Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO Mol Med ; 15(9): e17748, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37538042

RESUMEN

Hematopoietic cell transplantation (HCT) treats many blood conditions but remains underused due to complications such as graft-versus-host disease (GvHD). In GvHD, donor immune cells attack the patient, requiring powerful immunosuppressive drugs like glucocorticoids (GCs) to prevent death. In this study, we tested the hypothesis that donor cell conditioning with the glucocorticoid fluticasone propionate (FLU) prior to transplantation could increase hematopoietic stem cell (HSC) engraftment and reduce GvHD. Murine HSCs treated with FLU had increased HSC engraftment and reduced severity and incidence of GvHD after transplantation into allogeneic hosts. While most T cells died upon FLU treatment, donor T cells repopulated in the hosts and appeared less inflammatory and alloreactive. Regulatory T cells (Tregs) are immunomodulatory and survived FLU treatment, resulting in an increased ratio of Tregs to conventional T cells. Our results implicate an important role for Tregs in maintaining allogeneic tolerance in FLU-treated grafts and suggest a therapeutic strategy of pre-treating donor cells (and not the patients directly) with GCs to simultaneously enhance engraftment and reduce GvHD upon allogeneic HCT.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Ratones , Animales , Fluticasona/farmacología , Fluticasona/uso terapéutico , Trasplante Homólogo/efectos adversos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Enfermedad Injerto contra Huésped/prevención & control , Inmunosupresores
2.
Sci Rep ; 12(1): 3951, 2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35273232

RESUMEN

The SARS-CoV-2 pandemic has brought to light the need for expedient diagnostic testing. Cost and availability of large-scale testing capacity has led to a lag in turnaround time and hindered contact tracing efforts, resulting in a further spread of SARS-CoV-2. To increase the speed and frequency of testing, we developed a cost-effective single-tube approach for collection, denaturation, and analysis of clinical samples. The approach utilizes 1 µL microbiological inoculation loops to collect saliva, sodium dodecyl sulfate (SDS) to inactivate and release viral genomic RNA, and a diagnostic reaction mix containing polysorbate 80 (Tween 80). In the same tube, the SDS-denatured clinical samples are introduced to the mixtures containing all components for nucleic acids detection and Tween 80 micelles to absorb the SDS and allow enzymatic reactions to proceed, obviating the need for further handling of the samples. The samples can be collected by the tested individuals, further decreasing the need for trained personnel to administer the test. We validated this single-tube sample-to-assay method with reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) and reverse transcription loop-mediated isothermal amplification (RT-LAMP) and discovered little-to-no difference between Tween- and SDS-containing reaction mixtures, compared to control reactions. This approach reduces the logistical burden of traditional large-scale testing and provides a method of deployable point-of-care diagnostics to increase testing frequency.


Asunto(s)
Prueba de Ácido Nucleico para COVID-19/métodos , SARS-CoV-2/genética , Saliva/virología , Prueba de Ácido Nucleico para COVID-19/instrumentación , Humanos , Técnicas de Diagnóstico Molecular , Técnicas de Amplificación de Ácido Nucleico , Reacción en Cadena en Tiempo Real de la Polimerasa , Manejo de Especímenes/instrumentación , Manejo de Especímenes/métodos
3.
Glia ; 70(5): 875-891, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35025109

RESUMEN

Microglia are the primary resident myeloid cells of the brain responsible for maintaining homeostasis and protecting the central nervous system (CNS) from damage and infection. Monocytes and monocyte-derived macrophages arising from the periphery have also been implicated in CNS pathologies, however, distinguishing between different myeloid cell populations in the CNS has been difficult. Here, we set out to develop a reliable histological marker that can assess distinct myeloid cell heterogeneity and functional contributions, particularly in the context of disease and/or neuroinflammation. scRNAseq from brains of mice infected with the neurotropic JHM strain of the mouse hepatitis virus (JHMV), a mouse coronavirus, revealed that Lgals3 is highly upregulated in monocyte and macrophage populations, but not in microglia. Subsequent immunostaining for galectin-3 (encoded by Lgals3), also referred to as MAC2, highlighted the high expression levels of MAC2 protein in infiltrating myeloid cells in JHMV-infected and bone marrow (BM) chimeric mice, in stark contrast to microglia, which expressed little to no staining in these models. Expression of MAC2 was found even 6-10 months following BM-derived cell infiltration into the CNS. We also demonstrate that MAC2 is not a specific label for plaque-associated microglia in the 5xFAD mouse model, but only appears in a distinct subset of these cells in the presence of JHMV infection or during aging. Our data suggest that MAC2 can serve as a reliable and long-lasting histological marker for monocyte/macrophages in the brain, identifying an accessible approach to distinguishing resident microglia from infiltrating cells in the CNS under certain conditions.


Asunto(s)
Trasplante de Médula Ósea , Infecciones por Coronavirus , Animales , Encéfalo/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo
4.
Front Cell Dev Biol ; 9: 734176, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34513848

RESUMEN

Hematopoietic stem cells (HSCs) are defined by their self-renewal, multipotency, and bone marrow (BM) engraftment abilities. How HSCs emerge during embryonic development remains unclear, but are thought to arise from hemogenic endothelium through an intermediate precursor called "pre-HSCs." Pre-HSCs have self-renewal and multipotent activity, but lack BM engraftability. They can be identified functionally by transplantation into neonatal recipients, or by in vitro co-culture with cytokines and stroma followed by transplantation into adult recipients. While pre-HSCs express markers such as Kit and CD144, a precise surface marker identity for pre-HSCs has remained elusive due to the fluctuating expression of common HSC markers during embryonic development. We have previously determined that the lack of CD11a expression distinguishes HSCs in adults as well as multipotent progenitors in the embryo. Here, we use a neonatal transplantation assay to identify pre-HSC populations in the mouse embryo. We establish CD11a as a critical marker for the identification and enrichment of pre-HSCs in day 10.5 and 11.5 mouse embryos. Our proposed pre-HSC population, termed "11a- eKLS" (CD11a- Ter119- CD43+ Kit+ Sca1+ CD144+), contains all in vivo long-term engrafting embryonic progenitors. This population also displays a cell-cycle status expected of embryonic HSC precursors. Furthermore, we identify the neonatal liver as the likely source of signals that can mature pre-HSCs into BM-engraftable HSCs.

5.
Elife ; 102021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34423781

RESUMEN

Microglia, the brain's resident myeloid cells, play central roles in brain defense, homeostasis, and disease. Using a prolonged colony-stimulating factor 1 receptor inhibitor (CSF1Ri) approach, we report an unprecedented level of microglial depletion and establish a model system that achieves an empty microglial niche in the adult brain. We identify a myeloid cell that migrates from the subventricular zone and associated white matter areas. Following CSF1Ri, these amoeboid cells migrate radially and tangentially in a dynamic wave filling the brain in a distinct pattern, to replace the microglial-depleted brain. These repopulating cells are enriched in disease-associated microglia genes and exhibit similar phenotypic and transcriptional profiles to white-matter-associated microglia. Our findings shed light on the overlapping and distinct functional complexity and diversity of myeloid cells of the CNS and provide new insight into repopulating microglia function and dynamics in the mouse brain.


Asunto(s)
Ventrículos Laterales/fisiología , Microglía/fisiología , Sustancia Blanca/fisiología , Animales , Encéfalo , Modelos Animales de Enfermedad , Homeostasis , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
7.
Nat Commun ; 10(1): 1634, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30967552

RESUMEN

Gene correction in human long-term hematopoietic stem cells (LT-HSCs) could be an effective therapy for monogenic diseases of the blood and immune system. Here we describe an approach for X-linked sSevere cCombined iImmunodeficiency (SCID-X1) using targeted integration of a cDNA into the endogenous start codon to functionally correct disease-causing mutations throughout the gene. Using a CRISPR-Cas9/AAV6 based strategy, we achieve up to 20% targeted integration frequencies in LT-HSCs. As measures of the lack of toxicity we observe no evidence of abnormal hematopoiesis following transplantation and no evidence of off-target mutations using a high-fidelity Cas9 as a ribonucleoprotein complex. We achieve high levels of targeting frequencies (median 45%) in CD34+ HSPCs from six SCID-X1 patients and demonstrate rescue of lymphopoietic defect in a patient derived HSPC population in vitro and in vivo. In sum, our study provides specificity, toxicity and efficacy data supportive of clinical development of genome editing to treat SCID-Xl.


Asunto(s)
ADN Complementario/genética , Edición Génica/métodos , Trasplante de Células Madre Hematopoyéticas , Subunidad gamma Común de Receptores de Interleucina/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Animales , Antígenos CD34/metabolismo , Sistemas CRISPR-Cas/genética , Línea Celular , Codón Iniciador/genética , Dependovirus , Exones/genética , Sangre Fetal/citología , Vectores Genéticos/genética , Voluntarios Sanos , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Mutación , Parvovirinae/genética , Cultivo Primario de Células , Factores de Tiempo , Transducción Genética/métodos , Quimera por Trasplante/genética , Trasplante Heterólogo/métodos , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética
8.
Nat Commun ; 10(1): 2021, 2019 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-31028274

RESUMEN

The original version of this Article omitted the following from the Acknowledgements: "G.B. acknowledges the support from the Cancer Prevention and Research Institute of Texas (RR140081 and RR170721)."This has now been corrected in both the PDF and HTML versions of the Article.

9.
Stem Cell Reports ; 12(3): 451-460, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30745035

RESUMEN

In the CNS, perivascular cells ("pericytes") associate with endothelial cells to mediate the formation of tight junctions essential to the function of the blood-brain barrier (BBB). The BBB protects the CNS by regulating the flow of nutrients and toxins into and out of the brain. BBB dysfunction has been implicated in the progression of Alzheimer's disease (AD), but the role of pericytes in BBB dysfunction in AD is not well understood. In the developing embryo, CNS pericytes originate from two sources: mesoderm and neural crest. In this study, we report two protocols using mesoderm or neural crest intermediates, to generate brain-specific pericyte-like cells from induced pluripotent stem cell (iPSC) lines created from healthy and AD patients. iPSC-derived pericytes display stable expression of pericyte surface markers and brain-specific genes and are functionally capable of increasing vascular tube formation and endothelial barrier properties.


Asunto(s)
Barrera Hematoencefálica/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Mesodermo/fisiología , Cresta Neural/fisiología , Pericitos/fisiología , Células Madre Pluripotentes/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Biomarcadores/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiología , Humanos , Mesodermo/metabolismo , Mesodermo/patología , Cresta Neural/metabolismo , Cresta Neural/patología , Pericitos/metabolismo , Pericitos/patología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/patología , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Uniones Estrechas/fisiología
10.
Glia ; 67(5): 844-856, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30588668

RESUMEN

Alzheimer's disease (AD) is the leading cause of age-related neurodegeneration and is characterized neuropathologically by the accumulation of insoluble beta-amyloid (Aß) peptides. In AD brains, plaque-associated myeloid (PAM) cells cluster around Aß plaques but fail to effectively clear Aß by phagocytosis. PAM cells were originally thought to be brain-resident microglia. However, several studies have also suggested that Aß-induced inflammation causes peripheral monocytes to enter the otherwise immune-privileged brain. The relationship between AD progression and inflammation in the brain remains ambiguous because microglia and monocyte-derived macrophages are extremely difficult to distinguish from one another in an inflamed brain. Whether PAM cells are microglia, peripheral macrophages, or a mixture of both remains unclear. CD11a is a component of the ß2 integrin LFA1. We have determined that CD11a is highly expressed on peripheral immune cells, including macrophages, but is not expressed by mouse microglia. These expression patterns remain consistent in LPS-treated inflamed mice, as well as in two mouse models of AD. Thus, CD11a can be used as a marker to distinguish murine microglia from infiltrating peripheral immune cells. Using CD11a, we show that PAM cells in AD transgenic brains are comprised entirely of microglia. We also demonstrate a novel fluorescence-assisted quantification technique (FAQT), which reveals a significant increase in T lymphocytes, especially in the brains of female AD mice. Our findings support the notion that microglia are the lead myeloid players in AD and that rejuvenating their phagocytic potential may be an important therapeutic strategy.


Asunto(s)
Enfermedad de Alzheimer/patología , Antígeno CD11a/metabolismo , Microglía/metabolismo , Microglía/patología , Células Mieloides/metabolismo , Algoritmos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/cirugía , Animales , Animales Recién Nacidos , Trasplante de Médula Ósea , Encéfalo/metabolismo , Encéfalo/patología , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Inflamación/etiología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Células Mieloides/efectos de los fármacos , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Receptores CCR2/genética , Receptores CCR2/metabolismo , Toxoplasmosis/complicaciones
11.
Stem Cells Transl Med ; 7(6): 468-476, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29543389

RESUMEN

Hematopoietic stem cells (HSCs) are the self-renewing multipotent progenitors to all blood cell types. Identification and isolation of HSCs for study has depended on the expression of combinations of surface markers on HSCs that reliably distinguish them from other cell types. However, the increasing number of markers required to isolate HSCs has made it tedious, expensive, and difficult for newcomers, suggesting the need for a simpler panel of HSC markers. We previously showed that phenotypic HSCs could be separated based on expression of CD11a and that only the CD11a negative fraction contained true HSCs. Here, we show that CD11a and another HSC marker, endothelial protein C receptor (EPCR), can be used to effectively identify and purify HSCs. We introduce a new two-color HSC sorting method that can highly enrich for HSCs with efficiencies comparable to the gold standard combination of CD150 and CD48. Our results demonstrate that adding CD11a and EPCR to the HSC biologist's toolkit improves the purity of and simplifies isolation of HSCs. Stem Cells Translational Medicine 2018;7:468-476.


Asunto(s)
Biomarcadores/metabolismo , Antígeno CD11a/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Células Madre Hematopoyéticas/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Citometría de Flujo/métodos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Poli I-C/toxicidad
12.
Cell Rep ; 17(9): 2286-2298, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27880904

RESUMEN

The contribution of the different waves and sites of developmental hematopoiesis to fetal and adult blood production remains unclear. Here, we identify lymphatic vessel endothelial hyaluronan receptor-1 (LYVE1) as a marker of yolk sac (YS) endothelium and definitive hematopoietic stem and progenitor cells (HSPCs). Endothelium in mid-gestation YS and vitelline vessels, but not the dorsal aorta and placenta, were labeled by Lyve1-Cre. Most YS HSPCs and erythro-myeloid progenitors were Lyve1-Cre lineage traced, but primitive erythroid cells were not, suggesting that they represent distinct lineages. Fetal liver (FL) and adult HSPCs showed 35%-40% Lyve1-Cre marking. Analysis of circulation-deficient Ncx1-/- concepti identified the YS as a major source of Lyve1-Cre labeled HSPCs. FL proerythroblast marking was extensive at embryonic day (E) 11.5-13.5, but decreased to hematopoietic stem cell (HSC) levels by E16.5, suggesting that HSCs from multiple sources became responsible for erythropoiesis. Lyve1-Cre thus marks the divergence between YS primitive and definitive hematopoiesis and provides a tool for targeting YS definitive hematopoiesis and FL colonization.


Asunto(s)
Linaje de la Célula , Células Eritroides/citología , Células Eritroides/metabolismo , Hemangioblastos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Saco Vitelino/metabolismo , Envejecimiento , Animales , Eritropoyesis , Femenino , Feto/citología , Eliminación de Gen , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Integrasas/metabolismo , Hígado/embriología , Ratones Endogámicos C57BL , Embarazo , Factores de Tiempo
13.
Proc Natl Acad Sci U S A ; 113(9): E1316-25, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26884167

RESUMEN

The innate immune system is strongly implicated in the pathogenesis of Alzheimer's disease (AD). In contrast, the role of adaptive immunity in AD remains largely unknown. However, numerous clinical trials are testing vaccination strategies for AD, suggesting that T and B cells play a pivotal role in this disease. To test the hypothesis that adaptive immunity influences AD pathogenesis, we generated an immune-deficient AD mouse model that lacks T, B, and natural killer (NK) cells. The resulting "Rag-5xfAD" mice exhibit a greater than twofold increase in ß-amyloid (Aß) pathology. Gene expression analysis of the brain implicates altered innate and adaptive immune pathways, including changes in cytokine/chemokine signaling and decreased Ig-mediated processes. Neuroinflammation is also greatly exacerbated in Rag-5xfAD mice as indicated by a shift in microglial phenotype, increased cytokine production, and reduced phagocytic capacity. In contrast, immune-intact 5xfAD mice exhibit elevated levels of nonamyloid reactive IgGs in association with microglia, and treatment of Rag-5xfAD mice or microglial cells with preimmune IgG enhances Aß clearance. Last, we performed bone marrow transplantation studies in Rag-5xfAD mice, revealing that replacement of these missing adaptive immune populations can dramatically reduce AD pathology. Taken together, these data strongly suggest that adaptive immune cell populations play an important role in restraining AD pathology. In contrast, depletion of B cells and their appropriate activation by T cells leads to a loss of adaptive-innate immunity cross talk and accelerated disease progression.


Asunto(s)
Adaptación Fisiológica , Enfermedad de Alzheimer/fisiopatología , Microglía/patología , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/metabolismo , Animales , Humanos , Inmunoglobulina G/sangre , Ratones , Fagocitosis
14.
Exp Hematol ; 43(7): 578-85, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25892186

RESUMEN

Identification and isolation of hematopoietic stem cells (HSCs) in mice is most commonly based on the expression of surface molecules Kit and Sca-1 and the absence of markers of mature lineages. However, Sca-1 is absent or weakly expressed in hematopoietic progenitors in many strains, including nonobese diabetic (NOD), BALB/c, C3H, and CBA mice. In addition, both Kit and Sca-1 levels are modulated following bone marrow injury. In these cases, other markers and dye exclusion methods have been employed to identify HSCs, yet there is no antibody-based stain that enables identification of HSCs and early progenitors when Kit and Sca-1 are inadequate. CD201 is a marker that is highly restricted to HSCs and progenitors, and CD27 is expressed at moderate-to-high levels on HSCs. We show here that combining CD201 and CD27 enables highly efficient isolation of long-term HSCs in NOD mice as well as in other strains, including SJL, FVB, AKR, BALB/c, C3H, and CBA. We also find that HSCs appear to maintain expression of CD201 and CD27 after hematopoietic injury when Kit expression is downregulated. These results suggest a widely applicable yet simple alternative for HSC isolation in settings where Kit and Sca-1 expression are insufficient.


Asunto(s)
Células Sanguíneas/química , Separación Celular/métodos , Citometría de Flujo/métodos , Células Madre Hematopoyéticas/química , Ratones Endogámicos/sangre , Receptores de Superficie Celular/sangre , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/sangre , Animales , Antígenos Ly/genética , Antígenos Ly/fisiología , Autoinmunidad , Células Sanguíneas/citología , Médula Ósea/efectos de la radiación , Linaje de la Célula , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Receptor de Proteína C Endotelial , Expresión Génica , Células Madre Hematopoyéticas/citología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos NOD , Ratones Endogámicos/genética , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-kit/deficiencia , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/fisiología , Quimera por Radiación , Traumatismos Experimentales por Radiación/sangre , Traumatismos Experimentales por Radiación/patología , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
15.
Mol Biosyst ; 11(3): 783-90, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25502239

RESUMEN

The tamoxifen inducible Cre-ER/loxP system provides tissue specific temporal control of gene recombination events, and can be used to induce expression of reporter genes (e.g. GFP, LacZ) for lineage tracing studies. Cre enzyme fused with estrogen receptor (Cre-ER) is released upon tamoxifen binding, resulting in permanent activation of reporter genes within cells and their progeny. Tamoxifen and its active metabolite, hydroxytamoxifen (4OHT) diffuses rapidly in vivo, making it difficult to restrict labeling to specific locations. In this study, we developed a photocaged 4OHT molecule by covalently attaching 4OHT to an ortho-nitrobenzyl (ONB1) group, rendering 4OHT inactive. Exposure to UV radiation cleaves the bond between ONB1 and 4OHT, freeing the 4OHT to bind Cre-ER to result in downstream genetic recombination and reporter activation. We show that caged ONB1-4OHT crosses the cell membrane and uncages after short UV exposure, resulting in Cre-driven genetic recombination that can be localized to specific regions or tissues. ONB1-4OHT can provide spatial control of reporter activation and be adapted with any existing Cre-ER/loxP based system.


Asunto(s)
Regulación de la Expresión Génica/efectos de la radiación , Expresión Génica , Genes Reporteros , Recombinación Homóloga , Luz , Receptores de Estrógenos/genética , Tamoxifeno/análogos & derivados , Técnicas In Vitro , Tamoxifeno/química
16.
Stem Cell Reports ; 3(5): 707-15, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25418718

RESUMEN

Small numbers of hematopoietic stem cells (HSCs) generate large numbers of mature effector cells through the successive amplification of transiently proliferating progenitor cells. HSCs and their downstream progenitors have been extensively characterized based on their cell-surface phenotype and functional activities during transplantation assays. These cells dynamically lose and acquire specific sets of surface markers during differentiation, leading to the identification of markers that allow for more refined separation of HSCs from early hematopoietic progenitors. Here, we describe a marker, CD11A, which allows for the enhanced purification of mouse HSCs. We show through in vivo transplantations that upregulation of CD11A on HSCs denotes the loss of their long-term reconstitution potential. Surprisingly, nearly half of phenotypic HSCs (defined as Lin-KIT(+)SCA-1(+)CD150(+)CD34-) are CD11A(+) and lack long-term self-renewal potential. We propose that CD11A(+)Lin-KIT(+)SCA-1(+)CD150(+)CD34- cells are multipotent progenitors and CD11A-Lin-KIT(+)SCA-1(+)CD150(+)CD34- cells are true HSCs.


Asunto(s)
Antígeno CD11a/metabolismo , Diferenciación Celular , Proliferación Celular , Células Madre Hematopoyéticas/metabolismo , Regulación hacia Arriba , Animales , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Antígenos Ly/metabolismo , Antígeno CD11a/genética , Citometría de Flujo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de Superficie Celular/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria
17.
Cell Stem Cell ; 15(1): 37-50, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24813857

RESUMEN

Hematopoietic stem cells (HSCs) maintain homeostasis and regenerate the blood system throughout life. It has been postulated that HSCs may be uniquely capable of preserving their genomic integrity in order to ensure lifelong function. To directly test this, we quantified DNA damage in HSCs and downstream progenitors from young and old mice, revealing that strand breaks significantly accrue in HSCs during aging. DNA damage accumulation in HSCs was associated with broad attenuation of DNA repair and response pathways that was dependent upon HSC quiescence. Accordingly, cycling fetal HSCs and adult HSCs driven into cycle upregulated these pathways leading to repair of strand breaks. Our results demonstrate that HSCs are not comprehensively geno-protected during aging. Rather, HSC quiescence and concomitant attenuation of DNA repair and response pathways underlies DNA damage accumulation in HSCs during aging. These results provide a potential mechanism through which premalignant mutations accrue in HSCs.


Asunto(s)
Envejecimiento/fisiología , Células de la Médula Ósea/fisiología , Células Madre Hematopoyéticas/fisiología , Animales , Carcinogénesis/genética , Puntos de Control del Ciclo Celular/genética , Células Cultivadas , Daño del ADN , Reparación del ADN , Homeostasis , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Mutación/genética , Quimera por Trasplante
18.
Stem Cell Reports ; 2(4): 457-72, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24749071

RESUMEN

Hematopoiesis in the embryo proceeds in a series of waves, with primitive erythroid-biased waves succeeded by definitive waves, within which the properties of hematopoietic stem cells (multilineage potential, self-renewal, and engraftability) gradually arise. Whereas self-renewal and engraftability have previously been examined in the embryo, multipotency has not been thoroughly addressed, especially at the single-cell level or within well-defined populations. To identify when and where clonal multilineage potential arises during embryogenesis, we developed a single-cell multipotency assay. We find that, during the initiation of definitive hematopoiesis in the embryo, a defined population of multipotent, engraftable progenitors emerges that is much more abundant within the yolk sac (YS) than the aorta-gonad-mesonephros (AGM) or fetal liver. These experiments indicate that multipotent cells appear in concert within both the YS and AGM and strongly implicate YS-derived progenitors as contributors to definitive hematopoiesis.


Asunto(s)
Desarrollo Embrionario , Células Madre Hematopoyéticas/metabolismo , Células Madre Multipotentes/metabolismo , Animales , Antígenos de Superficie/metabolismo , Antígeno CD11a/genética , Diferenciación Celular , Linaje de la Célula , Ensayo de Unidades Formadoras de Colonias , Desarrollo Embrionario/genética , Femenino , Células Madre Hematopoyéticas/citología , Inmunofenotipificación , Leucosialina/genética , Leucosialina/metabolismo , Ratones , Células Madre Multipotentes/citología , Fenotipo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Saco Vitelino/embriología
19.
Proc Natl Acad Sci U S A ; 110(27): 11103-8, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23690610

RESUMEN

Mobilization of the T-cell response against cancer has the potential to achieve long-lasting cures. However, it is not known how to harness antigen-presenting cells optimally to achieve an effective antitumor T-cell response. In this study, we show that anti-CD47 antibody-mediated phagocytosis of cancer by macrophages can initiate an antitumor T-cell immune response. Using the ovalbumin model antigen system, anti-CD47 antibody-mediated phagocytosis of cancer cells by macrophages resulted in increased priming of OT-I T cells [cluster of differentiation 8-positive (CD8(+))] but decreased priming of OT-II T cells (CD4(+)). The CD4(+) T-cell response was characterized by a reduction in forkhead box P3-positive (Foxp3(+)) regulatory T cells. Macrophages following anti-CD47-mediated phagocytosis primed CD8(+) T cells to exhibit cytotoxic function in vivo. This response protected animals from tumor challenge. We conclude that anti-CD47 antibody treatment not only enables macrophage phagocytosis of cancer but also can initiate an antitumor cytotoxic T-cell immune response.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígeno CD47/inmunología , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Macrófagos/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Bloqueadores/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Neoplasias del Colon/patología , Citotoxicidad Inmunológica/genética , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagocitosis/inmunología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...