Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
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
2.
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
3.
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
4.
Genes Dev ; 23(20): 2376-81, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19833765

RESUMEN

Common lymphoid progenitors (CLPs) clonally produce both B- and T-cell lineages, but have little myeloid potential in vivo. However, some studies claim that the upstream lymphoid-primed multipotent progenitor (LMPP) is the thymic seeding population, and suggest that CLPs are primarily B-cell-restricted. To identify surface proteins that distinguish functional CLPs from B-cell progenitors, we used a new computational method of Mining Developmentally Regulated Genes (MiDReG). We identified Ly6d, which divides CLPs into two distinct populations: one that retains full in vivo lymphoid potential and produces more thymocytes at early timepoints than LMPP, and another that behaves essentially as a B-cell progenitor.


Asunto(s)
Antígenos Ly/metabolismo , Linfocitos B/citología , Linfocitos B/metabolismo , Diferenciación Celular , Células Progenitoras Linfoides/citología , Linfocitos T/citología , Linfocitos T/metabolismo , Animales , Antígenos Ly/genética , Células Cultivadas , Proteínas Ligadas a GPI , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL
5.
Nature ; 467(7313): 338-42, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20720541

RESUMEN

Epigenetic modifications must underlie lineage-specific differentiation as terminally differentiated cells express tissue-specific genes, but their DNA sequence is unchanged. Haematopoiesis provides a well-defined model to study epigenetic modifications during cell-fate decisions, as multipotent progenitors (MPPs) differentiate into progressively restricted myeloid or lymphoid progenitors. Although DNA methylation is critical for myeloid versus lymphoid differentiation, as demonstrated by the myeloerythroid bias in Dnmt1 hypomorphs, a comprehensive DNA methylation map of haematopoietic progenitors, or of any multipotent/oligopotent lineage, does not exist. Here we examined 4.6 million CpG sites throughout the genome for MPPs, common lymphoid progenitors (CLPs), common myeloid progenitors (CMPs), granulocyte/macrophage progenitors (GMPs), and thymocyte progenitors (DN1, DN2, DN3). Marked epigenetic plasticity accompanied both lymphoid and myeloid restriction. Myeloid commitment involved less global DNA methylation than lymphoid commitment, supported functionally by myeloid skewing of progenitors following treatment with a DNA methyltransferase inhibitor. Differential DNA methylation correlated with gene expression more strongly at CpG island shores than CpG islands. Many examples of genes and pathways not previously known to be involved in choice between lymphoid/myeloid differentiation have been identified, such as Arl4c and Jdp2. Several transcription factors, including Meis1, were methylated and silenced during differentiation, indicating a role in maintaining an undifferentiated state. Additionally, epigenetic modification of modifiers of the epigenome seems to be important in haematopoietic differentiation. Our results directly demonstrate that modulation of DNA methylation occurs during lineage-specific differentiation and defines a comprehensive map of the methylation and transcriptional changes that accompany myeloid versus lymphoid fate decisions.


Asunto(s)
Linaje de la Célula , Metilación de ADN , Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Línea Celular , Linaje de la Célula/genética , Islas de CpG/genética , Metilación de ADN/genética , Epigénesis Genética , Perfilación de la Expresión Génica , Genoma/genética , Hematopoyesis/genética , Linfocitos/citología , Linfocitos/metabolismo , Metaboloma , Metabolómica , Ratones , Células Mieloides/citología , Células Mieloides/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(9): 3405-10, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23391730

RESUMEN

A goal of regenerative medicine is to identify cardiovascular progenitors from human ES cells (hESCs) that can functionally integrate into the human heart. Previous studies to evaluate the developmental potential of candidate hESC-derived progenitors have delivered these cells into murine and porcine cardiac tissue, with inconclusive evidence regarding the capacity of these human cells to physiologically engraft in xenotransplantation assays. Further, the potential of hESC-derived cardiovascular lineage cells to functionally couple to human myocardium remains untested and unknown. Here, we have prospectively identified a population of hESC-derived ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells that give rise to cardiomyocytes, endothelial cells, and vascular smooth muscle cells in vitro at a clonal level. We observed rare clusters of ROR2(+) cells and diffuse expression of KDR and PDGFRα in first-trimester human fetal hearts. We then developed an in vivo transplantation model by transplanting second-trimester human fetal heart tissues s.c. into the ear pinna of a SCID mouse. ROR2(+)/CD13(+)/KDR(+)/PDGFRα(+) cells were delivered into these functioning fetal heart tissues: in contrast to traditional murine heart models for cell transplantation, we show structural and functional integration of hESC-derived cardiovascular progenitors into human heart.


Asunto(s)
Células Madre Embrionarias/citología , Feto/citología , Corazón/embriología , Miocitos Cardíacos/citología , Miocitos Cardíacos/trasplante , Trasplante de Células Madre , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Separación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Endotelio Vascular/citología , Feto/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Mesodermo/citología , Ratones , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Miocardio/citología , Miocitos Cardíacos/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Línea Primitiva/citología , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Supervivencia Tisular , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
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
8.
Proc Natl Acad Sci U S A ; 108(8): 3282-7, 2011 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-21300885

RESUMEN

The promise of pluripotent stem cells as a research and therapeutic tool is partly undermined by the technical challenges of generating and maintaining these cells in culture. Human embryonic stem cells (hESCs) are exquisitely sensitive to culture conditions, and require constant signaling by growth factors and cell-cell and cell-matrix interactions to prevent apoptosis, senescence, and differentiation. Previous work from our laboratory demonstrated that overexpression of the prosurvival gene BCL2 in mouse embryonic stem cells overrode the requirement of serum factors and feeder cells to maintain mESCs in culture. To determine whether this prosurvival gene could similarly protect hESCs, we generated hESC lines that constitutively or inducibly express BCL2. We find that BCL2 overexpression significantly decreases dissociation-induced apoptosis, resulting in enhanced colony formation from sorted single cells, and enhanced embryoid body formation. In addition, BCL2-hESCs exhibit normal growth in the absence of serum, but require basic fibroblast growth factor to remain undifferentiated. Furthermore, they maintain their pluripotency markers, form teratomas in vivo, and differentiate into all three germ layers. Our data suggest that the BCL2 signaling pathway plays an important role in inhibiting hESC apoptosis, such that its overexpression in hESCs offers both a survival benefit in conditions of stress by resisting apoptosis and obviates the requirement for serum or a feeder layer for maintenance.


Asunto(s)
Células Madre Embrionarias/citología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Supervivencia Celular/genética , Células Madre Embrionarias/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Genes bcl-2/fisiología , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Suero , Estrés Fisiológico
9.
Blood ; 118(20): 5439-47, 2011 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-21931117

RESUMEN

Natural killer (NK) cells develop in the bone marrow and are known to gradually acquire the ability to eliminate infected and malignant cells, yet the cellular stages of NK lineage commitment and maturation are incompletely understood. Using 12-color flow cytometry, we identified a novel NK-committed progenitor (pre-NKP) that is a developmental intermediate between the upstream common lymphoid progenitor and the downstream NKP, previously assumed to represent the first stage of NK lineage commitment. Our analysis also refined the purity of NKPs (rNKP) by 6-fold such that 50% of both pre-NKP and rNKP cells gave rise to NKp46+ NK cells at the single-cell level. On transplantation into unconditioned Rag2-/-Il2rγc-/- recipients, both pre-NKPs and rNKPs generated mature NK cells expressing a repertoire of Ly49 family members that degranulated on stimulation ex vivo. Intrathymic injection of these progenitors, however, yielded no NK cells, suggesting a separate origin of thymic NK cells. Unlike the rNKP, the pre-NKP does not express IL-2Rß (CD122), yet it is lineage committed toward the NK cell fate, adding support to the theory that IL-15 signaling is not required for NK commitment. Taken together, our data provide a high-resolution in vivo analysis of the earliest steps of NK cell commitment and maturation.


Asunto(s)
Células de la Médula Ósea/citología , Linaje de la Célula/inmunología , Células Asesinas Naturales/citología , Células Progenitoras Linfoides/citología , Timocitos/citología , Animales , Biomarcadores , Diferenciación Celular/inmunología , Separación Celular/métodos , Células Cultivadas , Citometría de Flujo/métodos , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes
10.
Proc Natl Acad Sci U S A ; 107(13): 5732-7, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20231483

RESUMEN

We present a method termed mining developmentally regulated genes (MiDReG) to predict genes whose expression is either activated or repressed as precursor cells differentiate. MiDReG does not require gene expression data from intermediate stages of development. MiDReG is based on the gene expression patterns between the initial and terminal stages of the differentiation pathway, coupled with "if-then" rules (Boolean implications) mined from large-scale microarray databases. MiDReG uses two gene expression-based seed conditions that mark the initial and the terminal stages of a given differentiation pathway and combines the statistically inferred Boolean implications from these seed conditions to identify the relevant genes. The method was validated by applying it to B-cell development. The algorithm predicted 62 genes that are expressed after the KIT+ progenitor cell stage and remain expressed through CD19+ and AICDA+ germinal center B cells. qRT-PCR of 14 of these genes on sorted B-cell progenitors confirmed that the expression of 10 genes is indeed stably established during B-cell differentiation. Review of the published literature of knockout mice revealed that of the predicted genes, 63.4% have defects in B-cell differentiation and function and 22% have a role in the B cell according to other experiments, and the remaining 14.6% are not characterized. Therefore, our method identified novel gene candidates for future examination of their role in B-cell development. These data demonstrate the power of MiDReG in predicting functionally important intermediate genes in a given developmental pathway that is defined by a mutually exclusive gene expression pattern.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Algoritmos , Animales , Antígenos CD19/genética , Biología Computacional , Citidina Desaminasa/genética , Minería de Datos/estadística & datos numéricos , Bases de Datos Genéticas , Perfilación de la Expresión Génica/estadística & datos numéricos , Humanos , Ratones , Modelos Estadísticos , Análisis de Secuencia por Matrices de Oligonucleótidos/estadística & datos numéricos , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/metabolismo , Factor de Células Madre/genética
11.
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
12.
J Exp Med ; 203(7): 1721-32, 2006 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-16785310

RESUMEN

V(D)J recombination of immunoglobulin (Ig) heavy (IgH) and light chain genes occurs sequentially in the pro- and pre-B cells. To identify cis-elements that dictate this order of rearrangement, we replaced the endogenous matrix attachment region/Igk intronic enhancer (MiE(kappa)) with its heavy chain counterpart (Emu) in mice. This replacement, denoted EmuR, substantially increases the accessibility of both V(kappa) and J(kappa) loci to V(D)J recombinase in pro-B cells and induces Igk rearrangement in these cells. However, EmuR does not support Igk rearrangement in pre-B cells. Similar to that in MiE(kappa)(-/-) pre-B cells, the accessibility of V(kappa) segments to V(D)J recombinase is considerably reduced in EmuR pre-B cells when compared with wild-type pre-B cells. Therefore, Emu and MiE(kappa) play developmental stage-specific roles in maintaining the sequential rearrangement of IgH and Igk loci by promoting the accessibility of V, D, and J loci to the V(D)J recombinase.


Asunto(s)
Elementos de Facilitación Genéticos/fisiología , Reordenamiento Génico de Cadena Pesada de Linfocito B , Reordenamiento Génico de Cadena Ligera de Linfocito B , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/genética , Intrones/fisiología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Ratones , Ratones Mutantes , Células Madre/inmunología , Células Madre/metabolismo
13.
Proc Natl Acad Sci U S A ; 106(6): 1930-5, 2009 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-19181846

RESUMEN

Hematopoiesis is a tightly controlled process maintained by a small pool of hematopoietic stem cells (HSCs). Here, we demonstrate that the LT-HSC, MPP, premegakaryocytic/erythroid, Pre CFU-E, Pre GM, MkP, and granulocyte-macrophage compartments were all significantly reduced in E2A-deficient bone marrow. Despite a severe depletion of erythroid progenitors, the erythrocyte and megakaryocyte compartments were equivalent in E2A-deficient bone marrow as compared with wild-type mice. E2A-deficient HSCs also failed to efficiently maintain the HSC pool on serial transplantation, and we demonstrate that the E2A proteins regulate cell cycle progression of HSCs by regulating the expression of p21(Cip1), p27(Kip1), and the thrombopoietin receptor, known regulators of HSC self-renewal activity. Based on these observations, we propose that the E2A proteins promote the developmental progression of the entire spectrum of early hematopoietic progenitors and to suppress an erythroid specific program of gene expression in alternative cell lineages. Last, the data mechanistically link E2A, cell cycle regulators, and the maintenance of the HSC pool in a common pathway.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Hematopoyesis , Células Madre Hematopoyéticas/citología , Animales , Ciclo Celular , Proteínas de Ciclo Celular , Diferenciación Celular/fisiología , Células Precursoras Eritroides , Células Progenitoras Linfoides , Ratones , Células Progenitoras Mieloides
14.
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
15.
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.

16.
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
17.
J Exp Med ; 200(9): 1205-11, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15504821

RESUMEN

The immunoglobulin kappa light chain intronic enhancer (iEkappa) activates kappa rearrangement and is required to maintain the earlier or more efficient rearrangement of kappa versus lambda (lambda). To understand the mechanism of how iEkappa regulates kappa rearrangement, we employed homologous recombination to mutate individual functional motifs within iE(kappa) in the endogenous kappa locus, including the NF-kappaB binding site (kappaB), as well as kappaE1, kappaE2, and kappaE3 E boxes. Analysis of the impacts of these mutations revealed that kappaE2 and to a lesser extent kappaE1, but not kappaE3, were important for activating kappa rearrangement. Surprisingly, mutation of the kappaB site had no apparent effect on kappa rearrangement. Comparable to the deletion of the entire iEkappa, simultaneous mutation of kappaE1 and kappaE2 reduces the efficiency of kappa rearrangement much more dramatically than either kappaE1 or kappaE2 mutation alone. Because E2A family proteins are the only known factors that bind to these E boxes, these findings provide unambiguous evidence that E2A is a key regulator of kappa rearrangement.


Asunto(s)
Sitios de Unión/inmunología , Elementos E-Box/genética , Reordenamiento Génico de Cadena Ligera de Linfocito B/genética , Cadenas kappa de Inmunoglobulina/genética , Animales , Linfocitos B/inmunología , Southern Blotting , Cartilla de ADN , Elementos E-Box/inmunología , Ratones , Ratones Mutantes , Mutagénesis Sitio-Dirigida , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/inmunología , Transfección
18.
Blood ; 111(12): 5562-70, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18424665

RESUMEN

Mature blood cells develop from multipotent hematopoietic stem cells through a series of sequential intermediates in which the developmental potential for particular blood lineages is progressively extinguished. We previously reported the identification of one of these developmental intermediates, the common lymphoid progenitor (CLP), which can give rise to T cells, B cells, dendritic cells (DCs), and natural killer cells (NKs), but lacks myeloid and erythroid potential. Recently, several studies have suggested that the T-cell and DC potential of CLP is limited or absent, and/or that CLP contains significant myeloid potential. Here, we show that the originally identified CLP population can be divided into functionally distinct subsets based on the expression of the tyrosine kinase receptor, Flk2. The Flk2(+) subset contains robust in vivo and in vitro T-cell, B-cell, DC, and NK potential, but lacks myeloid potential and, therefore, represents an oligopotent, lymphoid-restricted progenitor. This population of cells does not appear to be B cell-biased and robustly reconstitutes both B and T lineages in vivo, consistent with its being a physiologic progenitor of both of these subsets. Thus, Flk2 expression defines a homogeneous, readily obtainable subset of bone marrow CLP that is completely lymphoid-committed and can differentiate equivalently well into both B and T lineages.


Asunto(s)
Linfocitos B/citología , Linaje de la Célula/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Linfocitos T/citología , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Animales Congénicos , Linfocitos B/metabolismo , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Inmunofenotipificación , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Células Mieloides/citología , Células Mieloides/metabolismo , Linfocitos T/metabolismo
19.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA