Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 143(17): 1689-1701, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38364184

RESUMEN

ABSTRACT: Over the past 10 years, there has been a marked increase in recognition of the interplay between the intestinal microbiome and the hematopoietic system. Despite their apparent distance in the body, a large literature now supports the relevance of the normal intestinal microbiota to steady-state blood production, affecting both hematopoietic stem and progenitor cells as well as differentiated immune cells. Microbial metabolites enter the circulation where they can trigger cytokine signaling that influences hematopoiesis. Furthermore, the state of the microbiome is now recognized to affect outcomes from hematopoietic stem cell transplant, immunotherapy, and cellular therapies for hematologic malignancies. Here we review the mechanisms by which microbiotas influence hematopoiesis in development and adulthood as well as the avenues by which microbiotas are thought to impact stem cell transplant engraftment, graft-versus-host disease, and efficacy of cell and immunotherapies. We highlight areas of future research that may lead to reduced adverse effects of antibiotic use and improved outcomes for patients with hematologic conditions.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Hematológicas , Hematopoyesis , Humanos , Enfermedades Hematológicas/terapia , Enfermedades Hematológicas/microbiología , Animales , Trasplante de Células Madre Hematopoyéticas , Enfermedad Injerto contra Huésped/microbiología , Enfermedad Injerto contra Huésped/terapia , Enfermedad Injerto contra Huésped/inmunología
2.
Trends Immunol ; 44(10): 751-753, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37704548

RESUMEN

'Long COVID' affects nearly one in five adults who have had coronavirus disease 2019 (COVID-19), yet the mechanisms underlying this disorder remain poorly understood. In a new study, Cheong et al. show that the epigenetic and transcriptional state of myeloid immune cells and their progenitors are durably altered in patients following severe COVID-19.


Asunto(s)
COVID-19 , Adulto , Humanos , COVID-19/genética , SARS-CoV-2 , Síndrome Post Agudo de COVID-19 , Epigénesis Genética
3.
PLoS Biol ; 21(5): e3002104, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37141182

RESUMEN

Tumors protect themselves from immune clearance by promoting extramedullary hematopoiesis. A new study in PLOS Biology provides insights into the mechanisms underlying this process, which may hold the key to disrupting generation of the immunosuppressive tumor microenvironment.


Asunto(s)
Enfermedades Hematológicas , Hematopoyesis Extramedular , Neoplasias , Humanos , Factor Inhibidor de Leucemia , Interleucina-1alfa , Hematopoyesis , Microambiente Tumoral
4.
Blood ; 132(6): 559-564, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-29853538

RESUMEN

Recent studies have revealed that the intestinal bacterial microbiome plays an important role in the regulation of hematopoiesis. A correlation between adverse hematologic effects and imbalance of the intestinal microbiome, or dysbiosis, is evident in several human conditions, such as inflammatory bowel disease, obesity, and, critically, in the setting of antibiotic exposure. Here we review the effects of gut dysbiosis on the hematological compartment and our current understanding of the mechanisms through which changes in the bacterial microbiome affect hematopoiesis.


Asunto(s)
Disbiosis/complicaciones , Microbioma Gastrointestinal , Hematopoyesis , Animales , Antibacterianos/efectos adversos , Antibacterianos/farmacología , Médula Ósea/fisiología , Disbiosis/microbiología , Disbiosis/fisiopatología , Microbioma Gastrointestinal/efectos de los fármacos , Supervivencia de Injerto , Hematopoyesis/genética , Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/microbiología , Ratones , Modelos Inmunológicos , Factor 88 de Diferenciación Mieloide/fisiología , Neutropenia/inducido químicamente , Proteína Adaptadora de Señalización NOD1/fisiología , Trastornos Nutricionales/complicaciones , Trastornos Nutricionales/microbiología , Transducción de Señal , Organismos Libres de Patógenos Específicos , Receptores Toll-Like/fisiología
5.
Nature ; 510(7505): 393-6, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24870234

RESUMEN

A unique property of many adult stem cells is their ability to exist in a non-cycling, quiescent state. Although quiescence serves an essential role in preserving stem cell function until the stem cell is needed in tissue homeostasis or repair, defects in quiescence can lead to an impairment in tissue function. The extent to which stem cells can regulate quiescence is unknown. Here we show that the stem cell quiescent state is composed of two distinct functional phases, G0 and an 'alert' phase we term G(Alert). Stem cells actively and reversibly transition between these phases in response to injury-induced systemic signals. Using genetic mouse models specific to muscle stem cells (or satellite cells), we show that mTORC1 activity is necessary and sufficient for the transition of satellite cells from G0 into G(Alert) and that signalling through the HGF receptor cMet is also necessary. We also identify G0-to-G(Alert) transitions in several populations of quiescent stem cells. Quiescent stem cells that transition into G(Alert) possess enhanced tissue regenerative function. We propose that the transition of quiescent stem cells into G(Alert) functions as an 'alerting' mechanism, an adaptive response that positions stem cells to respond rapidly under conditions of injury and stress, priming them for cell cycle entry.


Asunto(s)
Ciclo Celular/fisiología , Complejos Multiproteicos/metabolismo , Músculo Esquelético/citología , Fase de Descanso del Ciclo Celular/fisiología , Células Satélite del Músculo Esquelético/citología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Ciclo Celular/genética , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos/genética , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Regeneración/fisiología , Fase de Descanso del Ciclo Celular/genética , Células Satélite del Músculo Esquelético/metabolismo , Serina-Treonina Quinasas TOR/genética
6.
Blood ; 129(6): 729-739, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-27879260

RESUMEN

Bone marrow suppression is an adverse effect associated with many antibiotics, especially when administered for prolonged treatment courses. Recent advances in our understanding of steady-state hematopoiesis have allowed us to explore the effects of antibiotics on hematopoietic progenitors in detail using a murine model. Antibiotic-treated mice exhibited anemia, thrombocytosis, and leukopenia, with pronounced pan-lymphopenia as demonstrated by flow cytometric analysis of peripheral blood. Bone marrow progenitor analysis revealed depletion of hematopoietic stem cells and multipotent progenitors across all subtypes. Granulocytes and B cells were also diminished in the bone marrow, whereas the number of CD8+ T cells increased. Reductions in progenitor activity were not observed when cells were directly incubated with antibiotics, suggesting that these effects are indirect. Hematopoietic changes were associated with a significant contraction of the fecal microbiome and were partially rescued by fecal microbiota transfer. Further, mice raised in germ-free conditions had hematopoietic abnormalities similar to those seen in antibiotic-treated mice, and antibiotic therapy of germ-free mice caused no additional abnormalities. The effects of antibiotics were phenocopied in Stat1-deficient mice, with no additional suppression by antibiotics in these mice. We conclude that microbiome depletion as a result of broad-spectrum antibiotic treatment disrupts basal Stat1 signaling and alters T-cell homeostasis, leading to impaired progenitor maintenance and granulocyte maturation. Methods to preserve the microbiome may reduce the incidence of antibiotic-associated bone marrow suppression.


Asunto(s)
Anemia/inducido químicamente , Antibacterianos/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Leucopenia/inducido químicamente , Factor de Transcripción STAT1/genética , Trombocitosis/inducido químicamente , Anemia/microbiología , Anemia/patología , Anemia/terapia , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/patología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/fisiología , Expresión Génica , Vida Libre de Gérmenes/efectos de los fármacos , Vida Libre de Gérmenes/genética , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Granulocitos/patología , Hematopoyesis/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Leucopenia/microbiología , Leucopenia/patología , Leucopenia/terapia , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT1/deficiencia , Transducción de Señal , Trombocitosis/microbiología , Trombocitosis/patología , Trombocitosis/terapia
7.
Eur J Haematol ; 97(3): 261-70, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26660446

RESUMEN

Heterozygous mutations in the transcriptional regulator GATA-2 associate with multilineage immunodeficiency, myelodysplastic syndrome (MDS), and acute myeloid leukemia (AML). The majority of these mutations localize in the zinc finger (ZnF) domains, which mediate GATA-2 DNA binding. Deregulated hematopoiesis with GATA-2 mutation frequently develops in adulthood, yet GATA-2 function in the bone marrow remains unresolved. To investigate this, we conditionally deleted the GATA-2 C-terminal ZnF (C-ZnF) coding sequences in adult mice. Upon Gata2 C-ZnF deletion, we observed rapid peripheral cytopenia, bone marrow failure, and decreased c-Kit expression on hematopoietic progenitors. Transplant studies indicated GATA-2 has a cell-autonomous role in bone marrow hematopoiesis. Moreover, myeloid lineage populations were particularly sensitive to Gata2 hemizygosity, while molecular assays indicated GATA-2 regulates c-Kit expression in multilineage progenitor cells. Enforced c-Kit expression in Gata2 C-ZnF-deficient hematopoietic progenitors enhanced myeloid colony activity, suggesting GATA-2 sustains myelopoiesis via a cell intrinsic role involving maintenance of c-Kit expression. Our results provide insight into mechanisms regulating hematopoiesis in bone marrow and may contribute to a better understanding of immunodeficiency and bone marrow failure associated with GATA-2 mutation.


Asunto(s)
Anemia Aplásica/genética , Enfermedades de la Médula Ósea/genética , Médula Ósea/patología , Factor de Transcripción GATA2/genética , Hemoglobinuria Paroxística/genética , Dominios y Motivos de Interacción de Proteínas/genética , Proteínas Proto-Oncogénicas c-kit/deficiencia , Eliminación de Secuencia , Dedos de Zinc/genética , Anemia Aplásica/diagnóstico , Anemia Aplásica/metabolismo , Anemia Aplásica/mortalidad , Animales , Biomarcadores , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Enfermedades de la Médula Ósea/diagnóstico , Enfermedades de la Médula Ósea/metabolismo , Enfermedades de la Médula Ósea/mortalidad , Trastornos de Fallo de la Médula Ósea , Huesos/patología , Inmunoprecipitación de Cromatina , Descalcificación Patológica/genética , Modelos Animales de Enfermedad , Factor de Transcripción GATA2/química , Factor de Transcripción GATA2/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Frecuencia de los Genes , Genes Reporteros , Genotipo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/metabolismo , Hemoglobinuria Paroxística/mortalidad , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunofenotipificación , Ratones , Ratones Noqueados , Pronóstico , Células de Población Lateral
8.
Nature ; 465(7299): 793-7, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20535209

RESUMEN

Lymphocytes and neutrophils are rapidly depleted by systemic infection. Progenitor cells of the haematopoietic system, such as common myeloid progenitors and common lymphoid progenitors, increase the production of immune cells to restore and maintain homeostasis during chronic infection, but the contribution of haematopoietic stem cells (HSCs) to this process is largely unknown. Here we show, using an in vivo mouse model of Mycobacterium avium infection, that an increased proportion of long-term repopulating HSCs proliferate during M. avium infection, and that this response requires interferon-gamma (IFN-gamma) but not interferon-alpha (IFN-alpha) signalling. Thus, the haematopoietic response to chronic bacterial infection involves the activation not only of intermediate blood progenitors but of long-term repopulating HSCs as well. IFN-gamma is sufficient to promote long-term repopulating HSC proliferation in vivo; furthermore, HSCs from IFN-gamma-deficient mice have a lower proliferative rate, indicating that baseline IFN-gamma tone regulates HSC activity. These findings implicate IFN-gamma both as a regulator of HSCs during homeostasis and under conditions of infectious stress. Our studies contribute to a deeper understanding of haematological responses in patients with chronic infections such as HIV/AIDS or tuberculosis.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Homeostasis/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Tuberculosis/inmunología , Animales , Trasplante de Médula Ósea , Recuento de Células , Proliferación Celular , Enfermedad Crónica , Homeostasis/fisiología , Interferón-alfa , Interferón gamma/deficiencia , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/citología , Células Madre Multipotentes/inmunología , Mycobacterium avium/inmunología , Transducción de Señal , Tuberculosis/sangre , Tuberculosis/microbiología
9.
Stem Cells ; 32(11): 3023-30, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25078851

RESUMEN

Interferon gamma (IFNγ) promotes cell division of hematopoietic stem cells (HSCs) without affecting the total HSC number. We postulated that IFNγ stimulates differentiation of HSCs as part of the innate immune response. Here, we report that type II interferon signaling is required, both at baseline and during an animal model of LCMV infection, to maintain normal myeloid development. By separately evaluating myeloid-biased and lymphoid-biased HSC subtypes, we found that myeloid-biased HSCs express higher levels of IFNγ receptor and are specifically activated to divide after recombinant IFNγ exposure in vivo. While both HSC subtypes show increased expression of the transcription factor C/EBPß after infection, only the myeloid-biased HSCs are transiently depleted from the marrow during the type II interferon-mediated immune response to Mycobacterium avium infection, as measured both functionally and phenotypically. These findings indicate that IFNγ selectively permits differentiation of myeloid-biased HSCs during an innate immune response to infection. This represents the first report of a context and a mechanism for discriminate utilization of the alternate HSC subtypes. Terminal differentiation, at the expense of self-renewal, may compromise HSC populations during states of chronic inflammation.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Células Madre Hematopoyéticas/metabolismo , Interferón gamma/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Proliferación Celular/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Ratones Endogámicos C57BL , Células Mieloides/citología , Transducción de Señal/genética
10.
Trends Immunol ; 32(2): 57-65, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21233016

RESUMEN

Hematopoietic stem cells (HSCs) are the progenitors of all blood and immune cells, yet their role in immunity is not well understood. Most studies have focused on the ability of committed lymphoid and myeloid precursors to replenish immune cells during infection. Recent studies, however, have indicated that HSCs also proliferate in response to systemic infection and replenish effector immune cells. Inflammatory signaling molecules including interferons, tumor necrosis factor-α and Toll-like receptors are essential to the HSC response. Observing the biology of HSCs through the lens of infection and inflammation has led to the discovery of an array of immune-mediators that serve crucial roles in HSC regulation and function.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Inflamación/inmunología , Animales , Proliferación Celular , Citocinas/inmunología , Células Madre Hematopoyéticas/citología , Humanos , Transducción de Señal , Receptores Toll-Like/inmunología
11.
Exp Hematol ; 134: 104215, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38580008

RESUMEN

Quiescence and differentiation of hematopoietic stem and progenitor cells (HSPC) can be modified by systemic inflammatory cues. Such cues can not only yield short-term changes in HSPCs such as in supporting emergency granulopoiesis but can also promote lasting influences on the HSPC compartment. First, inflammation can be a driver for clonal expansion, promoting clonal hematopoiesis for certain mutant clones, reducing overall clonal diversity, and reshaping the composition of the HSPC pool with significant health consequences. Second, inflammation can generate lasting cell-autonomous changes in HSPCs themselves, leading to changes in the epigenetic state, metabolism, and function of downstream innate immune cells. This concept, termed "trained immunity," suggests that inflammatory stimuli can alter subsequent immune responses leading to improved innate immunity or, conversely, autoimmunity. Both of these concepts have major implications in human health. Here we reviewed current literature about the lasting effects of inflammation on the HSPC compartment and opportunities for future advancement in this fast-developing field.


Asunto(s)
Células Madre Hematopoyéticas , Inflamación , Humanos , Inflamación/patología , Inflamación/inmunología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/citología , Animales , Inmunidad Innata , Epigénesis Genética , Diferenciación Celular , Hematopoyesis
12.
medRxiv ; 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38712139

RESUMEN

Hematologic side effects are associated with prolonged antibiotic exposure in up to 34% of patients. Neutropenia, reported in 10-15% of patients, increases the risk of sepsis and death. Murine studies have established a link between the intestinal microbiota and normal hematopoiesis. We sought to identify predisposing factors, presence of microbiota-derived metabolites, and changes in intestinal microbiota composition in otherwise healthy pediatric patients who developed neutropenia after prolonged courses of antibiotics. In this multi-center study, patients with infections requiring anticipated antibiotic treatment of two or more weeks were enrolled. Stool samples were obtained at the start and completion of antibiotics and at the time of neutropenia. We identified 10 patients who developed neutropenia on antibiotics and 29 controls matched for age, sex, race, and ethnicity. Clinical data demonstrated no association between neutropenia and type of infection or type of antibiotic used; however intensive care unit admission and length of therapy were associated with neutropenia. Reduced intestinal microbiome richness and decreased abundance of Lachnospiraceae family members correlated with neutropenia. Untargeted stool metabolomic profiling revealed several metabolites that were depleted exclusively in patients with neutropenia, including members of the urea cycle pathway, pyrimidine metabolism and fatty acid metabolism that are known to be produced by Lachnospiraceae . Our study confirms a relationship between intestinal microbiota disruption and abnormal hematopoiesis and identifies taxa and metabolites likely to contribute to microbiota-sustained hematopoiesis. As the microbiome is a key determinant of stem cell transplant and immunotherapy outcomes, these findings are likely to be of broad significance. Key Points: Neutropenia occurred in 17% of patients receiving prolonged antibiotic therapy.We found no association between neutropenia and type of infection or class of antibiotic used. Development of neutropenia after prolonged antibiotic treatment was associated with decreased prevalence of Lachnospiraceae and Lachnospiraceae metabolites such as citrulline.

13.
bioRxiv ; 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38826218

RESUMEN

Analysis of lung alveolar type 2 (AT2) progenitor stem cells has highlighted fundamental mechanisms that direct their differentiation into alveolar type 1 cells (AT1s) in lung repair and disease. However, microRNA (miRNA) mediated post-transcriptional mechanisms which govern this nexus remain understudied. We show here that the let-7 miRNA family serves a homeostatic role in governance of AT2 quiescence, specifically by preventing the uncontrolled accumulation of AT2 transitional cells and by promoting AT1 differentiation to safeguard the lung from spontaneous alveolar destruction and fibrosis. Using mice and organoid models with genetic ablation of let-7a1/let-7f1/let-7d cluster (let-7afd) in AT2 cells, we demonstrate prevents AT1 differentiation and results in aberrant accumulation of AT2 transitional cells in progressive pulmonary fibrosis. Integration of enhanced AGO2 UV-crosslinking and immunoprecipitation sequencing (AGO2-eCLIP) with RNA-sequencing from AT2 cells uncovered the induction of direct targets of let-7 in an oncogene feed-forward regulatory network including BACH1/EZH2 which drives an aberrant fibrotic cascade. Additional analyses by CUT&RUN-sequencing revealed loss of let-7afd hampers AT1 differentiation by eliciting aberrant histone EZH2 methylation which prevents the exit of AT2 transitional cells into terminal AT1s. This study identifies let-7 as a key gatekeeper of post-transcriptional and epigenetic chromatin signals to prevent AT2-driven pulmonary fibrosis.

14.
Blood ; 118(6): 1525-33, 2011 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-21633090

RESUMEN

The IFN-inducible immunity-related p47 GTPase Irgm1 has been linked to Crohn disease as well as susceptibility to tuberculosis. Previously we demonstrated that HSC quiescence and function are aberrant in mice lacking Irgm1. To investigate the molecular basis for these defects, we conducted microarray expression profiling of Irgm1-deficient HSCs. Cell-cycle and IFN-response genes are up-regulated in Irgm1(-/-) HSCs, consistent with dysregulated IFN signaling. To test the hypothesis that Irgm1 normally down-regulates IFN signaling in HSCs, we generated Irgm1(-/-)Ifngr1(-/-) and Irgm1(-/-)Stat1(-/-) double-knockout animals. Strikingly, hyperproliferation, self-renewal, and autophagy defects in Irgm1(-/-) HSCs were normalized in double-knockout animals. These defects were also abolished in Irgm1(-/-)Irgm3(-/-) double-knockout animals, indicating that Irgm1 may regulate Irgm3 activity. Furthermore, the number of HSCs was reduced in aged Irgm1(-/-) animals, suggesting that negative feedback inhibition of IFN signaling by Irgm1 is necessary to prevent hyperproliferation and depletion of the stem cell compartment. Collectively, our results indicate that Irgm1 is a powerful negative regulator of IFN-dependent stimulation in HSCs, with an essential role in preserving HSC number and function. The deleterious effects of excessive IFN signaling may explain how hematologic abnormalities arise in patients with inflammatory conditions.


Asunto(s)
Proteínas de Unión al GTP/genética , Células Madre Hematopoyéticas/metabolismo , Receptores de Interferón/genética , Transducción de Señal/genética , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Trasplante de Médula Ósea , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Proteínas de Unión al GTP/deficiencia , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Inmunohistoquímica , Interferones/genética , Interferones/metabolismo , Interferones/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Interferón/deficiencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Receptor de Interferón gamma
15.
Exp Hematol ; 127: 8-13, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37647982

RESUMEN

Chronic inflammation, although subtle, puts the body in a constant state of alertness and is associated with many diseases, including cancer and cardiovascular diseases. It leads hematopoietic cells to produce and release proinflammatory cytokines, which trigger specific signaling pathways in hematopoietic stem cells (HSCs) that cause changes in proliferation, differentiation, and migration. This response is essential when HSCs are needed to produce specific blood cells to eliminate an intruder, such as a pathogenic virus, but mutant HSCs can use these proinflammatory signals to their advantage and accelerate the development of hematologic disease or malignancy. Understanding this complex process is vital for monitoring and controlling disease progression in patients. In the 2023 International Society for Experimental Hematology winter webinar, Dr. Eric Pietras (University of Colorado Anschutz Medical Campus, United States) and Dr. Katherine Y. King (Baylor College of Medicine, United States) gave a presentation on this topic, which is summarized in this review article.


Asunto(s)
Enfermedades Hematológicas , Células Madre Hematopoyéticas , Humanos , Células Madre Hematopoyéticas/metabolismo , Diferenciación Celular , Transducción de Señal , Enfermedades Hematológicas/metabolismo , Inflamación/patología
16.
Transplant Cell Ther ; 29(3): 165.e1-165.e7, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36592718

RESUMEN

Chimeric antigen receptor (CAR) T cell (CAR-T) therapy represents a revolutionary treatment for patients with relapsed/refractory hematologic malignancies. However, its use can result in significant toxicities, including cytokine release syndrome (CRS), a potentially life-threatening clinical syndrome resulting from the release of proinflammatory cytokines upon T cell activation. In addition, patients who develop CRS often experience prolonged cytopenias, and those with the most severe CRS also have the longest delays in full marrow recovery. Although an association between CRS and delayed bone marrow recovery has been established, the precise mechanism underlying this phenomenon remains unknown. This study was conducted to test our hypothesis that delayed bone marrow recovery following CAR-T therapy is caused by elevation of proinflammatory cytokines, leading to apoptosis and depletion of hematopoietic stem and progenitor cells (HSPCs). SCID-beige mice bearing intraperitoneal CD19+ Raji cell tumors were treated with injection of human CD19.28z CAR T cells. Bone marrow was then harvested for analysis by flow cytometry, and HSPCs were isolated for whole-transcriptome analysis by RNA sequencing. Complete blood counts and serum cytokine levels were measured as well. A second model was developed in which SCID-beige mice were treated with murine IFN-γ (mIFN-γ), murine IL-6 (mIL-6), or both. Bone marrow was harvested, and flow cytometry assays were conducted to evaluate the degree of apoptosis and proliferation on specific HSPC populations. SCID-beige mice bearing intraperitoneal Raji cell tumors that were treated with CAR-T therapy developed CRS, with elevations of several proinflammatory cytokines, including profound elevation of human IFN-γ. Gene set enrichment analysis of RNA sequencing data revealed that genes associated with apoptosis were significantly upregulated in HSPCs from mice that developed CRS. Endothelial protein C receptor (EPCR)-negative HSCs, a subset of HSCs that is poised for terminal differentiation, was found to be specifically decreased in mice that were treated with CAR T cells. Furthermore, HSPCs were found to have increased levels of apoptosis upon treatment with mIFN-γ and mIL-6, whereas short-term HSCs and multipotent progenitors exhibited increases in proliferation with mIFN-γ treatment alone. The results from this study provide evidence that the elevation of proinflammatory cytokines following CAR-T therapy impacts the bone marrow through a combined mechanism: pluripotent HSCs that are exposed to elevated levels of IFN-γ and IL-6 undergo increased cell death, while more committed progenitor cells become more proliferative in response to elevated IFN-γ. These combined effects lead to depleted stores of repopulating HSCs and ultimately cytopenias. © 2023 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc.


Asunto(s)
Enfermedades de la Médula Ósea , Inmunoterapia Adoptiva , Neoplasias , Receptores Quiméricos de Antígenos , Animales , Humanos , Ratones , Apoptosis , Médula Ósea/patología , Enfermedades de la Médula Ósea/metabolismo , Enfermedades de la Médula Ósea/patología , Citocinas/metabolismo , Células Madre Hematopoyéticas , Interleucina-6/metabolismo , Ratones SCID , Inmunoterapia Adoptiva/efectos adversos
17.
iScience ; 26(2): 106059, 2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36824275

RESUMEN

Basic leucine zipper ATF-like transcription factor 2 (BATF2), an interferon-activated immune response regulator, is a key factor responsible for myeloid differentiation and depletion of HSC during chronic infection. To delineate the mechanism of BATF2 function in HSCs, we assessed Batf2 KO mice during chronic infection and found that they produced less pro-inflammatory cytokines, less immune cell recruitment to the spleen, and impaired myeloid differentiation with better preservation of HSC capacity compared to WT. Co-IP analysis revealed that BATF2 forms a complex with JUN to amplify pro-inflammatory signaling pathways including CCL5 during infection. Blockade of CCL5 receptors phenocopied Batf2 KO differentiation defects, whereas treatment with recombinant CCL5 was sufficient to rescue IFNγ-induced myeloid differentiation and recruit more immune cells to the spleen in Batf2 KO mice. By revealing the mechanism of BATF2-induced myeloid differentiation of HSCs, these studies elucidate potential therapeutic strategies to boost immunity while preserving HSC function during chronic infection.

18.
iScience ; 26(9): 107596, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37664586

RESUMEN

Recent studies suggest that infection reprograms hematopoietic stem and progenitor cells (HSPCs) to enhance innate immune responses upon secondary infectious challenge, a process called "trained immunity." However, the specificity and cell types responsible for this response remain poorly defined. We established a model of trained immunity in mice in response to Mycobacterium avium infection. scRNA-seq analysis revealed that HSPCs activate interferon gamma-response genes heterogeneously upon primary challenge, while rare cell populations expand. Macrophages derived from trained HSPCs demonstrated enhanced bacterial killing and metabolism, and a single dose of recombinant interferon gamma exposure was sufficient to induce similar training. Mice transplanted with influenza-trained HSPCs displayed enhanced immunity against M. avium challenge and vice versa, demonstrating cross protection against antigenically distinct pathogens. Together, these results indicate that heterogeneous responses to infection by HSPCs can lead to long-term production of bone marrow derived macrophages with enhanced function and confer cross-protection against alternative pathogens.

19.
Exp Hematol ; 112-113: 35-43, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35768035

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are responsible for the production of all immune and blood cells in both steady state and emergency settings. The rates at which HSPCs divide and differentiate vary widely in accordance with both cell intrinsic and cell extrinsic factors. However, the kinetics of these events remain poorly understood. In prior work, we determined that the inflammatory cytokine interferon-γ (IFN-γ) induces HSPC division and differentiation. Here, we report that a subset of hematopoietic stem cells (HSCs) that express Fgd5 do not divide or differentiate in response to IFN-γ. This suggests that FGD5 marks a subset of HSCs that remains unperturbed during emergency hematopoiesis and is potentially a mechanism of preservation of the HSC compartment.


Asunto(s)
Células Madre Hematopoyéticas , Interferón gamma , Diferenciación Celular , Citocinas , Hematopoyesis/fisiología , Interferón gamma/farmacología
20.
Elife ; 112022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35166205

RESUMEN

New therapeutic strategies to reduce sepsis-related mortality are urgently needed, as sepsis accounts for one in five deaths worldwide. Since hematopoietic stem and progenitor cells (HSPCs) are responsible for producing blood and immune cells, including in response to immunological stress, we explored their potential for treating sepsis. In a mouse model of Group A Streptococcus (GAS)-induced sepsis, severe immunological stress was associated with significant depletion of bone marrow HSPCs and mortality within approximately 5-7 days. We hypothesized that the inflammatory environment of GAS infection drives rapid HSPC differentiation and depletion that can be rescued by infusion of donor HSPCs. Indeed, infusion of 10,000 naïve HSPCs into GAS-infected mice resulted in rapid myelopoiesis and a 50-60% increase in overall survival. Surprisingly, mice receiving donor HSPCs displayed a similar pathogen load compared to untreated mice. Flow cytometric analysis revealed a significantly increased number of myeloid-derived suppressor cells in HSPC-infused mice, which correlated with reduced inflammatory cytokine levels and restored HSPC levels. These findings suggest that HSPCs play an essential immunomodulatory role that may translate into new therapeutic strategies for sepsis.


Asunto(s)
Diferenciación Celular/inmunología , Células Madre Hematopoyéticas/inmunología , Inmunomodulación , Sepsis/inmunología , Células Madre/inmunología , Infecciones Estreptocócicas/sangre , Animales , Citocinas/inmunología , Femenino , Trasplante de Células Madre Hematopoyéticas/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Sepsis/terapia , Trasplante de Células Madre/métodos , Infecciones Estreptocócicas/inmunología , Streptococcus/inmunología , Streptococcus/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA