Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.490
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(12): 3120-3140.e29, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38714197

RESUMEN

Non-hematopoietic cells are essential contributors to hematopoiesis. However, heterogeneity and spatial organization of these cells in human bone marrow remain largely uncharacterized. We used single-cell RNA sequencing (scRNA-seq) to profile 29,325 non-hematopoietic cells and discovered nine transcriptionally distinct subtypes. We simultaneously profiled 53,417 hematopoietic cells and predicted their interactions with non-hematopoietic subsets. We employed co-detection by indexing (CODEX) to spatially profile over 1.2 million cells. We integrated scRNA-seq and CODEX data to link predicted cellular signaling with spatial proximity. Our analysis revealed a hyperoxygenated arterio-endosteal neighborhood for early myelopoiesis, and an adipocytic localization for early hematopoietic stem and progenitor cells (HSPCs). We used our CODEX atlas to annotate new images and uncovered mesenchymal stromal cell (MSC) expansion and spatial neighborhoods co-enriched for leukemic blasts and MSCs in acute myeloid leukemia (AML) patient samples. This spatially resolved, multiomic atlas of human bone marrow provides a reference for investigation of cellular interactions that drive hematopoiesis.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Células Madre Mesenquimatosas , Proteómica , Análisis de la Célula Individual , Transcriptoma , Humanos , Análisis de la Célula Individual/métodos , Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Proteómica/métodos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Hematopoyesis , Nicho de Células Madre , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/citología
2.
Cell ; 182(2): 372-387.e14, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32610084

RESUMEN

Acute psychological stress has long been known to decrease host fitness to inflammation in a wide variety of diseases, but how this occurs is incompletely understood. Using mouse models, we show that interleukin-6 (IL-6) is the dominant cytokine inducible upon acute stress alone. Stress-inducible IL-6 is produced from brown adipocytes in a beta-3-adrenergic-receptor-dependent fashion. During stress, endocrine IL-6 is the required instructive signal for mediating hyperglycemia through hepatic gluconeogenesis, which is necessary for anticipating and fueling "fight or flight" responses. This adaptation comes at the cost of enhancing mortality to a subsequent inflammatory challenge. These findings provide a mechanistic understanding of the ontogeny and adaptive purpose of IL-6 as a bona fide stress hormone coordinating systemic immunometabolic reprogramming. This brain-brown fat-liver axis might provide new insights into brown adipose tissue as a stress-responsive endocrine organ and mechanistic insight into targeting this axis in the treatment of inflammatory and neuropsychiatric diseases.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Interleucina-6/metabolismo , Estrés Psicológico , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Encéfalo/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Gluconeogénesis , Hiperglucemia/metabolismo , Hiperglucemia/patología , Interleucina-6/sangre , Interleucina-6/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Adrenérgicos beta 3/metabolismo , Receptores de Interleucina-6/metabolismo , Proteína Desacopladora 1/deficiencia , Proteína Desacopladora 1/genética
3.
Annu Rev Immunol ; 30: 1-22, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22136168

RESUMEN

A properly functioning adaptive immune system signifies the best features of life. It is diverse beyond compare, tolerant without fail, and capable of behaving appropriately with a myriad of infections and other challenges. Dendritic cells are required to explain how this remarkable system is energized and directed. I frame this article in terms of the major decisions that my colleagues and I have made in dendritic cell science and some of the guiding themes at the time the decisions were made. As a result of progress worldwide, there is now evidence of a central role for dendritic cells in initiating antigen-specific immunity and tolerance. The in vivo distribution and development of a previously unrecognized white cell lineage is better understood, as is the importance of dendritic cell maturation to link innate and adaptive immunity in response to many stimuli. Our current focus is on antigen uptake receptors on dendritic cells. These receptors enable experiments involving selective targeting of antigens in situ and new approaches to vaccine design in preclinical and clinical systems.


Asunto(s)
Alergia e Inmunología/tendencias , Células Dendríticas/inmunología , Alergia e Inmunología/historia , Animales , Antígenos CD/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Tolerancia Inmunológica/inmunología , Inmunidad Celular/inmunología , Lectinas Tipo C/metabolismo , Antígenos de Histocompatibilidad Menor , Modelos Animales , Receptores de Superficie Celular/metabolismo , Vacunas/inmunología
4.
Cell ; 176(6): 1265-1281.e24, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30827681

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous disease that resides within a complex microenvironment, complicating efforts to understand how different cell types contribute to disease progression. We combined single-cell RNA sequencing and genotyping to profile 38,410 cells from 40 bone marrow aspirates, including 16 AML patients and five healthy donors. We then applied a machine learning classifier to distinguish a spectrum of malignant cell types whose abundances varied between patients and between subclones in the same tumor. Cell type compositions correlated with prototypic genetic lesions, including an association of FLT3-ITD with abundant progenitor-like cells. Primitive AML cells exhibited dysregulated transcriptional programs with co-expression of stemness and myeloid priming genes and had prognostic significance. Differentiated monocyte-like AML cells expressed diverse immunomodulatory genes and suppressed T cell activity in vitro. In conclusion, we provide single-cell technologies and an atlas of AML cell states, regulators, and markers with implications for precision medicine and immune therapies. VIDEO ABSTRACT.


Asunto(s)
Leucemia Mieloide Aguda/genética , Transcriptoma/genética , Adulto , Secuencia de Bases/genética , Médula Ósea , Células de la Médula Ósea/citología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Genotipo , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/fisiopatología , Aprendizaje Automático , Masculino , Persona de Mediana Edad , Mutación , Pronóstico , ARN , Transducción de Señal , Análisis de la Célula Individual/métodos , Microambiente Tumoral , Secuenciación del Exoma/métodos
5.
Cell ; 178(5): 1102-1114.e17, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442403

RESUMEN

Caloric restriction is known to improve inflammatory and autoimmune diseases. However, the mechanisms by which reduced caloric intake modulates inflammation are poorly understood. Here we show that short-term fasting reduced monocyte metabolic and inflammatory activity and drastically reduced the number of circulating monocytes. Regulation of peripheral monocyte numbers was dependent on dietary glucose and protein levels. Specifically, we found that activation of the low-energy sensor 5'-AMP-activated protein kinase (AMPK) in hepatocytes and suppression of systemic CCL2 production by peroxisome proliferator-activator receptor alpha (PPARα) reduced monocyte mobilization from the bone marrow. Importantly, we show that fasting improves chronic inflammatory diseases without compromising monocyte emergency mobilization during acute infectious inflammation and tissue repair. These results reveal that caloric intake and liver energy sensors dictate the blood and tissue immune tone and link dietary habits to inflammatory disease outcome.


Asunto(s)
Restricción Calórica , Monocitos/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Adulto , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Quimiocina CCL2/deficiencia , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Femenino , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , PPAR alfa/deficiencia , PPAR alfa/genética , PPAR alfa/metabolismo
6.
Cell ; 173(7): 1796-1809.e17, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29779944

RESUMEN

Non-coding genetic variation is a major driver of phenotypic diversity and allows the investigation of mechanisms that control gene expression. Here, we systematically investigated the effects of >50 million variations from five strains of mice on mRNA, nascent transcription, transcription start sites, and transcription factor binding in resting and activated macrophages. We observed substantial differences associated with distinct molecular pathways. Evaluating genetic variation provided evidence for roles of ∼100 TFs in shaping lineage-determining factor binding. Unexpectedly, a substantial fraction of strain-specific factor binding could not be explained by local mutations. Integration of genomic features with chromatin interaction data provided evidence for hundreds of connected cis-regulatory domains associated with differences in transcription factor binding and gene expression. This system and the >250 datasets establish a substantial new resource for investigation of how genetic variation affects cellular phenotypes.


Asunto(s)
Variación Genética , Macrófagos/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Células de la Médula Ósea/citología , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Análisis por Conglomerados , Elementos de Facilitación Genéticos/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética
7.
Nat Immunol ; 20(7): 852-864, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31213723

RESUMEN

Dendritic cells (DC) are currently classified as conventional DCs (cDCs) and plasmacytoid DCs (pDCs). Through a combination of single-cell transcriptomic analysis, mass cytometry, in vivo fate mapping and in vitro clonal assays, here we show that, at the single-cell level, the priming of mouse hematopoietic progenitor cells toward the pDC lineage occurs at the common lymphoid progenitor stage, indicative of early divergence of the pDC and cDC lineages. We found the transcriptional signature of a pDC precursor stage, defined here, in the IL-7Rα+ common lymphoid progenitor population and identified Ly6D, IL-7Rα, CD81 and CD2 as key markers of pDC differentiation, which distinguish pDC precursors from cDC precursors. In conclusion, pDCs developed in the bone marrow from a Ly6DhiCD2hi lymphoid progenitor cell and differentiated independently of the myeloid cDC lineage.


Asunto(s)
Antígenos Ly/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Citometría de Flujo , Proteínas Ligadas a GPI/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Ratones , Transcriptoma
8.
Cell ; 167(5): 1296-1309.e10, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27839867

RESUMEN

The ability of cells to count and remember their divisions could underlie many alterations that occur during development, aging, and disease. We tracked the cumulative divisional history of slow-cycling hematopoietic stem cells (HSCs) throughout adult life. This revealed a fraction of rarely dividing HSCs that contained all the long-term HSC (LT-HSC) activity within the aging HSC compartment. During adult life, this population asynchronously completes four traceable symmetric self-renewal divisions to expand its size before entering a state of dormancy. We show that the mechanism of expansion involves progressively lengthening periods between cell divisions, with long-term regenerative potential lost upon a fifth division. Our data also show that age-related phenotypic changes within the HSC compartment are divisional history dependent. These results suggest that HSCs accumulate discrete memory stages over their divisional history and provide evidence for the role of cellular memory in HSC aging.


Asunto(s)
Envejecimiento/patología , Células de la Médula Ósea/citología , Células Madre Hematopoyéticas/citología , Animales , Trasplante de Médula Ósea , Ciclo Celular , División Celular , Ratones , Ratones Endogámicos C57BL , Glicoproteína IIb de Membrana Plaquetaria/metabolismo
9.
Nature ; 626(8001): 1102-1107, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38355795

RESUMEN

Plasma cells produce large quantities of antibodies and so play essential roles in immune protection1. Plasma cells, including a long-lived subset, reside in the bone marrow where they depend on poorly defined microenvironment-linked survival signals1. We show that bone marrow plasma cells use the ligand-gated purinergic ion channel P2RX4 to sense extracellular ATP released by bone marrow osteoblasts through the gap-junction protein pannexin 3 (PANX3). Mutation of Panx3 or P2rx4 each caused decreased serum antibodies and selective loss of bone marrow plasma cells. Compared to their wild-type counterparts, PANX3-null osteoblasts secreted less extracellular ATP and failed to support plasma cells in vitro. The P2RX4-specific inhibitor 5-BDBD abrogated the impact of extracellular ATP on bone marrow plasma cells in vitro, depleted bone marrow plasma cells in vivo and reduced pre-induced antigen-specific serum antibody titre with little posttreatment rebound. P2RX4 blockade also reduced autoantibody titre and kidney disease in two mouse models of humoral autoimmunity. P2RX4 promotes plasma cell survival by regulating endoplasmic reticulum homeostasis, as short-term P2RX4 blockade caused accumulation of endoplasmic reticulum stress-associated regulatory proteins including ATF4 and B-lineage mutation of the pro-apoptotic ATF4 target Chop prevented bone marrow plasma cell demise on P2RX4 inhibition. Thus, generating mature protective and pathogenic plasma cells requires P2RX4 signalling controlled by PANX3-regulated extracellular ATP release from bone marrow niche cells.


Asunto(s)
Adenosina Trifosfato , Células de la Médula Ósea , Células Plasmáticas , Animales , Ratones , Adenosina Trifosfato/metabolismo , Autoanticuerpos/inmunología , Autoinmunidad/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Linaje de la Célula , Conexinas/genética , Conexinas/metabolismo , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Mutación , Osteoblastos/metabolismo , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptores Purinérgicos P2X4/metabolismo , Transducción de Señal
10.
Nature ; 626(8000): 864-873, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38326607

RESUMEN

Macrophage activation is controlled by a balance between activating and inhibitory receptors1-7, which protect normal tissues from excessive damage during infection8,9 but promote tumour growth and metastasis in cancer7,10. Here we report that the Kupffer cell lineage-determining factor ID3 controls this balance and selectively endows Kupffer cells with the ability to phagocytose live tumour cells and orchestrate the recruitment, proliferation and activation of natural killer and CD8 T lymphoid effector cells in the liver to restrict the growth of a variety of tumours. ID3 shifts the macrophage inhibitory/activating receptor balance to promote the phagocytic and lymphoid response, at least in part by buffering the binding of the transcription factors ELK1 and E2A at the SIRPA locus. Furthermore, loss- and gain-of-function experiments demonstrate that ID3 is sufficient to confer this potent anti-tumour activity to mouse bone-marrow-derived macrophages and human induced pluripotent stem-cell-derived macrophages. Expression of ID3 is therefore necessary and sufficient to endow macrophages with the ability to form an efficient anti-tumour niche, which could be harnessed for cell therapy in cancer.


Asunto(s)
Proteínas Inhibidoras de la Diferenciación , Macrófagos del Hígado , Neoplasias , Animales , Humanos , Ratones , Células de la Médula Ósea/citología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linaje de la Célula , Células Madre Pluripotentes Inducidas/citología , Proteínas Inhibidoras de la Diferenciación/deficiencia , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Macrófagos del Hígado/citología , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Hígado/inmunología , Hígado/patología , Activación de Macrófagos , Proteínas de Neoplasias , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Fagocitosis
11.
Immunity ; 53(2): 319-334.e6, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32814027

RESUMEN

Neutrophils are the most abundant peripheral immune cells and thus, are continually replenished by bone marrow-derived progenitors. Still, how newly identified neutrophil subsets fit into the bone marrow neutrophil lineage remains unclear. Here, we use mass cytometry to show that two recently defined human neutrophil progenitor populations contain a homogeneous progenitor subset we term "early neutrophil progenitors" (eNePs) (Lin-CD66b+CD117+CD71+). Surface marker- and RNA-expression analyses, together with in vitro colony formation and in vivo adoptive humanized mouse transfers, indicate that eNePs are the earliest human neutrophil progenitors. Furthermore, we identified CD71 as a marker associated with the earliest neutrophil developmental stages. Expression of CD71 marks proliferating neutrophils, which were expanded in the blood of melanoma patients and detectable in blood and tumors from lung cancer patients. In summary, we establish CD117+CD71+ eNeP as the inceptive human neutrophil progenitor and propose a refined model of the neutrophil developmental lineage in bone marrow.


Asunto(s)
Antígenos CD/metabolismo , Células de la Médula Ósea/citología , Células Progenitoras Mieloides/metabolismo , Neutrófilos/citología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de Transferrina/metabolismo , Traslado Adoptivo , Animales , Médula Ósea/metabolismo , Linaje de la Célula , Humanos , Masculino , Melanoma/sangre , Ratones , Ratones Endogámicos NOD , Células Progenitoras Mieloides/citología
12.
Immunity ; 53(3): 641-657.e14, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32888418

RESUMEN

Metabolic-associated fatty liver disease (MAFLD) represents a spectrum of disease states ranging from simple steatosis to non-alcoholic steatohepatitis (NASH). Hepatic macrophages, specifically Kupffer cells (KCs), are suggested to play important roles in the pathogenesis of MAFLD through their activation, although the exact roles played by these cells remain unclear. Here, we demonstrated that KCs were reduced in MAFLD being replaced by macrophages originating from the bone marrow. Recruited macrophages existed in two subsets with distinct activation states, either closely resembling homeostatic KCs or lipid-associated macrophages (LAMs) from obese adipose tissue. Hepatic LAMs expressed Osteopontin, a biomarker for patients with NASH, linked with the development of fibrosis. Fitting with this, LAMs were found in regions of the liver with reduced numbers of KCs, characterized by increased Desmin expression. Together, our data highlight considerable heterogeneity within the macrophage pool and suggest a need for more specific macrophage targeting strategies in MAFLD.


Asunto(s)
Células de la Médula Ósea/citología , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Osteopontina/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Desmina/metabolismo , Femenino , Macrófagos del Hígado/citología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteoma/metabolismo , Transcriptoma/genética
13.
Cell ; 157(2): 340-356, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725403

RESUMEN

Innate lymphoid cells (ILCs) are a recently recognized group of lymphocytes that have important functions in protecting epithelial barriers against infections and in maintaining organ homeostasis. ILCs have been categorized into three distinct groups, transcriptional circuitry and effector functions of which strikingly resemble the various T helper cell subsets. Here, we identify a common, Id2-expressing progenitor to all interleukin 7 receptor-expressing, "helper-like" ILC lineages, the CHILP. Interestingly, the CHILP differentiated into ILC2 and ILC3 lineages, but not into conventional natural killer (cNK) cells that have been considered an ILC1 subset. Instead, the CHILP gave rise to a peculiar NKp46(+) IL-7Rα(+) ILC lineage that required T-bet for specification and was distinct of cNK cells or other ILC lineages. Such ILC1s coproduced high levels of IFN-γ and TNF and protected against infections with the intracellular parasite Toxoplasma gondii. Our data significantly advance our understanding of ILC differentiation and presents evidence for a new ILC lineage that protects barrier surfaces against intracellular infections.


Asunto(s)
Diferenciación Celular , Linfocitos/citología , Linfocitos/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Factor de Transcripción GATA3/metabolismo , Inmunidad Innata , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-7/metabolismo , Células Madre/citología , Toxoplasma , Toxoplasmosis/inmunología
14.
Cell ; 156(6): 1223-1234, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24630724

RESUMEN

Splenic red pulp macrophages (RPM) degrade senescent erythrocytes and recycle heme-associated iron. The transcription factor SPI-C is selectively expressed by RPM and is required for their development, but the physiologic stimulus inducing Spic is unknown. Here, we report that Spic also regulated the development of F4/80(+)VCAM1(+) bone marrow macrophages (BMM) and that Spic expression in BMM and RPM development was induced by heme, a metabolite of erythrocyte degradation. Pathologic hemolysis induced loss of RPM and BMM due to excess heme but induced Spic in monocytes to generate new RPM and BMM. Spic expression in monocytes was constitutively inhibited by the transcriptional repressor BACH1. Heme induced proteasome-dependent BACH1 degradation and rapid Spic derepression. Furthermore, cysteine-proline dipeptide motifs in BACH1 that mediate heme-dependent degradation were necessary for Spic induction by heme. These findings are the first example of metabolite-driven differentiation of a tissue-resident macrophage subset and provide new insights into iron homeostasis.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Hemo/metabolismo , Hierro/metabolismo , Monocitos/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Macrófagos/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Bazo/citología , Bazo/metabolismo
15.
Nature ; 607(7919): 578-584, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35636458

RESUMEN

The nervous and immune systems are intricately linked1. Although psychological stress is known to modulate immune function, mechanistic pathways linking stress networks in the brain to peripheral leukocytes remain poorly understood2. Here we show that distinct brain regions shape leukocyte distribution and function throughout the body during acute stress in mice. Using optogenetics and chemogenetics, we demonstrate that motor circuits induce rapid neutrophil mobilization from the bone marrow to peripheral tissues through skeletal-muscle-derived neutrophil-attracting chemokines. Conversely, the paraventricular hypothalamus controls monocyte and lymphocyte egress from secondary lymphoid organs and blood to the bone marrow through direct, cell-intrinsic glucocorticoid signalling. These stress-induced, counter-directional, population-wide leukocyte shifts are associated with altered disease susceptibility. On the one hand, acute stress changes innate immunity by reprogramming neutrophils and directing their recruitment to sites of injury. On the other hand, corticotropin-releasing hormone neuron-mediated leukocyte shifts protect against the acquisition of autoimmunity, but impair immunity to SARS-CoV-2 and influenza infection. Collectively, these data show that distinct brain regions differentially and rapidly tailor the leukocyte landscape during psychological stress, therefore calibrating the ability of the immune system to respond to physical threats.


Asunto(s)
Encéfalo , Miedo , Leucocitos , Neuronas Motoras , Vías Nerviosas , Estrés Psicológico , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Encéfalo/citología , Encéfalo/fisiología , COVID-19/inmunología , Quimiocinas/inmunología , Susceptibilidad a Enfermedades , Miedo/fisiología , Glucocorticoides/metabolismo , Humanos , Leucocitos/citología , Leucocitos/inmunología , Linfocitos/citología , Linfocitos/inmunología , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Ratones , Monocitos/citología , Monocitos/inmunología , Neuronas Motoras/citología , Neuronas Motoras/fisiología , Neutrófilos/citología , Neutrófilos/inmunología , Optogenética , Infecciones por Orthomyxoviridae/inmunología , Núcleo Hipotalámico Paraventricular/fisiología , SARS-CoV-2/inmunología , Estrés Psicológico/inmunología , Estrés Psicológico/fisiopatología
16.
Annu Rev Cell Dev Biol ; 30: 677-704, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25150008

RESUMEN

Two opposing descriptions of so-called mesenchymal stem cells (MSCs) exist at this time. One sees MSCs as the postnatal, self-renewing, and multipotent stem cells for the skeleton. This cell coincides with a specific type of bone marrow perivascular cell. In skeletal physiology, this skeletal stem cell is pivotal to the growth and lifelong turnover of bone and to its native regeneration capacity. In hematopoietic physiology, its role as a key player in maintaining hematopoietic stem cells in their niche and in regulating the hematopoietic microenvironment is emerging. In the alternative description, MSCs are ubiquitous in connective tissues and are defined by in vitro characteristics and by their use in therapy, which rests on their ability to modulate the function of host tissues rather than on stem cell properties. Here, I discuss how the two views developed, conceptually and experimentally, and attempt to clarify the confusion arising from their collision.


Asunto(s)
Células Madre Mesenquimatosas/citología , Animales , Células de la Médula Ósea/clasificación , Células de la Médula Ósea/citología , Huesos/citología , Antígeno CD146/análisis , Separación Celular/métodos , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Células Clonales/citología , Tejido Conectivo/inmunología , Humanos , Inmunomodulación , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/clasificación , Ratones , Modelos Biológicos , Pericitos/citología , Células Madre Pluripotentes/citología , Quimera por Radiación , Nicho de Células Madre , Células del Estroma/clasificación , Células del Estroma/citología , Trasplante Heterotópico
17.
Nat Methods ; 21(5): 868-881, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38374263

RESUMEN

The human bone marrow (BM) niche sustains hematopoiesis throughout life. We present a method for generating complex BM-like organoids (BMOs) from human induced pluripotent stem cells (iPSCs). BMOs consist of key cell types that self-organize into spatially defined three-dimensional structures mimicking cellular, structural and molecular characteristics of the hematopoietic microenvironment. Functional properties of BMOs include the presence of an in vivo-like vascular network, the presence of multipotent mesenchymal stem/progenitor cells, the support of neutrophil differentiation and responsiveness to inflammatory stimuli. Single-cell RNA sequencing revealed a heterocellular composition including the presence of a hematopoietic stem/progenitor (HSPC) cluster expressing genes of fetal HSCs. BMO-derived HSPCs also exhibited lymphoid potential and a subset demonstrated transient engraftment potential upon xenotransplantation in mice. We show that the BMOs could enable the modeling of hematopoietic developmental aspects and inborn errors of hematopoiesis, as shown for human VPS45 deficiency. Thus, iPSC-derived BMOs serve as a physiologically relevant in vitro model of the human BM microenvironment to study hematopoietic development and BM diseases.


Asunto(s)
Diferenciación Celular , Hematopoyesis , Células Madre Pluripotentes Inducidas , Organoides , Humanos , Organoides/citología , Organoides/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Ratones , Células Madre Hematopoyéticas/citología , Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Técnicas de Cultivo de Célula/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo
18.
Nat Immunol ; 16(10): 1044-50, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26280998

RESUMEN

The cellular and molecular events that drive the early development of innate lymphoid cells (ILCs) remain poorly understood. We show that the transcription factor TCF-1 is required for the efficient generation of all known adult ILC subsets and their precursors. Using novel reporter mice, we identified a new subset of early ILC progenitors (EILPs) expressing high amounts of TCF-1. EILPs lacked efficient T and B lymphocyte potential but efficiently gave rise to NK cells and all known adult helper ILC lineages, indicating that they are the earliest ILC-committed progenitors identified so far. Our results suggest that upregulation of TCF-1 expression denotes the earliest stage of ILC fate specification. The discovery of EILPs provides a basis for deciphering additional signals that specify ILC fate.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Inmunidad Innata , Linfocitos/citología , Linfocitos/inmunología , Factor 1 de Transcripción de Linfocitos T/genética , Regulación hacia Arriba , Animales , Células Cultivadas , Citometría de Flujo , Ratones , Análisis por Micromatrices , Factor 1 de Transcripción de Linfocitos T/metabolismo
19.
Immunity ; 49(4): 627-639.e6, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30314756

RESUMEN

The non-hematopoietic cell fraction of the bone marrow (BM) is classically identified as CD45- Ter119- CD31- (herein referred to as triple-negative cells or TNCs). Although TNCs are believed to contain heterogeneous stromal cell populations, they remain poorly defined. Here we showed that the vast majority of TNCs (∼85%) have a hematopoietic rather than mesenchymal origin. Single cell RNA-sequencing revealed erythroid and lymphoid progenitor signatures among CD51- TNCs. Ly6D+ CD44+ CD51- TNCs phenotypically and functionally resembled CD45+ pro-B lymphoid cells, whereas Ly6D- CD44+ CD51- TNCs were enriched in previously unappreciated stromal-dependent erythroid progenitors hierarchically situated between preCFU-E and proerythroblasts. Upon adoptive transfer, CD44+ CD51- TNCs contributed to repopulate the B-lymphoid and erythroid compartments. CD44+ CD51- TNCs also expanded during phenylhydrazine-induced acute hemolysis or in a model of sickle cell anemia. These findings thus uncover physiologically relevant new classes of stromal-associated functional CD45- hematopoietic progenitors.


Asunto(s)
Células de la Médula Ósea/inmunología , Células Eritroides/inmunología , Células Progenitoras Linfoides/inmunología , Células del Estroma/inmunología , Traslado Adoptivo/métodos , Animales , Antígenos de Grupos Sanguíneos/inmunología , Antígenos de Grupos Sanguíneos/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Células Eritroides/citología , Células Eritroides/metabolismo , Antígenos Comunes de Leucocito/inmunología , Antígenos Comunes de Leucocito/metabolismo , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo
20.
Nature ; 595(7867): 421-425, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34030176

RESUMEN

Long-lived bone marrow plasma cells (BMPCs) are a persistent and essential source of protective antibodies1-7. Individuals who have recovered from COVID-19 have a substantially lower risk of reinfection with SARS-CoV-28-10. Nonetheless, it has been reported that levels of anti-SARS-CoV-2 serum antibodies decrease rapidly in the first few months after infection, raising concerns that long-lived BMPCs may not be generated and humoral immunity against SARS-CoV-2 may be short-lived11-13. Here we show that in convalescent individuals who had experienced mild SARS-CoV-2 infections (n = 77), levels of serum anti-SARS-CoV-2 spike protein (S) antibodies declined rapidly in the first 4 months after infection and then more gradually over the following 7 months, remaining detectable at least 11 months after infection. Anti-S antibody titres correlated with the frequency of S-specific plasma cells in bone marrow aspirates from 18 individuals who had recovered from COVID-19 at 7 to 8 months after infection. S-specific BMPCs were not detected in aspirates from 11 healthy individuals with no history of SARS-CoV-2 infection. We show that S-binding BMPCs are quiescent, which suggests that they are part of a stable compartment. Consistently, circulating resting memory B cells directed against SARS-CoV-2 S were detected in the convalescent individuals. Overall, our results indicate that mild infection with SARS-CoV-2 induces robust antigen-specific, long-lived humoral immune memory in humans.


Asunto(s)
Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , COVID-19/inmunología , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Adulto , Anciano , Supervivencia Celular , Femenino , Humanos , Memoria Inmunológica , Masculino , Persona de Mediana Edad , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA