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.
Cell Stem Cell ; 22(2): 177-190.e7, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29395054

RESUMO

The development of cell therapy for repairing damaged or diseased skeletal muscle has been hindered by the inability to significantly expand immature, transplantable myogenic stem cells (MuSCs) in culture. To overcome this limitation, a deeper understanding of the mechanisms regulating the transition between activated, proliferating MuSCs and differentiation-primed, poorly engrafting progenitors is needed. Here, we show that methyltransferase Setd7 facilitates such transition by regulating the nuclear accumulation of ß-catenin in proliferating MuSCs. Genetic or pharmacological inhibition of Setd7 promotes in vitro expansion of MuSCs and increases the yield of primary myogenic cell cultures. Upon transplantation, both mouse and human MuSCs expanded with a Setd7 small-molecule inhibitor are better able to repopulate the satellite cell niche, and treated mouse MuSCs show enhanced therapeutic potential in preclinical models of muscular dystrophy. Thus, Setd7 inhibition may help bypass a key obstacle in the translation of cell therapy for muscle disease.


Assuntos
Desenvolvimento Muscular , Proteínas Metiltransferases/antagonistas & inibidores , Transplante de Células-Tronco , Células-Tronco/citologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Autorrenovação Celular/efeitos dos fármacos , Células Cultivadas , Deleção de Genes , Histona-Lisina N-Metiltransferase , Camundongos , Músculo Esquelético/fisiologia , Proteína MyoD/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteínas Metiltransferases/metabolismo , Pirrolidinas/farmacologia , Regeneração/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Sulfonamidas/farmacologia , Tetra-Hidroisoquinolinas/farmacologia , beta Catenina/metabolismo
2.
Front Neurol ; 8: 75, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28337172

RESUMO

Bone marrow-derived cells (BMDCs) are capable of migrating across the blood-brain barrier (BBB) and accumulating in the central nervous system (CNS) when transplanted into recipients conditioned with whole-body irradiation or chemotherapy. We used the chemotherapeutic agents busulfan and treosulfan to condition recipient mice for transplantation with bone marrow (BM) cells isolated from donor mice ubiquitously expressing green fluorescent protein. We attempted to increase the accumulation of BMDCs in the CNS by mobilization of BMDCs using either, or both, granulocyte colony-stimulating factor (GCSF) or plerixafor (AMD3100). We also used several concentrations of busulfan. We hypothesized that higher concentrations of busulfan and BMDC mobilization would increase numbers of GFP+ cells in the CNS. The doses of busulfan employed (60-125 mg/kg) all resulted in high levels of sustained chimerism (>85% 1 year post-transplant) in both the blood and BM of wild-type (WT) mice and an amyotrophic lateral sclerosis (ALS) mouse model. Moreover, cells accumulated within the CNS in a dose-, time-, and disease-dependent manner. Conditioning with the hydrophilic busulfan analog treosulfan, which is unable to cross the BBB efficiently, also resulted in a high degree of BM chimerism. However, few GFP+ BMDCs were found within the CNS of WT or ALS mice of treosulfan-conditioned mice. Mobilization of BMDCs into the circulation using GCSF and/or AMD3100 did not lead to increased accumulation of GFP+ BMDCs within the CNS of WT or ALS mice. Weekly analysis of BMDC accumulation revealed that BMDCs accumulated more rapidly and to a greater extent in the CNS of ALS mice conditioned with a high dose (125 mg/kg) of busulfan compared to a lower dose (80 mg/kg). The number of GFP+ BMDCs in the CNS labeling with the proliferation marker Ki67 increased in parallel with BMDC accumulation within the CNS. Our results indicate that establishment of high levels of blood and BM chimerism alone is not sufficient to induce BMDC accumulation within the CNS and that CNS conditioning is a crucial requirement for BMDC accumulation to occur. Moreover, it appears that proliferation of BMDCs that infiltrate the CNS is partly responsible for cell accumulation in busulfan-conditioned ALS mice.

3.
J Vis Exp ; (98): e52553, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25867947

RESUMO

Bone marrow transplantation (BMT) is often used to replace the bone marrow (BM) compartment of recipient mice with BM cells expressing a distinct biomarker isolated from donor mice. This technique allows for identification of donor-derived hematopoietic cells within the recipient mice, and can be used to isolate and characterize donor cells using various biochemical techniques. BMT typically relies on myeloablative conditioning with total body irradiation to generate niche space within the BM compartment of recipient mice for donor cell engraftment. The protocol we describe here uses myelosuppressive conditioning with the chemotherapeutic agent busulfan. Unlike irradiation, which requires the use of specialized facilities, busulfan conditioning is performed using intraperitoneal injections of 20 mg/kg busulfan until a total dose of 60-100 mg/kg has been administered. Moreover, myeloablative irradiation can have toxic side effects and requires successful engraftment of donor cells for survival of recipient mice. In contrast, busulfan conditioning using these doses is generally well tolerated and mice survive without donor cell support. Donor BM cells are isolated from the femurs and tibiae of mice ubiquitously expressing green fluorescent protein (GFP), and injected into the lateral tail vein of conditioned recipient mice. BM chimerism is estimated by quantifying the number of GFP+ cells within the peripheral blood following BMT. Levels of chimerism >80% are typically observed in the peripheral blood 3-4 weeks post-transplant and remain established for at least 1 year. As with irradiation, conditioning with busulfan and BMT allows for the accumulation of donor BM-derived cells within the central nervous system (CNS), particularly in mouse models of neurodegeneration. This busulfan-mediated CNS accumulation may be more physiological than total body irradiation, as the busulfan treatment is less toxic and CNS inflammation appears to be less extensive. We hypothesize that these cells can be genetically engineered to deliver therapeutics to the CNS.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Transplante de Medula Óssea/métodos , Bussulfano/farmacologia , Agonistas Mieloablativos/farmacologia , Quimeras de Transplante , Condicionamento Pré-Transplante/métodos , Animais , Células da Medula Óssea/citologia , Sistema Nervoso Central/citologia , Camundongos , Camundongos Endogâmicos C57BL , Irradiação Corporal Total
4.
Neurosci Lett ; 588: 196-201, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25582787

RESUMO

Previous work has suggested that bone marrow (BM)-derived cells (BMDCs) accumulate within the CNS and could potentially associate with ß-amyloid plaques in Alzheimer's disease (AD). To explore the accumulation of BMDCs in murine AD, we transplanted green fluorescent protein (GFP)-labeled BM cells into triple transgenic (3×Tg) and wild-type (wt) mice using non-irradiative myelosuppresive conditioning with busulfan (BU). We find that BU (80mg/kg) is sufficient to obtain adequate chimerism (>85%) in wt mice. In order to obtain appreciable non-irradiative chimerism in the 3×Tg mice (>80%), anti-asialo ganglio-N-tetraosylceramide (α-ASGM-1) antibody was also used to reduce natural killer cell function and thereby abrogate the hybrid resistance of the 3×Tg mouse strain. Using BU conditioning and α-ASGM-1 together, we observed sustained BM chimerism and BMDC accumulation within the CNS of the 3×Tg and wt mice. In cortex and hippocampus, BMDC accumulation was perivascular in distribution and similar between 3×Tg and wt mice, with no clear association between BMDCs and AD plaques. We conclude that non-irradiative BM chimerism can be achieved with BU in 3×Tg mice, but requires α-ASGM-1 (or similar appropriate NK-cell depletion). Use of this chimerism protocol permits BMDCs accumulation in the CNS of mixed strain recipient mice although BMDCs appear to be largely perivascular within cortex and hippocampus.


Assuntos
Doença de Alzheimer/patologia , Células da Medula Óssea/efeitos dos fármacos , Bussulfano/farmacologia , Animais , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Encéfalo/patologia , Camundongos Transgênicos , Medula Espinal/patologia , Quimeras de Transplante , Condicionamento Pré-Transplante
5.
Acta Neuropathol ; 128(3): 363-80, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25107477

RESUMO

Microglia have long been the focus of much attention due to their strong proliferative response (microgliosis) to essentially any kind of damage to the CNS. More recently, we reached the realization that these cells play specific roles in determining progression and outcomes of essentially all CNS disease. Thus, microglia has ceased to be viewed as an accessory to underlying pathologies and has now taken center stage as a therapeutic target. Here, we review how our understanding of microglia's involvement in promoting or limiting the pathogenesis of diseases such as amyotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease, multiple sclerosis, X-linked adrenoleukodystrophy (X-ALD) and lysosomal storage diseases (LSD) has changed over time. While strategies to suppress the deleterious and promote the virtuous functions of microglia will undoubtedly be forthcoming, replacement of these cells has already proven its usefulness in a clinical setting. Over the past few years, we have reached the realization that microglia have a developmental origin that is distinct from that of bone marrow-derived myelomonocytic cells. Nevertheless, microglia can be replaced, in specific situations, by the progeny of hematopoietic stem cells (HSCs), pointing to a strategy to engineer the CNS environment through the transplantation of modified HSCs. Thus, microglia replacement has been successfully exploited to deliver therapeutics to the CNS in human diseases such as X-ALD and LSD. With this outlook in mind, we will discuss the evidence existing so far for microglial involvement in the pathogenesis and the therapy of specific CNS disease.


Assuntos
Doenças do Sistema Nervoso Central , Microglia/fisiologia , Doenças do Sistema Nervoso Central/patologia , Doenças do Sistema Nervoso Central/fisiopatologia , Doenças do Sistema Nervoso Central/terapia , Humanos
6.
PLoS One ; 8(4): e60661, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593276

RESUMO

Myeloablative preconditioning using irradiation is the most commonly used technique to generate rodents having chimeric bone marrow, employed for the study of bone marrow-derived cell accumulation in the healthy and diseased central nervous system. However, irradiation has been shown to alter the blood-brain barrier, potentially creating confounding artefacts. To better study the potential of bone marrow-derived cells to function as treatment vehicles for neurodegenerative diseases alternative preconditioning regimens must be developed. We treated transgenic mice that over-express human mutant superoxide dismutase 1, a model of amyotrophic lateral sclerosis, with busulfan to determine whether this commonly used chemotherapeutic leads to stable chimerism and promotes the entry of bone marrow-derived cells into spinal cord. Intraperitoneal treatment with busulfan at 60 mg/kg or 80 mg/kg followed by intravenous injection of green fluorescent protein-expressing bone marrow resulted in sustained levels of chimerism (~80%). Bone marrow-derived cells accumulated in the lumbar spinal cord of diseased mice at advanced stages of pathology at both doses, with limited numbers of bone marrow derived cells observed in the spinal cords of similarly treated, age-matched controls; the majority of bone marrow-derived cells in spinal cord immunolabelled for macrophage antigens. Comparatively, significantly greater numbers of bone marrow-derived cells were observed in lumbar spinal cord following irradiative myeloablation. These results demonstrate bone marrow-derived cell accumulation in diseased spinal cord is possible without irradiative preconditioning.


Assuntos
Esclerose Lateral Amiotrófica/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Transplante de Medula Óssea , Bussulfano/farmacologia , Medula Espinal/imunologia , Condicionamento Pré-Transplante/métodos , Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/cirurgia , Animais , Células da Medula Óssea/imunologia , Contagem de Células , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Fenótipo , Superóxido Dismutase/genética , Superóxido Dismutase-1
7.
Glia ; 57(13): 1410-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19243075

RESUMO

Amyotrophic lateral sclerosis (ALS) is associated with increased numbers of microglia within the CNS. However, it is unclear to what extent bone marrow (BM)-derived cells contribute to this microgliosis. We have studied the adoptive transfer of green fluorescent protein (GFP)-labeled whole BM cells and BM from mice that express GFP only in CX(3)CR1+ cells (CX(3)CR1(+/GFP)) into the CNS of a murine model of ALS having over-expression of mutant superoxide dismutase (mSOD), and wt littermates. We find that most GFP+ and CX(3)CR1(+/GFP) cells are found adjacent to the microvasculature within the CNS, both in mSOD and wt mice. GFP+ and CX(3)CR1(+/GFP) cells within the CNS have a variety of morphologies, including cells with an elongated appearance, weak Iba-1 immunoreactivity, and often mannose receptor immunoreactivity, indicating that these cells are perivascular microglia. Typically, less than 10% of BM-derived cells had a stellate-shape and expressed strong Iba-1 immunoreactivity, as expected for parenchymal microglia, indicating that BM-derived cells uncommonly generate parenchymal microglia. Adoptive transfer of BM-derived cells from CX(3)CR1(+/GFP) mice revealed that many elongated cells are GFP+, demonstrating that some perivascular cells are derived from BM cells of the CX(3)CR1+ lineage. The significantly greater numbers of BM cells in mSOD than in control mice indicate that the presence of these BM cells in the spinal cord is regulated by conditioning stimuli that may include irradiation and inflammatory factors within the CNS.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Vasos Sanguíneos/fisiopatologia , Células da Medula Óssea/fisiologia , Receptores de Quimiocinas/metabolismo , Medula Espinal/fisiopatologia , Esclerose Lateral Amiotrófica/patologia , Animais , Vasos Sanguíneos/citologia , Células da Medula Óssea/citologia , Receptor 1 de Quimiocina CX3C , Contagem de Células , Forma Celular , Quimera , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microvasos/fisiologia , Mutação de Sentido Incorreto , Medula Espinal/irrigação sanguínea , Medula Espinal/citologia , Superóxido Dismutase/genética , Superóxido Dismutase-1
8.
Glia ; 53(7): 744-53, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16518833

RESUMO

Amyotrophic lateral sclerosis (ALS) is associated with increased numbers of microglia within the central nervous system (CNS). However, it is unknown whether the microgliosis results from proliferation of CNS resident microglia, or recruitment of bone marrow (BM)-derived microglial precursors. Here we assess the distribution and number of BM-derived cells in spinal cord using transplantation of green fluorescent protein (GFP)-labeled BM cells into myelo-ablated mice over-expressing human mutant superoxide dismutase 1 (mSOD), a murine model of ALS. Transplantation of GFP+ BM did not affect the rate of disease progression in mSOD mice. Mean numbers of microglia and GFP+ cells in spinal cords of control mice were not significantly different from those in asymptomatic mSOD mice and showed no change with animal age. The number of GFP+ cells and microglia (F4/80+ and CD11b+ cells) within the spinal cord of mSOD mice increased compared to age-matched controls at a time when mSOD mice exhibited disease symptoms, continuing up to disease end-stage. Although we observed an increase in the number of GFP+ cells in spinal cords of mSOD mice with disease symptoms, mean numbers of GFP+ F4/80+ cells comprised less than 20% of all F4/80+ cells and did not increase with disease progression. Furthermore, the relative rates of proliferation in CD45+GFP- and CD45+GFP+ cells were comparable. Thus, we demonstrate that the microgliosis present in spinal cord tissue of mSOD mice is primarily due to an expansion of resident microglia and not to the recruitment of microglial precursors from the circulation.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Células da Medula Óssea/citologia , Linhagem da Célula/fisiologia , Gliose/fisiopatologia , Microglia/citologia , Medula Espinal/fisiopatologia , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Animais , Células da Medula Óssea/metabolismo , Contagem de Células , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Gliose/metabolismo , Gliose/patologia , Proteínas de Fluorescência Verde , Humanos , Antígenos Comuns de Leucócito/imunologia , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Superóxido Dismutase/genética , Superóxido Dismutase-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA