCns vs. the number of errors on a horizontal ladder beam task revealed a strong correlation between these variables (r = -0.78, p<0.05), suggesting that survival and engraftment were directly related to a quantitative measure of recovery. CONCLUSIONS: Altogether, the data suggest that the locomotor improvements associated with hCNS-SCns transplantation were not due to modifications within the host microenvironment, supporting the hypothesis that human cell integration within the host circuitry mediates functional recovery following a 9 day delayed transplant.


Assuntos
Sistema Nervoso Central/patologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco , Animais , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Estudos de Coortes , Citoplasma/metabolismo , Toxina Diftérica/metabolismo , Feminino , Humanos , Camundongos , Camundongos SCID , Neurônios/metabolismo , Oligodendroglia/fisiologia , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA