Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Surgery ; 175(2): 242-249, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37661485

RESUMO

BACKGROUND: Crohn-related rectovaginal fistulas are notoriously difficult to treat. Studies of mesenchymal stem cells for the treatment of perianal Crohn fistulizing disease have largely excluded rectovaginal fistulas. The aim of this study was to determine the safety and efficacy of mesenchymal stem cells for refractory rectovaginal fistulizing Crohn disease. METHODS: A phase IB/IIA randomized control trial was performed in a 3:1, single-blinded study. Patients included were adult women with an anovaginal/rectovaginal fistula in the setting of Crohn disease. Seventy-five million mesenchymal stem cells were administered with a 22G needle after curettage and primary closure of the fistula tract at day 0 and month 3. Adverse and serious adverse events were recorded at post-procedure day 1, week 2, week 6, month 3, month 6, and month 12, along with clinical healing, magnetic resonance imaging, and patient-reported outcomes. RESULTS: A total of 19 patients were enrolled and treated-15 treatment and 4 control. There were no adverse or serious adverse events related to mesenchymal stem cell therapy. At 6 months, 50% of the treatment group and 0% of the control had complete clinical and radiographic healing; 91.7% of the treatment group had improvement at 6 months with only one patient having a lack of response, whereas only 50% of the control group had improvement at 6 months. CONCLUSION: Bone marrow-derived mesenchymal stem cells offer a safe alternative treatment approach for rectovaginal fistulas in the setting of Crohn disease. Complete healing was achieved in half of the patients.


Assuntos
Doença de Crohn , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Mesenquimais , Fístula Retal , Adulto , Humanos , Feminino , Doença de Crohn/complicações , Doença de Crohn/terapia , Fístula Retovaginal/etiologia , Fístula Retovaginal/cirurgia , Medula Óssea , Fístula Retal/etiologia , Fístula Retal/terapia , Resultado do Tratamento
2.
JCI Insight ; 7(21)2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36345941

RESUMO

HIV-specific chimeric antigen receptor-T cell (CAR T cell) therapies are candidates to functionally cure HIV infection in people with HIV (PWH) by eliminating reactivated HIV-infected cells derived from latently infected cells within the HIV reservoir. Paramount to translating such therapeutic candidates successfully into the clinic will require anti-HIV CAR T cells to localize to lymphoid tissues in the body and eliminate reactivated HIV-infected cells such as CD4+ T cells and monocytes/macrophages. Here we show that i.v. injected anti-HIV duoCAR T cells, generated using a clinical-grade anti-HIV duoCAR lentiviral vector, localized to the site of active HIV infection in the spleen of humanized mice and eliminated HIV-infected PBMCs. CyTOF analysis of preinfusion duoCAR T cells revealed an early memory phenotype composed predominantly of CCR7+ stem cell-like/central memory T cells (TSCM/TCM) with expression of some effector-like molecules. In addition, we show that anti-HIV duoCAR T cells effectively sense and kill HIV-infected CD4+ T cells and monocytes/macrophages. Furthermore, we demonstrate efficient genetic modification of T cells from PWH on suppressive ART into anti-HIV duoCAR T cells that subsequently kill autologous PBMCs superinfected with HIV. These studies support the safety and efficacy of anti-HIV duoCAR T cell therapy in our presently open phase I/IIa clinical trial (NCT04648046).


Assuntos
Infecções por HIV , HIV-1 , Receptores de Antígenos Quiméricos , Animais , Camundongos , Linfócitos T CD4-Positivos , Infecções por HIV/tratamento farmacológico , Leucócitos Mononucleares , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto
3.
Cancer Discov ; 12(8): 1886-1903, 2022 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-35554512

RESUMO

Chimeric antigen receptor T-cell (CAR-T cell) therapy directed at CD19 produces durable remissions in the treatment of relapsed/refractory non-Hodgkin lymphoma (NHL). Nonetheless, many patients receiving CD19 CAR-T cells fail to respond for unknown reasons. To reveal changes in 4-1BB-based CD19 CAR-T cells and identify biomarkers of response, we used single-cell RNA sequencing and protein surface marker profiling of patient CAR-T cells pre- and postinfusion into patients with NHL. At the transcriptional and protein levels, we note the evolution of CAR-T cells toward a nonproliferative, highly differentiated, and exhausted state, with an enriched exhaustion profile in CAR-T cells of patients with poor response marked by TIGIT expression. Utilizing in vitro and in vivo studies, we demonstrate that TIGIT blockade alone improves the antitumor function of CAR-T cells. Altogether, we provide evidence of CAR-T cell dysfunction marked by TIGIT expression driving a poor response in patients with NHL. SIGNIFICANCE: This is the first study investigating the mechanisms linked to CAR-T patient responses based on the sequential analysis of manufactured and infused CAR-T cells using single-cell RNA and protein expression data. Furthermore, our findings are the first to demonstrate an improvement of CAR-T cell efficacy with TIGIT inhibition alone. This article is highlighted in the In This Issue feature, p. 1825.


Assuntos
Linfoma não Hodgkin , Receptores de Antígenos Quiméricos , Receptores Imunológicos , Linfócitos T , Antígenos CD19 , Humanos , Imunoterapia Adotiva , Linfoma não Hodgkin/genética , Receptores de Antígenos de Linfócitos T , Receptores de Antígenos Quiméricos/genética , Receptores Imunológicos/genética , Linfócitos T/patologia
4.
Front Immunol ; 11: 1941, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849651

RESUMO

Chimeric antigen receptor T cells (CAR-T cell) targeting CD19 are effective against several subtypes of CD19-expressing hematologic malignancies. Centralized manufacturing has allowed rapid expansion of this cellular therapy, but it may be associated with treatment delays due to the required logistics. We hypothesized that point of care manufacturing of CAR-T cells on the automated CliniMACS Prodigy® device allows reproducible and fast delivery of cells for the treatment of patients with non-Hodgkin lymphoma. Here we describe cell manufacturing results and characterize the phenotype and effector function of CAR-T cells used in a phase I/II study. We utilized a lentiviral vector delivering a second-generation CD19 CAR construct with 4-1BB costimulatory domain and TNFRSF19 transmembrane domain. Our data highlight the successful generation of CAR-T cells at numbers sufficient for all patients treated, a shortened duration of production from 12 to 8 days followed by fresh infusion into patients, and the detection of CAR-T cells in patient circulation up to 1-year post-infusion.


Assuntos
Antígenos CD19/imunologia , Engenharia Celular , Imunoterapia Adotiva , Linfoma não Hodgkin/terapia , Sistemas Automatizados de Assistência Junto ao Leito , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/transplante , Animais , Antígenos CD19/genética , Antígenos CD19/metabolismo , Automação , Técnicas de Cultura de Células , Células Cultivadas , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Citotoxicidade Imunológica , Humanos , Linfoma não Hodgkin/imunologia , Linfoma não Hodgkin/metabolismo , Camundongos Endogâmicos NOD , Fenótipo , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transplante Autólogo , Resultado do Tratamento , Carga de Trabalho , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Stem Cells Transl Med ; 5(11): 1506-1514, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27400792

RESUMO

: Human mesenchymal stem cells (hMSCs) are being increasingly pursued as potential therapies for immune-mediated conditions, including multiple sclerosis. Although they can suppress human Th1 responses, they reportedly can reciprocally enhance human Th17 responses. Here, we investigated the mechanisms underlying the capacity of hMSCs to modulate human Th1 and Th17 responses. Human adult bone marrow-derived MSCs were isolated, and their purity and differentiation capacity were confirmed. Human venous peripheral blood mononuclear cells (PBMC) were activated, alone, together with hMSC, or in the presence of hMSC-derived supernatants (sups). Cytokine expression by CD4+ T-cell subsets (intracellular staining by fluorescence-activated cell sorting) and secreted cytokines (enzyme-linked immunosorbent assay) were then quantified. The contribution of prostaglandin E2 (PGE2) as well as of myeloid cells to the hMSC-mediated regulation of T-cell responses was investigated by selective depletion of PGE2 from the hMSC sups (anti-PGE2 beads) and by the selective removal of CD14+ cells from the PBMC (magnetic-activated cell sorting separation). Human MSC-secreted products could reciprocally induce interleukin-17 expression while decreasing interferon-γ expression by human CD4+ T cells, both in coculture and through soluble products. Pre-exposure of hMSCs to IL-1ß accentuated their capacity to reciprocally regulate Th1 and Th17 responses. Human MSCs secreted high levels of PGE2, which correlated with their capacity to regulate the T-cell responses. Selective removal of PGE2 from the hMSC supernatants abrogated the impact of hMSC on the T cells. Selective removal of CD14+ cells from the PBMCs also limited the capacity of hMSC-secreted PGE2 to affect T-cell responses. Our discovery of a novel PGE2-dependent and myeloid cell-mediated mechanism by which human MSCs can reciprocally induce human Th17 while suppressing Th1 responses has implications for the use of, as well as monitoring of, MSCs as a potential therapeutic for patients with multiple sclerosis and other immune-mediated diseases. SIGNIFICANCE: Although animal studies have generated a growing interest in the anti-inflammatory potential of mesenchymal stem cells (MSCs) for the treatment of autoimmune diseases, MSCs possess the capacity to both limit and promote immune responses. Yet relatively little is known about human-MSC modulation of human disease-implicated T-cell responses, or the mechanisms underlying such modulation. The current study reveals a novel prostaglandin E2-dependent and myeloid cell-mediated mechanism by which human MSCs can reciprocally regulate human Th17 and Th1 responses, with implications for the use of MSCs as a potential therapeutic for patients with multiple sclerosis and other immune-mediated diseases.

6.
Virus Res ; 196: 170-80, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25479595

RESUMO

The DNA repair gene O(6)-methylguanine-DNA methyltransferase (MGMT) allows efficient in vivo enrichment of transduced hematopoietic stem cells (HSC). Thus, linking this selection strategy to therapeutic gene expression offers the potential to reconstitute diseased hematopoietic tissue with gene-corrected cells. However, different dual-gene expression vector strategies are limited by poor expression of one or both transgenes. To evaluate different co-expression strategies in the context of MGMT-mediated HSC enrichment, we compared selection and expression efficacies in cells cotransduced with separate single-gene MGMT and GFP lentivectors to those obtained with dual-gene vectors employing either encephalomyocarditis virus (EMCV) internal ribosome entry site (IRES) or foot and mouth disease virus (FMDV) 2A elements for co-expression strategies. Each strategy was evaluated in vitro and in vivo using equivalent multiplicities of infection (MOI) to transduce 5-fluorouracil (5-FU) or Lin(-)Sca-1(+)c-kit(+) (LSK)-enriched murine bone marrow cells (BMCs). The highest dual-gene expression (MGMT(+)GFP(+)) percentages were obtained with the FMDV-2A dual-gene vector, but half of the resulting gene products existed as fusion proteins. Following selection, dual-gene expression percentages in single-gene vector cotransduced and dual-gene vector transduced populations were similar. Equivalent MGMT expression levels were obtained with each strategy, but GFP expression levels derived from the IRES dual-gene vector were significantly lower. In mice, vector-insertion averages were similar among cells enriched after dual-gene vectors and those cotransduced with single-gene vectors. These data demonstrate the limitations and advantages of each strategy in the context of MGMT-mediated selection, and may provide insights into vector design with respect to a particular therapeutic gene or hematologic defect.


Assuntos
Expressão Gênica , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Lentivirus/genética , O(6)-Metilguanina-DNA Metiltransferase/genética , Animais , Fluoruracila/farmacologia , Dosagem de Genes , Ordem dos Genes , Técnicas de Transferência de Genes , Genes Reporter , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Células K562 , Camundongos , Transdução Genética
7.
ISRN Hematol ; 2012: 212586, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22888445

RESUMO

The P140K point mutant of MGMT allows robust hematopoietic stem cell (HSC) enrichment in vivo. Thus, dual-gene vectors that couple MGMT and therapeutic gene expression have allowed enrichment of gene-corrected HSCs in animal models. However, expression levels from dual-gene vectors are often reduced for one or both genes. Further, it may be desirable to express selection and therapeutic genes at distinct stages of cell differentiation. In this regard, we evaluated whether hematopoietic cells could be efficiently cotransduced using low MOIs of two separate single-gene lentiviruses, including MGMT for dual-positive cell enrichment. Cotransduction efficiencies were evaluated using a range of MGMT : GFP virus ratios, MOIs, and selection stringencies in vitro. Cotransduction was optimal when equal proportions of each virus were used, but low MGMT : GFP virus ratios resulted in the highest proportion of dual-positive cells after selection. This strategy was then evaluated in murine models for in vivo selection of HSCs cotransduced with a ubiquitous MGMT expression vector and an erythroid-specific GFP vector. Although the MGMT and GFP expression percentages were variable among engrafted recipients, drug selection enriched MGMT-positive leukocyte and GFP-positive erythroid cell populations. These data demonstrate cotransduction as a mean to rapidly enrich and evaluate therapeutic lentivectors in vivo.

8.
Stem Cells ; 26(3): 675-81, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18192231

RESUMO

Previous studies have suggested that donor bone marrow-derived cells can differentiate into lung epithelial cells at low frequency. We investigated whether we could enrich the number of donor-derived hematopoietic cells that have type II pneumocyte characteristics by overexpression of the drug resistance gene methylguanine DNA methyltransferase (MGMT). MGMT encodes O(6)-alkylguanine DNA alkyltransferase (AGT), a drug resistance protein for DNA damage induced by N,N'-bis(2-chloroethyl)-N-nitrosourea (BCNU), and the mutant P140K MGMT confers resistance to BCNU and the AGT inactivator O(6)-benzylguanine (BG). For this study, we used two MGMT selection models: one in which donor cells had a strong selection advantage because the recipient lung lacked MGMT expression, and another in which drug resistance was conferred by gene transfer of P140K MGMT. In both models, we saw an increase in the total number of donor-derived cells in the lung after BCNU treatment. Analysis of single-cell suspensions from 28 mice showed donor-derived cells with characteristics of type II pneumocytes, determined by surfactant protein C (SP-C) expression. Furthermore, an increase in the percentage of donor-derived SP-C cells was noted after BCNU or BG and BCNU treatment. This study demonstrates that bone marrow cells expressing MGMT can engraft in the lung and convert into cells expressing the type II pneumocyte protein SP-C. Furthermore, these cells can be enriched in response to alkylating agent-mediated lung injury. These results suggest that expression of MGMT could enhance the capacity of bone marrow-derived cells to repopulate lung epithelium, and when used in combination with a gene of interest, MGMT could have therapeutic applications.


Assuntos
Células da Medula Óssea/citologia , Resistência a Medicamentos , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Pulmão/citologia , Pulmão/enzimologia , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Alquilantes/toxicidade , Animais , Células da Medula Óssea/efeitos dos fármacos , Carmustina/toxicidade , Fusão Celular , Galinhas , DNA/metabolismo , Resistência a Medicamentos/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Pulmão/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Proteína B Associada a Surfactante Pulmonar/genética , Proteína B Associada a Surfactante Pulmonar/metabolismo , Retroviridae
9.
J Clin Invest ; 112(10): 1561-70, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14617757

RESUMO

Infusion of transduced hematopoietic stem cells into nonmyeloablated hosts results in ineffective in vivo levels of transduced cells. To increase the proportion of transduced cells in vivo, selection based on P140K O6-methylguanine-DNA-methyltransferase (MGMT[P140K]) gene transduction and O6-benzylguanine/1,3-bis(2-chloroethyl)-1-nitrosourea (BG/BCNU) treatment has been devised. In this study, we transduced human NOD/SCID repopulating cells (SRCs) with MGMT(P140K) using a lentiviral vector and infused them into BG/BCNU-conditioned NOD/SCID mice before rounds of BG/BCNU treatment as a model for in vivo selection. Engraftment was not observed until the second round of BG/BCNU treatment, at which time human cells emerged to compose up to 20% of the bone marrow. Furthermore, 99% of human CFCs derived from NOD/SCID mice were positive for provirus as measured by PCR, compared with 35% before transplant and 11% in untreated irradiation-preconditioned mice, demonstrating selection. Bone marrow showed BG-resistant O6-alkylguanine-DNA-alkyltransferase (AGT) activity, and CFUs were stained intensely for AGT protein, indicating high transgene expression. Real-time PCR estimates of the number of proviral insertions in individual CFUs ranged from 3 to 22. Selection resulted in expansion of one or more SRC clones containing similar numbers of proviral copies per mouse. To our knowledge, these results provide the first evidence of potent in vivo selection of MGMT(P140K) lentivirus-transduced human SRCs following BG/BCNU treatment.


Assuntos
Técnicas de Transferência de Genes , Guanina/análogos & derivados , Transplante de Células-Tronco Hematopoéticas , Lentivirus/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/genética , Imunodeficiência Combinada Severa , Transdução Genética , Condicionamento Pré-Transplante , Animais , Antígenos CD34/metabolismo , Antineoplásicos/metabolismo , Carmustina/metabolismo , Vetores Genéticos , Guanina/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Humanos , Lentivirus/genética , Camundongos , Camundongos SCID , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Radiação , Transgenes
10.
J Hematother Stem Cell Res ; 12(4): 375-87, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12965075

RESUMO

Optimized hematopoietic gene therapy requires vectors with strong expression in the desired target cell population and the ability to select for the expressing transduced cells. In the context of drug resistance selection of repopulating hematopoietic stem cells in the mouse, we examined tissue expression after transduced marrow transplantation of the drug selection gene, G156A mutant O6-methylguanine-DNA methyltransferase (G156A MGMT). To gain more experience with the rigor of the impact of selection on tissue-specific gene expression, we also asked whether there are expression differences between three different onco-retroviral backbones--MPSV, SF, and MFG. MGMT expression was compared after O6-benzylguanine (BG) and 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU) drug selection in vivo. After mice were transplanted with cells transduced with MPSV, MFG, or SF retroviral vectors expressing G156A MGMT and drug treated, nearly complete replacement by transduced progenitors was observed in the marrow. Each backbone supported MGMT expression in all four hematopoietic lineages in vivo indicating that MGMT-mediated selection is indeed robust. Expression in marrow, spleen, and thymus was very similar between the vectors and differences were most likely due to differences in gene copy number per selected cell. In primary and secondary recipients, the highest expression was observed in MFG and this was the vector that transduced at the greatest proviral copy number per cell. These data indicate that strong selection pressure using the MGMT gene to protect primary and secondary repopulating murine stem cells from the toxicity of BCNU. Regardless of the vector backbone used, multiorgan expression was observed without evidence of gene silencing. These data help establish mutant, BG-resistant MGMT as a potent selection gene for stem cell selection in vivo.


Assuntos
Guanina/análogos & derivados , Células-Tronco Hematopoéticas/metabolismo , Mutação , O(6)-Metilguanina-DNA Metiltransferase/genética , Proteínas Oncogênicas de Retroviridae/genética , Transgenes , Animais , Antineoplásicos/farmacologia , Células da Medula Óssea/citologia , Linhagem Celular , Linhagem da Célula , Separação Celular , Clonagem Molecular , Relação Dose-Resposta a Droga , Citometria de Fluxo , Inativação Gênica , Vetores Genéticos , Genoma , Guanina/farmacologia , Humanos , Células K562 , Camundongos , Camundongos Endogâmicos C3H , Modelos Genéticos , O(6)-Metilguanina-DNA Metiltransferase/farmacologia , Reação em Cadeia da Polimerase , Células-Tronco/citologia , Fatores de Tempo
11.
Mol Ther ; 8(1): 42-50, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12842427

RESUMO

Gene transduction of hematopoietic progenitors capable of reconstituting both primary and secondary recipients is an important milestone in preclinical development of gene therapy. Myeloablation conditioning prior to infusion of transduced stem cells causes significant host morbidity. In contrast, drug-resistance gene transfer utilizes judicious in vivo selection of transduced stem cells over time, reaching only the level of transduction and expression required. The O(6)-benzylguanine (BG)-resistant mutant O(6)-methylguanine-DNA methyltransferase (MGMT) gene is a potent selection gene for transduced cells. Using two different mutant MGMTs, G156A and P140K, that vary in BG resistance by a factor of 1:20, we asked whether long-term repopulating and secondary mouse-repopulating cells could be transduced, transplanted, and selected for in the nonmyeloablated recipient and whether the mutant MGMT would continue to be expressed in secondary recipient repopulating cells. We found that under stringent drug-selection competition, cells expressing the more BG-resistant variant, P140K-MGMT, were enriched over G156A-MGMT-expressing progenitors. In addition, the MFG retroviral vector transmitted the mutant MGMT gene to long-term repopulating cells that, after selective enrichment in the nonmyeloablated primary recipient, repopulated secondary mice and continued to express the transgene. Thus, MFG mutant MGMT vectors transduce repopulating hematopoietic stem cells that may be used both for chemotherapeutic drug resistance and to enrich for second therapeutic genes.


Assuntos
Resistência a Medicamentos , Terapia Genética/métodos , Guanina/análogos & derivados , O(6)-Metilguanina-DNA Metiltransferase/genética , Animais , Antineoplásicos Alquilantes/farmacologia , Southern Blotting , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Carmustina/farmacologia , Separação Celular , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Técnicas de Transferência de Genes , Vetores Genéticos , Guanina/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C3H , Modelos Genéticos , Mutação , Transplante de Neoplasias , O(6)-Metilguanina-DNA Metiltransferase/biossíntese , Reação em Cadeia da Polimerase , Células-Tronco/metabolismo , Fatores de Tempo
12.
Blood ; 102(5): 1626-33, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12730104

RESUMO

Mismatch repair deficiency is associated with carcinogenesis, increased spontaneous and induced mutagenesis, and resistance to methylating agents. In humans, leukemias and lymphomas arise in the background of mismatch repair deficiency, raising the possibility that hematopoiesis is abnormal as well. To address hematopoiesis in MSH2-/- mice, we collected marrow and performed serial transplantations of these cells, alone or mixed with wild-type cells, into lethally irradiated healthy mice. Transplant recipients were observed or treated with the methylating agent, temozolomide (TMZ). Methylating agent tolerance was evident by the competitive survival advantage of MSH2-/- marrow progenitors compared with wild-type cells after each TMZ exposure. However, serial repopulation by MSH2-/- cells was deficient compared with wild-type cells. In recipients of mixed populations, the MSH 2-/- cells were lost from the marrow, and mice receiving MSH2-/- cells plus TMZ could not be reconstituted in the third passage, whereas all wild-type cell recipients survived. No differences in telomere length, cell cycle distribution, or homing were observed, but an increase in microsatellite instability was seen in the MSH2-/- early progenitor colony-forming unit (CFU) and Sca+Kit+lin--derived clones. Thus, mismatch repair deficiency is associated with a hematopoietic repopulation defect and stem cell exhaustion because of accumulation of genomic instability.


Assuntos
Transplante de Medula Óssea , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA , Dacarbazina/análogos & derivados , Hematopoese/genética , Células-Tronco Hematopoéticas/fisiologia , Proteínas Proto-Oncogênicas/genética , Animais , Antineoplásicos Alquilantes/farmacologia , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dacarbazina/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Repetições de Microssatélites/genética , Proteína 2 Homóloga a MutS , Telômero/fisiologia , Temozolomida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...