Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cells Dev ; 174: 203844, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37100116

RESUMO

Just over one decade ago, it was discovered that hematopoietic stem cells (HSCs) could directly respond to inflammatory cytokines by mounting a proliferative response thought to mediate the emergency production of mature blood cells. In the intervening years, we have gained mechanistic insight into this so-called activation process and have started to learn such a response may come at a cost in terms of ultimately resulting in HSC exhaustion and hematologic dysfunction. In this review article, we report the progress we have made in understanding the interplay between infection, inflammation and HSCs during the funding period of the Collaborative Research Center 873 "Maintenance and Differentiation of Stem Cells in Development and Disease", and place this work within the context of recent output by others working within this field.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas , Humanos , Hematopoese/fisiologia , Diferenciação Celular , Citocinas , Inflamação
2.
Cell Rep ; 41(1): 111447, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36198277

RESUMO

Respiratory tract infections are among the deadliest communicable diseases worldwide. Severe cases of viral lung infections are often associated with a cytokine storm and alternating platelet numbers. We report that hematopoietic stem and progenitor cells (HSPCs) sense a non-systemic influenza A virus (IAV) infection via inflammatory cytokines. Irrespective of antiviral treatment or vaccination, at a certain threshold of IAV titer in the lung, CD41-positive hematopoietic stem cells (HSCs) enter the cell cycle while endothelial protein C receptor-positive CD41-negative HSCs remain quiescent. Active CD41-positive HSCs represent the source of megakaryocytes, while their multi-lineage reconstitution potential is reduced. This emergency megakaryopoiesis is thrombopoietin independent and attenuated in IAV-infected interleukin-1 receptor-deficient mice. Newly produced platelets during IAV infection are immature and hyper-reactive. After viral clearance, HSC quiescence is re-established. Our study reveals that non-systemic viral respiratory infection has an acute impact on HSCs via inflammatory cytokines to counteract IAV-induced thrombocytopenia.


Assuntos
Vírus da Influenza A , Influenza Humana , Animais , Antivirais/metabolismo , Citocinas/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Hematopoese , Humanos , Influenza Humana/metabolismo , Megacariócitos/metabolismo , Camundongos , Receptores de Interleucina-1/metabolismo , Trombopoetina/metabolismo
3.
Cell Stem Cell ; 29(8): 1273-1284.e8, 2022 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-35858618

RESUMO

Hematopoietic stem cells (HSCs) mediate regeneration of the hematopoietic system following injury, such as following infection or inflammation. These challenges impair HSC function, but whether this functional impairment extends beyond the duration of inflammatory exposure is unknown. Unexpectedly, we observed an irreversible depletion of functional HSCs following challenge with inflammation or bacterial infection, with no evidence of any recovery up to 1 year afterward. HSCs from challenged mice demonstrated multiple cellular and molecular features of accelerated aging and developed clinically relevant blood and bone marrow phenotypes not normally observed in aged laboratory mice but commonly seen in elderly humans. In vivo HSC self-renewal divisions were absent or extremely rare during both challenge and recovery periods. The progressive, irreversible attrition of HSC function demonstrates that temporally discrete inflammatory events elicit a cumulative inhibitory effect on HSCs. This work positions early/mid-life inflammation as a mediator of lifelong defects in tissue maintenance and regeneration.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas , Idoso , Envelhecimento , Animais , Medula Óssea , Humanos , Inflamação , Camundongos
4.
Cell Stem Cell ; 29(5): 760-775.e10, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35523139

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) are responsible for the production of blood and immune cells. Throughout life, HSPCs acquire oncogenic aberrations that can cause hematological cancers. Although molecular programs maintaining stem cell integrity have been identified, safety mechanisms eliminating malignant HSPCs from the stem cell pool remain poorly characterized. Here, we show that HSPCs constitutively present antigens via major histocompatibility complex class II. The presentation of immunogenic antigens, as occurring during malignant transformation, triggers bidirectional interactions between HSPCs and antigen-specific CD4+ T cells, causing stem cell proliferation, differentiation, and specific exhaustion of aberrant HSPCs. This immunosurveillance mechanism effectively eliminates transformed HSPCs from the hematopoietic system, thereby preventing leukemia onset. Together, our data reveal a bidirectional interaction between HSPCs and CD4+ T cells, demonstrating that HSPCs are not only passive receivers of immunological signals but also actively engage in adaptive immune responses to safeguard the integrity of the stem cell pool.


Assuntos
Apresentação de Antígeno , Células-Tronco Hematopoéticas , Diferenciação Celular , Linfócitos T
5.
Exp Hematol ; 96: 1-12, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33571568

RESUMO

Interferons are an ancient and well-conserved group of inflammatory cytokines most famous for their role in viral immunity. A decade ago, we discovered that interferons also play an important role in the biology of hematopoietic stem cells (HSCs), which are responsible for lifelong blood production. Though we have learned a great deal about the role of interferons on HSC quiescence, differentiation, and self-renewal, there remains some controversy regarding how interferons impact these stem cells, with differing conclusions depending on experimental models and clinical context. Here, we review the contradictory roles of Type 1 and 2 interferons in hematopoiesis. Specifically, we highlight the roles of interferons in embryonic and adult hematopoiesis, along with short-term and long-term adaptive and maladaptive responses to inflammation. We discuss experimental challenges in the study of these powerful yet short-lived cytokines and strategies to address those challenges. We further review the contribution by interferons to disease states including bone marrow failure and aplastic anemia as well as their therapeutic use to treat myeloproliferative neoplasms and viral infections, including SARS-CoV2. Understanding the opposing effects of interferons on hematopoiesis will elucidate immune responses and bone marrow failure syndromes, and future therapeutic approaches for patients undergoing HSC transplantation or fighting infectious diseases and cancer.


Assuntos
Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Fatores Imunológicos/uso terapêutico , Interferons/uso terapêutico , Animais , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/imunologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Fatores Imunológicos/imunologia , Interferons/imunologia
6.
Cell Stem Cell ; 26(5): 611-612, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32386549

RESUMO

Immune memory was thought to be unique to cells of the adaptive immune system. In this issue of Cell Stem Cell, de Laval et al. (2020) describe persistent epigenetic modifications in hematopoietic stem cells following an inflammatory insult with LPS as a mechanism by which immune memory may be established.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT , Células-Tronco Hematopoéticas , Epigênese Genética , Humanos , Sistema Imunitário , Inflamação/genética
7.
Nat Commun ; 11(1): 1494, 2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-32198421

RESUMO

Metastatic colonization relies on interactions between disseminated cancer cells and the microenvironment in secondary organs. Here, we show that disseminated breast cancer cells evoke phenotypic changes in lung fibroblasts, forming a supportive metastatic niche. Colonization of the lungs confers an inflammatory phenotype in metastasis-associated fibroblasts. Specifically, IL-1α and IL-1ß secreted by breast cancer cells induce CXCL9 and CXCL10 production in lung fibroblasts via NF-κB signaling, fueling the growth of lung metastases. Notably, we find that the chemokine receptor CXCR3, that binds CXCL9/10, is specifically expressed in a small subset of breast cancer cells, which exhibits tumor-initiating ability when co-transplanted with fibroblasts and has high JNK signaling that drives IL-1α/ß expression. Importantly, disruption of the intercellular JNK-IL-1-CXCL9/10-CXCR3 axis reduces metastatic colonization in xenograft and syngeneic mouse models. These data mechanistically demonstrate an essential role for the molecular crosstalk between breast cancer cells and their fibroblast niche in the progression of metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Fibroblastos/metabolismo , Neoplasias Pulmonares/metabolismo , Pulmão/metabolismo , Metástase Neoplásica , Microambiente Tumoral/fisiologia , Animais , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/metabolismo , Feminino , Fibroblastos/patologia , Técnicas de Silenciamento de Genes , Humanos , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Receptores CXCR3/metabolismo , Transdução de Sinais , Transcriptoma , Transplante Heterólogo
8.
Cell Rep ; 19(11): 2345-2356, 2017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28614719

RESUMO

Quiescent long-term hematopoietic stem cells (LT-HSCs) are efficiently activated by type I interferon (IFN-I). However, this effect remains poorly investigated in the context of IFN-I-inducing virus infections. Here we report that both vesicular stomatitis virus (VSV) and murine cytomegalovirus (MCMV) infection induce LT-HSC activation that substantially differs from the effects triggered upon injection of synthetic IFN-I-inducing agents. In both infections, inflammatory responses had to exceed local thresholds within the bone marrow to confer LT-HSC cell cycle entry, and IFN-I receptor triggering was not critical for this activation. After resolution of acute MCMV infection, LT-HSCs returned to phenotypic quiescence. However, non-acute MCMV infection induced a sustained inflammatory milieu within the bone marrow that was associated with long-lasting impairment of LT-HSC function. In conclusion, our results show that systemic virus infections fundamentally affect LT-HSCs and that also non-acute inflammatory stimuli in bone marrow donors can affect the reconstitution potential of bone marrow transplants.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Infecções/virologia , Animais , Ciclo Celular , Proliferação de Células , Células-Tronco Hematopoéticas/citologia , Camundongos , Transdução de Sinais
9.
Nat Cell Biol ; 19(4): 271-281, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28319093

RESUMO

Blood formation is believed to occur through stepwise progression of haematopoietic stem cells (HSCs) following a tree-like hierarchy of oligo-, bi- and unipotent progenitors. However, this model is based on the analysis of predefined flow-sorted cell populations. Here we integrated flow cytometric, transcriptomic and functional data at single-cell resolution to quantitatively map early differentiation of human HSCs towards lineage commitment. During homeostasis, individual HSCs gradually acquire lineage biases along multiple directions without passing through discrete hierarchically organized progenitor populations. Instead, unilineage-restricted cells emerge directly from a 'continuum of low-primed undifferentiated haematopoietic stem and progenitor cells' (CLOUD-HSPCs). Distinct gene expression modules operate in a combinatorial manner to control stemness, early lineage priming and the subsequent progression into all major branches of haematopoiesis. These data reveal a continuous landscape of human steady-state haematopoiesis downstream of HSCs and provide a basis for the understanding of haematopoietic malignancies.


Assuntos
Linhagem da Célula , Células-Tronco Hematopoéticas/citologia , Adulto , Animais , Antígenos CD/metabolismo , Linhagem da Célula/genética , Proliferação de Células/genética , Feminino , Redes Reguladoras de Genes , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Masculino , Camundongos , Modelos Biológicos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Transcriptoma/genética
10.
Haematologica ; 102(3): 445-453, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27742772

RESUMO

In the bone marrow, endothelial cells are a major component of the hematopoietic stem cell vascular niche and are a first line of defense against inflammatory stress and infection. The primary response of an organism to infection involves the synthesis of immune-modulatory cytokines, including interferon alpha. In the bone marrow, interferon alpha induces rapid cell cycle entry of hematopoietic stem cells in vivo However, the effect of interferon alpha on bone marrow endothelial cells has not been described. Here, we demonstrate that acute interferon alpha treatment leads to rapid stimulation of bone marrow endothelial cells in vivo, resulting in increased bone marrow vascularity and vascular leakage. We find that activation of bone marrow endothelial cells involves the expression of key inflammatory and endothelial cell-stimulatory markers. This interferon alpha-mediated activation of bone marrow endothelial cells is dependent in part on vascular endothelial growth factor signaling in bone marrow hematopoietic cell types, including hematopoietic stem cells. Thus, this implies a role for hematopoietic stem cells in remodeling of the bone marrow niche in vivo following inflammatory stress. These data increase our current understanding of the relationship between hematopoietic stem cells and the bone marrow niche under inflammatory stress and also clarify the response of bone marrow niche endothelial cells to acute interferon alpha treatment in vivo.


Assuntos
Medula Óssea/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Interferon-alfa/metabolismo , Nicho de Células-Tronco , Animais , Biomarcadores , Medula Óssea/irrigação sanguínea , Células da Medula Óssea/metabolismo , Permeabilidade Capilar , Proliferação de Células , Microambiente Celular , Feminino , Hematopoese , Células-Tronco Hematopoéticas/citologia , Imunofenotipagem , Interferon-alfa/genética , Interferon-alfa/farmacologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Neovascularização Fisiológica , Fenótipo , Poli I-C/farmacologia , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico , Fator A de Crescimento do Endotélio Vascular/biossíntese
11.
Cell Stem Cell ; 19(2): 143-144, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27494667

RESUMO

Changes in cellular metabolism drive hematopoietic stem cell (HSC) behavior during homeostasis, although whether they control HSC behavior during stress conditions is unclear. In this issue of Cell Stem Cell, Karigane et al. (2016) identify a p38α-dependent pathway that alters purine metabolism in HSCs during stress hematopoiesis, promoting hematopoietic recovery.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas , Homeostase , Purinas
13.
Cell ; 164(4): 668-80, 2016 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-26871632

RESUMO

Mouse embryonic stem cells (ESCs) are maintained in a naive ground state of pluripotency in the presence of MEK and GSK3 inhibitors. Here, we show that ground-state ESCs express low Myc levels. Deletion of both c-myc and N-myc (dKO) or pharmacological inhibition of Myc activity strongly decreases transcription, splicing, and protein synthesis, leading to proliferation arrest. This process is reversible and occurs without affecting pluripotency, suggesting that Myc-depleted stem cells enter a state of dormancy similar to embryonic diapause. Indeed, c-Myc is depleted in diapaused blastocysts, and the differential expression signatures of dKO ESCs and diapaused epiblasts are remarkably similar. Following Myc inhibition, pre-implantation blastocysts enter biosynthetic dormancy but can progress through their normal developmental program after transfer into pseudo-pregnant recipients. Our study shows that Myc controls the biosynthetic machinery of stem cells without affecting their potency, thus regulating their entry and exit from the dormant state.


Assuntos
Células-Tronco Embrionárias/citologia , Genes myc , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Blastocisto/metabolismo , Proliferação de Células , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/metabolismo , Feminino , Técnicas de Inativação de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL
14.
J Allergy Clin Immunol ; 138(1): 219-228.e9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26853280

RESUMO

BACKGROUND: Defects in phagocytic nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) function cause chronic granulomatous disease (CGD), a primary immunodeficiency characterized by dysfunctional microbicidal activity and chronic inflammation. OBJECTIVE: We sought to study the effect of chronic inflammation on the hematopoietic compartment in patients and mice with X-linked chronic granulomatous disease (X-CGD). METHODS: We used immunostaining and functional analyses to study the hematopoietic compartment in patients with CGD. RESULTS: An analysis of bone marrow cells from patients and mice with X-CGD revealed a dysregulated hematopoiesis characterized by increased numbers of hematopoietic progenitor cells (HPCs) at the expense of repopulating hematopoietic stem cells (HSCs). In patients with X-CGD, there was a clear reduction in the proportion of HSCs in bone marrow and peripheral blood, and they were also more rapidly exhausted after in vitro culture. In mice with X-CGD, increased cycling of HSCs, expansion of HPCs, and impaired long-term engraftment capacity were found to be associated with high concentrations of proinflammatory cytokines, including IL-1ß. Treatment of wild-type mice with IL-1ß induced enhanced cell-cycle entry of HSCs, expansion of HPCs, and defects in long-term engraftment, mimicking the effects observed in mice with X-CGD. Inhibition of cytokine signaling in mice with X-CGD reduced HPC numbers but had only minor effects on the repopulating ability of HSCs. CONCLUSIONS: Persistent chronic inflammation in patients with CGD is associated with hematopoietic proliferative stress, leading to a decrease in the functional activity of HSCs. Our observations have clinical implications for the development of successful autologous cell therapy approaches.


Assuntos
Doença Granulomatosa Crônica/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Adolescente , Adulto , Animais , Biomarcadores , Estudos de Casos e Controles , Contagem de Células , Diferenciação Celular , Criança , Pré-Escolar , Ensaio de Unidades Formadoras de Colônias , Citocinas/metabolismo , Citocinas/farmacologia , Modelos Animais de Doenças , Sobrevivência de Enxerto , Doença Granulomatosa Crônica/etiologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Imunofenotipagem , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Fenótipo , Transdução de Sinais , Adulto Jovem
15.
Cell Stem Cell ; 17(4): 422-34, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26299573

RESUMO

Infections are associated with extensive platelet consumption, representing a high risk for health. However, the mechanism coordinating the rapid regeneration of the platelet pool during such stress conditions remains unclear. Here, we report that the phenotypic hematopoietic stem cell (HSC) compartment contains stem-like megakaryocyte-committed progenitors (SL-MkPs), a cell population that shares many features with multipotent HSCs and serves as a lineage-restricted emergency pool for inflammatory insults. During homeostasis, SL-MkPs are maintained in a primed but quiescent state, thus contributing little to steady-state megakaryopoiesis. Even though lineage-specific megakaryocyte transcripts are expressed, protein synthesis is suppressed. In response to acute inflammation, SL-MkPs become activated, resulting in megakaryocyte protein production from pre-existing transcripts and a maturation of SL-MkPs and other megakaryocyte progenitors. This results in an efficient replenishment of platelets that are lost during inflammatory insult. Thus, our study reveals an emergency machinery that counteracts life-threatening platelet depletions during acute inflammation.


Assuntos
Plaquetas/patologia , Inflamação/patologia , Células Progenitoras de Megacariócitos/patologia , Trombopoese , Animais , Plaquetas/fisiologia , Linhagem da Célula , Proliferação de Células , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/fisiologia , Células Progenitoras de Megacariócitos/fisiologia , Camundongos
16.
Nature ; 520(7548): 549-52, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25707806

RESUMO

Haematopoietic stem cells (HSCs) are responsible for the lifelong production of blood cells. The accumulation of DNA damage in HSCs is a hallmark of ageing and is probably a major contributing factor in age-related tissue degeneration and malignant transformation. A number of accelerated ageing syndromes are associated with defective DNA repair and genomic instability, including the most common inherited bone marrow failure syndrome, Fanconi anaemia. However, the physiological source of DNA damage in HSCs from both normal and diseased individuals remains unclear. Here we show in mice that DNA damage is a direct consequence of inducing HSCs to exit their homeostatic quiescent state in response to conditions that model physiological stress, such as infection or chronic blood loss. Repeated activation of HSCs out of their dormant state provoked the attrition of normal HSCs and, in the case of mice with a non-functional Fanconi anaemia DNA repair pathway, led to a complete collapse of the haematopoietic system, which phenocopied the highly penetrant bone marrow failure seen in Fanconi anaemia patients. Our findings establish a novel link between physiological stress and DNA damage in normal HSCs and provide a mechanistic explanation for the universal accumulation of DNA damage in HSCs during ageing and the accelerated failure of the haematopoietic system in Fanconi anaemia patients.


Assuntos
Ciclo Celular , Dano ao DNA , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Animais , Medula Óssea/patologia , Morte Celular , Proliferação de Células , Anemia de Fanconi/metabolismo , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Estresse Fisiológico
17.
Cell Stem Cell ; 15(4): 507-522, 2014 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-25158935

RESUMO

In this study, we present integrated quantitative proteome, transcriptome, and methylome analyses of hematopoietic stem cells (HSCs) and four multipotent progenitor (MPP) populations. From the characterization of more than 6,000 proteins, 27,000 transcripts, and 15,000 differentially methylated regions (DMRs), we identified coordinated changes associated with early differentiation steps. DMRs show continuous gain or loss of methylation during differentiation, and the overall change in DNA methylation correlates inversely with gene expression at key loci. Our data reveal the differential expression landscape of 493 transcription factors and 682 lncRNAs and highlight specific expression clusters operating in HSCs. We also found an unexpectedly dynamic pattern of transcript isoform regulation, suggesting a critical regulatory role during HSC differentiation, and a cell cycle/DNA repair signature associated with multipotency in MPP2 cells. This study provides a comprehensive genome-wide resource for the functional exploration of molecular, cellular, and epigenetic regulation at the top of the hematopoietic hierarchy.


Assuntos
Metilação de DNA/genética , Redes Reguladoras de Genes , Células-Tronco Hematopoéticas/metabolismo , Proteoma/metabolismo , Transcriptoma/genética , Adulto , Diferenciação Celular/genética , Linhagem da Célula/genética , Análise por Conglomerados , Epigênese Genética , Perfilação da Expressão Gênica , Genoma Humano/genética , Impressão Genômica , Células-Tronco Hematopoéticas/citologia , Humanos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Longo não Codificante/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
18.
J Exp Med ; 211(5): 769-79, 2014 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-24752302

RESUMO

The serine protease granzyme B (GzmB) is stored in the granules of cytotoxic T and NK cells and facilitates immune-mediated destruction of virus-infected cells. In this study, we use genetic tools to report novel roles for GzmB as an important regulator of hematopoietic stem cell (HSC) function in response to stress. HSCs lacking the GzmB gene show improved bone marrow (BM) reconstitution associated with increased HSC proliferation and mitochondrial activity. In addition, recipients deficient in GzmB support superior engraftment of wild-type HSCs compared with hosts with normal BM niches. Stimulation of mice with lipopolysaccharide strongly induced GzmB protein expression in HSCs, which was mediated by the TLR4-TRIF-p65 NF-κB pathway. This is associated with increased cell death and GzmB secretion into the BM environment, suggesting an extracellular role of GzmB in modulating HSC niches. Moreover, treatment with the chemotherapeutic agent 5-fluorouracil (5-FU) also induces GzmB production in HSCs. In this situation GzmB is not secreted, but instead causes cell-autonomous apoptosis. Accordingly, GzmB-deficient mice are more resistant to serial 5-FU treatments. Collectively, these results identify GzmB as a negative regulator of HSC function that is induced by stress and chemotherapy in both HSCs and their niches. Blockade of GzmB production may help to improve hematopoiesis in various situations of BM stress.


Assuntos
Regulação da Expressão Gênica/imunologia , Granzimas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Transdução de Sinais/imunologia , Estresse Fisiológico/fisiologia , Animais , Apoptose/efeitos dos fármacos , Medula Óssea/fisiologia , Proliferação de Células/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Primers do DNA/genética , Tratamento Farmacológico , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Fluoruracila/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Granzimas/deficiência , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Imuno-Histoquímica , Lipopolissacarídeos/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Reação em Cadeia da Polimerase em Tempo Real
19.
Ann N Y Acad Sci ; 1310: 51-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24611873

RESUMO

The immune response to infection is a rapid and multifaceted process. Infection affects homeostasis within the hematopoietic stem cell (HSC) niche, as lost immune cells must be replaced by HSCs. During the immune response, interferon is produced. Surprisingly, HSCs respond directly to interferon, entering the cell cycle from even the most dormant state. The complex response of both the HSCs and the niche to infection is a unique platform on which to consider HSC-niche interactions. Here, we comment on the contribution of the immune system to the niche and on the direct and indirect effect that infection has on HSCs in the niche.


Assuntos
Células-Tronco Hematopoéticas/imunologia , Imunidade Inata/fisiologia , Infecções/imunologia , Nicho de Células-Tronco/imunologia , Animais , Humanos , Camundongos
20.
Nature ; 458(7240): 904-8, 2009 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-19212321

RESUMO

Maintenance of the blood system is dependent on dormant haematopoietic stem cells (HSCs) with long-term self-renewal capacity. After injury these cells are induced to proliferate to quickly re-establish homeostasis. The signalling molecules promoting the exit of HSCs out of the dormant stage remain largely unknown. Here we show that in response to treatment of mice with interferon-alpha (IFNalpha), HSCs efficiently exit G(0) and enter an active cell cycle. HSCs respond to IFNalpha treatment by the increased phosphorylation of STAT1 and PKB/Akt (also known as AKT1), the expression of IFNalpha target genes, and the upregulation of stem cell antigen-1 (Sca-1, also known as LY6A). HSCs lacking the IFNalpha/beta receptor (IFNAR), STAT1 (ref. 3) or Sca-1 (ref. 4) are insensitive to IFNalpha stimulation, demonstrating that STAT1 and Sca-1 mediate IFNalpha-induced HSC proliferation. Although dormant HSCs are resistant to the anti-proliferative chemotherapeutic agent 5-fluoro-uracil, HSCs pre-treated (primed) with IFNalpha and thus induced to proliferate are efficiently eliminated by 5-fluoro-uracil exposure in vivo. Conversely, HSCs chronically activated by IFNalpha are functionally compromised and are rapidly out-competed by non-activatable Ifnar(-/-) cells in competitive repopulation assays. Whereas chronic activation of the IFNalpha pathway in HSCs impairs their function, acute IFNalpha treatment promotes the proliferation of dormant HSCs in vivo. These data may help to clarify the so far unexplained clinical effects of IFNalpha on leukaemic cells, and raise the possibility for new applications of type I interferons to target cancer stem cells.


Assuntos
Ciclo Celular/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Interferon-alfa/farmacologia , Animais , Antígenos Ly/metabolismo , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Fluoruracila/farmacologia , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/metabolismo , Fator de Transcrição STAT1/deficiência , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA