Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nature ; 595(7868): 578-584, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34135508

RESUMEN

Macrophages have a key role in shaping the tumour microenvironment (TME), tumour immunity and response to immunotherapy, which makes them an important target for cancer treatment1,2. However, modulating macrophages has proved extremely difficult, as we still lack a complete understanding of the molecular and functional diversity of the tumour macrophage compartment. Macrophages arise from two distinct lineages. Tissue-resident macrophages self-renew locally, independent of adult haematopoiesis3-5, whereas short-lived monocyte-derived macrophages arise from adult haematopoietic stem cells, and accumulate mostly in inflamed lesions1. How these macrophage lineages contribute to the TME and cancer progression remains unclear. To explore the diversity of the macrophage compartment in human non-small cell lung carcinoma (NSCLC) lesions, here we performed single-cell RNA sequencing of tumour-associated leukocytes. We identified distinct populations of macrophages that were enriched in human and mouse lung tumours. Using lineage tracing, we discovered that these macrophage populations differ in origin and have a distinct temporal and spatial distribution in the TME. Tissue-resident macrophages accumulate close to tumour cells early during tumour formation to promote epithelial-mesenchymal transition and invasiveness in tumour cells, and they also induce a potent regulatory T cell response that protects tumour cells from adaptive immunity. Depletion of tissue-resident macrophages reduced the numbers and altered the phenotype of regulatory T cells, promoted the accumulation of CD8+ T cells and reduced tumour invasiveness and growth. During tumour growth, tissue-resident macrophages became redistributed at the periphery of the TME, which becomes dominated by monocyte-derived macrophages in both mouse and human NSCLC. This study identifies the contribution of tissue-resident macrophages to early lung cancer and establishes them as a target for the prevention and treatment of early lung cancer lesions.


Asunto(s)
Carcinogénesis , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Macrófagos/inmunología , Microambiente Tumoral , Animales , Linfocitos T CD8-positivos/inmunología , Transición Epitelial-Mesenquimal , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Linfocitos T Reguladores/inmunología
2.
Proc Natl Acad Sci U S A ; 120(22): e2302019120, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37216517

RESUMEN

Mammalian aging is associated with multiple defects of hematopoiesis, most prominently with the impaired development of T and B lymphocytes. This defect is thought to originate in hematopoietic stem cells (HSCs) of the bone marrow, specifically due to the age-dependent accumulation of HSCs with preferential megakaryocytic and/or myeloid potential ("myeloid bias"). Here, we tested this notion using inducible genetic labeling and tracing of HSCs in unmanipulated animals. We found that the endogenous HSC population in old mice shows reduced differentiation into all lineages including lymphoid, myeloid, and megakaryocytic. Single-cell RNA sequencing and immunophenotyping (CITE-Seq) showed that HSC progeny in old animals comprised balanced lineage spectrum including lymphoid progenitors. Lineage tracing using the aging-induced HSC marker Aldh1a1 confirmed the low contribution of old HSCs across all lineages. Competitive transplantations of total bone marrow cells with genetically marked HSCs revealed that the contribution of old HSCs was reduced, but compensated by other donor cells in myeloid cells but not in lymphocytes. Thus, the HSC population in old animals becomes globally decoupled from hematopoiesis, which cannot be compensated in lymphoid lineages. We propose that this partially compensated decoupling, rather than myeloid bias, is the primary cause of the selective impairment of lymphopoiesis in older mice.


Asunto(s)
Envejecimiento , Células Madre Hematopoyéticas , Ratones , Animales , Linaje de la Célula , Diferenciación Celular , Médula Ósea , Hematopoyesis , Mamíferos
3.
Immunity ; 45(3): 597-609, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27590115

RESUMEN

Hematopoietic stem cells (HSCs) sustain long-term reconstitution of hematopoiesis in transplantation recipients, yet their role in the endogenous steady-state hematopoiesis remains unclear. In particular, recent studies suggested that HSCs provide a relatively minor contribution to immune cell development in adults. We directed transgene expression in a fraction of HSCs that maintained reconstituting activity during serial transplantations. Inducible genetic labeling showed that transgene-expressing HSCs gave rise to other phenotypic HSCs, confirming their top position in the differentiation hierarchy. The labeled HSCs rapidly contributed to committed progenitors of all lineages and to mature myeloid cells and lymphocytes, but not to B-1a cells or tissue macrophages. Importantly, labeled HSCs gave rise to more than two-thirds of all myeloid cells and platelets in adult mice, and this contribution could be accelerated by an induced interferon response. Thus, classically defined HSCs maintain immune cell development in the steady state and during systemic cytokine responses.


Asunto(s)
Linaje de la Célula/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Animales , Linfocitos B/metabolismo , Linfocitos B/fisiología , Plaquetas/metabolismo , Plaquetas/fisiología , Diferenciación Celular/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/metabolismo , Interferones/metabolismo , Macrófagos/metabolismo , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Células Mieloides/fisiología
4.
Curr Opin Hematol ; 27(4): 241-247, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32398457

RESUMEN

PURPOSE OF REVIEW: Although hematopoietic stem cell (HSC) function has long been studied by transplantation assays, this does not reflect what HSCs actually do in their native context. Here, we review recent technologic advances that facilitate the study of HSCs in their native context focusing on inducible HSC-specific lineage tracing and inference of hematopoietic trajectories through single-cell RNA sequencing (scRNA-Seq). RECENT FINDINGS: Lineage tracing of HSCs at the population level using multiple systems has suggested that HSCs make a major contribution to steady-state hematopoiesis. Although several genetic systems and novel methods for lineage tracing individual hematopoietic clones have been described, the technology for tracking these cellular barcodes (in particular mutations or insertion sites) is still in its infancy. Thus, lineage tracing of HSC clones in the adult bone marrow remains elusive. Static snapshots of scRNA-Seq of hematopoietic populations have captured the heterogeneity of transcriptional profiles of HSCs and progenitors, with some cells displaying a unilineage signature as well as others with bi or multipotent lineage profiles. Kinetic analysis using HSC-specific lineage tracing combined with scRNA-Seq confirmed this heterogeneity of progenitor populations and revealed a rapid and early emergence of megakaryocytic progeny, followed by erythroid and myeloid lineages, whereas lymphoid differentiation emerged last. SUMMARY: New approaches to study HSCs both in vivo through lineage tracing and at a high-resolution molecular level through scRNA-Seq are providing key insight into HSC differentiation in the absence of transplantation. Recent studies using these approaches are discussed here. These studies pave the way for integration of in-vivo clonal analysis of HSC behavior over time with single-cell sequencing data, including but not limited to transcriptomic, proteomic, and epigenomic, to establish a comprehensive molecular and cellular map of hematopoiesis.


Asunto(s)
Células Madre Adultas/metabolismo , Diferenciación Celular , Células Madre Hematopoyéticas/metabolismo , Adulto , Células Madre Adultas/citología , Genómica , Células Madre Hematopoyéticas/citología , Humanos , RNA-Seq
5.
Immunity ; 28(3): 335-45, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18280186

RESUMEN

Productive rearrangement of the immunoglobulin heavy-chain locus triggers a major developmental checkpoint that promotes limited clonal expansion of pre-B cells, thereby culminating in cell-cycle arrest and rearrangement of light-chain loci. By using Irf4-/-Irf8-/- pre-B cells, we demonstrated that two pathways converge to synergistically drive light-chain rearrangement, but not simply as a consequence of cell-cycle exit. One pathway was directly dependent on transcription factor IRF-4, whose expression was elevated by pre-B cell receptor signaling. IRF-4 targeted the immunoglobulin 3'Ekappa and Elambda enhancers and positioned a kappa allele away from pericentromeric heterochromatin. The other pathway was triggered by attenuation of IL-7 signaling and activated the iEkappa enhancer via binding of the transcription factor E2A. IRF-4 also regulated expression of chemokine receptor Cxcr4 and promoted migration of pre-B cells in response to the chemokine ligand CXCL12. We propose that IRF-4 coordinates the two pathways regulating light-chain recombination by positioning pre-B cells away from IL-7-expressing stromal cells.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Reordenamiento Génico de Cadena Ligera de Linfocito B/inmunología , Factores Reguladores del Interferón/inmunología , Interleucina-7/inmunología , Transducción de Señal/inmunología , Animales , Linfocitos B/citología , Western Blotting , Movimiento Celular/inmunología , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Inmunoprecipitación , Hibridación Fluorescente in Situ , Factores Reguladores del Interferón/metabolismo , Interleucina-7/metabolismo , Ratones , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Células Madre/inmunología
6.
Elife ; 132024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39017663

RESUMEN

A new mathematical model can estimate the number of precursor cells that contribute to regenerating blood cells in mice.


Asunto(s)
Modelos Teóricos , Animales , Ratones , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología
7.
Biol Rev Camb Philos Soc ; 98(5): 1668-1686, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37157910

RESUMEN

Cancers rely on multiple, heterogeneous processes at different scales, pertaining to many biomedical fields. Therefore, understanding cancer is necessarily an interdisciplinary task that requires placing specialised experimental and clinical research into a broader conceptual, theoretical, and methodological framework. Without such a framework, oncology will collect piecemeal results, with scant dialogue between the different scientific communities studying cancer. We argue that one important way forward in service of a more successful dialogue is through greater integration of applied sciences (experimental and clinical) with conceptual and theoretical approaches, informed by philosophical methods. By way of illustration, we explore six central themes: (i) the role of mutations in cancer; (ii) the clonal evolution of cancer cells; (iii) the relationship between cancer and multicellularity; (iv) the tumour microenvironment; (v) the immune system; and (vi) stem cells. In each case, we examine open questions in the scientific literature through a philosophical methodology and show the benefit of such a synergy for the scientific and medical understanding of cancer.


Asunto(s)
Neoplasias , Filosofía , Investigación , Estudios Interdisciplinarios
8.
Cell Stem Cell ; 27(2): 336-345.e4, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32589864

RESUMEN

Adult mammalian hematopoietic stem cells (HSCs) reside in the bone marrow (BM) but can be mobilized into blood for use in transplantation. HSCs interact with BM niche cells that produce growth factor c-Kit ligand (Kitl/SCF) and chemokine CXCL12, and were thought to be static and sessile. We used two-photon laser scanning microscopy to visualize genetically labeled HSCs in the BM of live mice for several hours. The majority of HSCs showed a dynamic non-spherical morphology and significant motility, undergoing slow processive motion interrupted by short stretches of confined motion. HSCs moved in the perivascular space and showed intermittent close contacts with SCF-expressing perivascular stromal cells. In contrast, mobilization-inducing blockade of CXCL12 receptor CXCR4 and integrins rapidly abrogated HSC motility and shape dynamics in real time. Our results reveal an unexpectedly dynamic nature of HSC residence in the BM and interaction with the SCF+ stromal niche, which is disrupted during HSC mobilization.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Animales , Células de la Médula Ósea , Movimiento Celular , Quimiocina CXCL12 , Microscopía Intravital , Ratones , Nicho de Células Madre
9.
Sci Immunol ; 5(45)2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32220976

RESUMEN

Tissue-resident macrophages are a diverse population of cells that perform specialized functions including sustaining tissue homeostasis and tissue surveillance. Here, we report an interstitial subset of CD169+ lung-resident macrophages that are transcriptionally and developmentally distinct from alveolar macrophages (AMs). They are primarily localized around the airways and are found in close proximity to the sympathetic nerves in the bronchovascular bundle. These nerve- and airway-associated macrophages (NAMs) are tissue resident, yolk sac derived, self-renewing, and do not require CCR2+ monocytes for development or maintenance. Unlike AMs, the development of NAMs requires CSF1 but not GM-CSF. Bulk population and single-cell transcriptome analysis indicated that NAMs are distinct from other lung-resident macrophage subsets and highly express immunoregulatory genes under steady-state and inflammatory conditions. NAMs proliferated robustly after influenza infection and activation with the TLR3 ligand poly(I:C), and in their absence, the inflammatory response was augmented, resulting in excessive production of inflammatory cytokines and innate immune cell infiltration. Overall, our study provides insights into a distinct subset of airway-associated pulmonary macrophages that function to maintain immune and tissue homeostasis.


Asunto(s)
Macrófagos Alveolares/inmunología , Neuronas/inmunología , Animales , Homeostasis/inmunología , Factor Estimulante de Colonias de Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Saco Vitelino/citología , Saco Vitelino/inmunología
10.
Exp Hematol ; 61: 26-35, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29501466

RESUMEN

The production of blood cells is dependent on the activity of a rare stem cell population that normally resides in the bone marrow (BM) of the organism. These hematopoietic stem cells (HSCs) have the ability to both self-renew and differentiate, ensuring this lifelong hematopoiesis. Determining the regulation of HSC functions should thus provide critical insight to advancing regenerative medicine. Until quite recently, HSCs were primarily studied using in vitro studies and transplantations into immunodeficient hosts. Indeed, the definition of a bona fide HSC is its ability to reconstitute lymphopenic hosts. In this review, we discuss the development of novel, HSC-specific genetic reporter systems that enable the prospective identification of HSCs and the study of their functions in the absence of transplantation. Coupled with additional technological advances, these studies are now defining the fundamental properties of HSCs in vivo. Furthermore, complex cellular and molecular mechanisms that regulate HSC dormancy, self-renewal, and differentiation are being identified and further dissected. These novel reporter systems represent a major technological advance for the stem cell field and allow new questions to be addressed.


Asunto(s)
Células de la Médula Ósea/citología , Técnicas Genéticas/tendencias , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Linaje de la Célula/genética , Genes Reporteros/genética , Hematopoyesis , Humanos
11.
J Exp Med ; 215(11): 2815-2832, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30291161

RESUMEN

Adult hematopoiesis has been studied in terms of progenitor differentiation potentials, whereas its kinetics in vivo is poorly understood. We combined inducible lineage tracing of endogenous adult hematopoietic stem cells (HSCs) with flow cytometry and single-cell RNA sequencing to characterize early steps of hematopoietic differentiation in the steady-state. Labeled cells, comprising primarily long-term HSCs and some short-term HSCs, produced megakaryocytic lineage progeny within 1 wk in a process that required only two to three cell divisions. Erythroid and myeloid progeny emerged simultaneously by 2 wk and included a progenitor population with expression features of both lineages. Myeloid progenitors at this stage showed diversification into granulocytic, monocytic, and dendritic cell types, and rare intermediate cell states could be detected. In contrast, lymphoid differentiation was virtually absent within the first 3 wk of tracing. These results show that continuous differentiation of HSCs rapidly produces major hematopoietic lineages and cell types and reveal fundamental kinetic differences between megakaryocytic, erythroid, myeloid, and lymphoid differentiation.


Asunto(s)
Células Madre Adultas/inmunología , Diferenciación Celular/inmunología , División Celular/inmunología , Células Madre Hematopoyéticas/inmunología , Células Madre Adultas/citología , Animales , Células Dendríticas/citología , Células Dendríticas/inmunología , Granulocitos/citología , Granulocitos/inmunología , Células Madre Hematopoyéticas/citología , Cinética , Megacariocitos/citología , Megacariocitos/inmunología , Ratones , Ratones Transgénicos , Monocitos/citología , Monocitos/inmunología
12.
Front Immunol ; 9: 2475, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30410494

RESUMEN

Inflammatory bowel disease (IBD) is a chronic inflammatory condition caused by an aberrant immune response to microbial components of the gastrointestinal tract. Plasmacytoid dendritic cells (pDCs) are innate immune cells specialized in the production of type I interferons and were recently implicated in the pathogenesis of autoimmune disorders such as lupus and scleroderma. While pDCs were shown to infiltrate intestinal mucosa of IBD patients and proposed to participate in intestinal inflammation, their net contribution to the disease remains unclear. We addressed this question by targeting the pDC-specific transcription factor TCF4 (E2-2) in experimental IBD caused by deficiency of Wiskott-Aldrich syndrome protein (WASP) or of interleukin-10 (IL-10). Monoallelic Tcf4 deletion, which was previously shown to abrogate experimental lupus, did not affect autoimmunity manifestations or colitis in WASP-deficient animals. Furthermore, conditional biallelic Tcf4 targeting resulted in a near-complete pDC ablation, yet had no effect on the development of colitis in IL-10-deficient mice. Our results suggest that, in contrast to other inflammatory and autoimmune diseases, pDCs do not play a major role in the pathogenesis of intestinal inflammation during IBD.


Asunto(s)
Colitis/inmunología , Células Dendríticas/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Nefritis Lúpica/inmunología , Factor de Transcripción 4/metabolismo , Animales , Colitis/genética , Humanos , Inmunidad Innata , Interleucina-10/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Factor de Transcripción 4/genética , Proteína del Síndrome de Wiskott-Aldrich/genética
13.
J Exp Med ; 210(11): 2151-9, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-24101375

RESUMEN

Plasmacytoid dendritic cells (pDCs) rapidly produce type I interferon (IFN-I) in response to viruses and are essential for antiviral immune responses. Although related to classical DCs (cDCs) in their development and expression profile, pDCs possess many distinct features. Unlike cDCs, pDCs develop in the bone marrow (BM) and emerge into peripheral lymphoid organs and tissues as fully differentiated cells. We now report that pDCs specifically express Runx2, a Runt family transcription factor that is essential for bone development. pDCs in Runx2-deficient mice developed normally in the BM but were greatly reduced in the periphery. The defect was cell-intrinsic and was associated with the retention of mature Ly49Q(+) pDCs in the BM. Runx2 was required for the expression of several pDC-enriched genes, including the chemokine receptors Ccr2 and Ccr5. Mature pDCs expressed high levels of Ccr5 at the cell surface, and Ccr5-deficient pDCs in a competitive setting were reduced in the periphery relative to the BM. Thus, Runx2 is required for the emergence of mature BM pDCs into the periphery, in a process that is partially dependent on Ccr5. These results establish Runx2 as a lineage-specific regulator of immune system development.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Factores de Transcripción/metabolismo , Animales , Células de la Médula Ósea/citología , Eliminación de Gen , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Receptores CCR5/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo
14.
Cancer Cell ; 22(4): 452-65, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-23079656

RESUMEN

D-type cyclins form complexes with cyclin-dependent kinases (CDK4/6) and promote cell cycle progression. Although cyclin D functions appear largely tissue specific, we demonstrate that cyclin D3 has unique functions in lymphocyte development and cannot be replaced by cyclin D2, which is also expressed during blood differentiation. We show that only combined deletion of p27(Kip1) and retinoblastoma tumor suppressor (Rb) is sufficient to rescue the development of Ccnd3(-/-) thymocytes. Furthermore, we show that a small molecule targeting the kinase function of cyclin D3:CDK4/6 inhibits both cell cycle entry in human T cell acute lymphoblastic leukemia (T-ALL) and disease progression in animal models of T-ALL. These studies identify unique functions for cyclin D3:CDK4/6 complexes and suggest potential therapeutic protocols for this devastating blood tumor.


Asunto(s)
Ciclina D3/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Animales , Ciclina D2/fisiología , Ciclina D3/fisiología , Quinasa 4 Dependiente de la Ciclina/fisiología , Quinasa 6 Dependiente de la Ciclina/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Humanos , Linfocitos/fisiología , Ratones , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Receptor Notch1/fisiología , Proteína de Retinoblastoma/fisiología
15.
Nat Immunol ; 7(5): 489-97, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16582912

RESUMEN

During hematopoiesis, stem cell proliferation is dependent on expression of the D-type cyclins. However, little is known about how each cyclin D contributes to the development of specific hematopoietic lineages. Here, analysis of Ccnd1(-/-), Ccnd2(-/-), Ccnd3(-/-) and Ccnd2(-/-)Ccnd3(-/-) mice showed that cyclin D3 was uniquely required for the development of pre-B cells. Transcription of Ccnd3 was dependent on expression of the common gamma-chain. In contrast, expression of the pre-B cell receptor and activation of 'downstream' signaling pathways prevented proteasome-mediated degradation of cyclin D3. Cyclin D3 has a key function in B cell development by integrating cytokine and pre-B cell receptor-dependent signals to expand the pool of pre-B cells that have successfully rearranged immunoglobulin heavy chain.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Ciclinas/fisiología , Animales , Linfocitos B/citología , Línea Celular , Ciclina D3 , Ciclinas/genética , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA