Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 422
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(6): 1179-1194.e15, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36931245

RESUMEN

The human brain undergoes rapid development at mid-gestation from a pool of neural stem and progenitor cells (NSPCs) that give rise to the neurons, oligodendrocytes, and astrocytes of the mature brain. Functional study of these cell types has been hampered by a lack of precise purification methods. We describe a method for prospectively isolating ten distinct NSPC types from the developing human brain using cell-surface markers. CD24-THY1-/lo cells were enriched for radial glia, which robustly engrafted and differentiated into all three neural lineages in the mouse brain. THY1hi cells marked unipotent oligodendrocyte precursors committed to an oligodendroglial fate, and CD24+THY1-/lo cells marked committed excitatory and inhibitory neuronal lineages. Notably, we identify and functionally characterize a transcriptomically distinct THY1hiEGFRhiPDGFRA- bipotent glial progenitor cell (GPC), which is lineage-restricted to astrocytes and oligodendrocytes, but not to neurons. Our study provides a framework for the functional study of distinct cell types in human neurodevelopment.


Asunto(s)
Células-Madre Neurales , Ratones , Animales , Humanos , Células-Madre Neurales/metabolismo , Neuronas , Diferenciación Celular/fisiología , Neuroglía/metabolismo , Encéfalo , Astrocitos
2.
Cell ; 175(1): 43-56.e21, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30241615

RESUMEN

Stem cell regulation and hierarchical organization of human skeletal progenitors remain largely unexplored. Here, we report the isolation of a self-renewing and multipotent human skeletal stem cell (hSSC) that generates progenitors of bone, cartilage, and stroma, but not fat. Self-renewing and multipotent hSSCs are present in fetal and adult bones and can also be derived from BMP2-treated human adipose stroma (B-HAS) and induced pluripotent stem cells (iPSCs). Gene expression analysis of individual hSSCs reveals overall similarity between hSSCs obtained from different sources and partially explains skewed differentiation toward cartilage in fetal and iPSC-derived hSSCs. hSSCs undergo local expansion in response to acute skeletal injury. In addition, hSSC-derived stroma can maintain human hematopoietic stem cells (hHSCs) in serum-free culture conditions. Finally, we combine gene expression and epigenetic data of mouse skeletal stem cells (mSSCs) and hSSCs to identify evolutionarily conserved and divergent pathways driving SSC-mediated skeletogenesis. VIDEO ABSTRACT.


Asunto(s)
Desarrollo Óseo/fisiología , Huesos/citología , Células Madre Hematopoyéticas/citología , Animales , Huesos/metabolismo , Cartílago/citología , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Análisis de la Célula Individual/métodos , Células Madre/citología , Células del Estroma/citología , Transcriptoma/genética
3.
Cell ; 168(6): 1041-1052.e18, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28283060

RESUMEN

Most secreted growth factors and cytokines are functionally pleiotropic because their receptors are expressed on diverse cell types. While important for normal mammalian physiology, pleiotropy limits the efficacy of cytokines and growth factors as therapeutics. Stem cell factor (SCF) is a growth factor that acts through the c-Kit receptor tyrosine kinase to elicit hematopoietic progenitor expansion but can be toxic when administered in vivo because it concurrently activates mast cells. We engineered a mechanism-based SCF partial agonist that impaired c-Kit dimerization, truncating downstream signaling amplitude. This SCF variant elicited biased activation of hematopoietic progenitors over mast cells in vitro and in vivo. Mouse models of SCF-mediated anaphylaxis, radioprotection, and hematopoietic expansion revealed that this SCF partial agonist retained therapeutic efficacy while exhibiting virtually no anaphylactic off-target effects. The approach of biasing cell activation by tuning signaling thresholds and outputs has applications to many dimeric receptor-ligand systems.


Asunto(s)
Anafilaxia/metabolismo , Células Madre Hematopoyéticas/inmunología , Mastocitos/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal , Factor de Células Madre/metabolismo , Anafilaxia/inmunología , Animales , Dimerización , Humanos , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Ingeniería de Proteínas , Proteínas Proto-Oncogénicas c-kit/agonistas , Proteínas Proto-Oncogénicas c-kit/química , Factor de Células Madre/química , Factor de Células Madre/genética
4.
Cell ; 164(4): 593-5, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26871622

RESUMEN

Double-strand break repair is required for neural development, and brain cells contain somatic genomic variations. Now, Wei et al. demonstrate that neural stem and progenitor cells undergo very frequent DNA breaks in a very restricted set of genes involved in neural cell adhesion and synapse function.


Asunto(s)
Roturas del ADN , Células-Madre Neurales/metabolismo , Animales , Humanos
5.
Cell ; 166(2): 451-467, 2016 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-27419872

RESUMEN

Stem-cell differentiation to desired lineages requires navigating alternating developmental paths that often lead to unwanted cell types. Hence, comprehensive developmental roadmaps are crucial to channel stem-cell differentiation toward desired fates. To this end, here, we map bifurcating lineage choices leading from pluripotency to 12 human mesodermal lineages, including bone, muscle, and heart. We defined the extrinsic signals controlling each binary lineage decision, enabling us to logically block differentiation toward unwanted fates and rapidly steer pluripotent stem cells toward 80%-99% pure human mesodermal lineages at most branchpoints. This strategy enabled the generation of human bone and heart progenitors that could engraft in respective in vivo models. Mapping stepwise chromatin and single-cell gene expression changes in mesoderm development uncovered somite segmentation, a previously unobservable human embryonic event transiently marked by HOPX expression. Collectively, this roadmap enables navigation of mesodermal development to produce transplantable human tissue progenitors and uncover developmental processes. VIDEO ABSTRACT.


Asunto(s)
Mesodermo/citología , Transducción de Señal , Proteínas Morfogenéticas Óseas/metabolismo , Huesos/citología , Huesos/metabolismo , Corazón/crecimiento & desarrollo , Proteínas de Homeodominio/metabolismo , Humanos , Mesodermo/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/metabolismo , Línea Primitiva/citología , Línea Primitiva/metabolismo , Análisis de la Célula Individual , Somitos/metabolismo , Células Madre , Proteínas Supresoras de Tumor/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/metabolismo
6.
Nat Immunol ; 19(1): 76-84, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29180808

RESUMEN

Exciting progress in the field of cancer immunotherapy has renewed the urgency of the need for basic studies of immunoregulation in both adaptive cell lineages and innate cell lineages. Here we found a central role for major histocompatibility complex (MHC) class I in controlling the phagocytic function of macrophages. Our results demonstrated that expression of the common MHC class I component ß2-microglobulin (ß2M) by cancer cells directly protected them from phagocytosis. We further showed that this protection was mediated by the inhibitory receptor LILRB1, whose expression was upregulated on the surface of macrophages, including tumor-associated macrophages. Disruption of either MHC class I or LILRB1 potentiated phagocytosis of tumor cells both in vitro and in vivo, which defines the MHC class I-LILRB1 signaling axis as an important regulator of the effector function of innate immune cells, a potential biomarker for therapeutic response to agents directed against the signal-regulatory protein CD47 and a potential target of anti-cancer immunotherapy.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Receptor Leucocitario Tipo Inmunoglobulina B1/inmunología , Macrófagos/inmunología , Neoplasias/inmunología , Fagocitosis/inmunología , Animales , Línea Celular Tumoral , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoterapia/métodos , Receptor Leucocitario Tipo Inmunoglobulina B1/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/metabolismo , Neoplasias/terapia , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia
7.
Immunity ; 54(3): 586-602.e8, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33691136

RESUMEN

To identify disease-relevant T cell receptors (TCRs) with shared antigen specificity, we analyzed 778,938 TCRß chain sequences from 178 non-small cell lung cancer patients using the GLIPH2 (grouping of lymphocyte interactions with paratope hotspots 2) algorithm. We identified over 66,000 shared specificity groups, of which 435 were clonally expanded and enriched in tumors compared to adjacent lung. The antigenic epitopes of one such tumor-enriched specificity group were identified using a yeast peptide-HLA A∗02:01 display library. These included a peptide from the epithelial protein TMEM161A, which is overexpressed in tumors and cross-reactive epitopes from Epstein-Barr virus and E. coli. Our findings suggest that this cross-reactivity may underlie the presence of virus-specific T cells in tumor infiltrates and that pathogen cross-reactivity may be a feature of multiple cancers. The approach and analytical pipelines generated in this work, as well as the specificity groups defined here, present a resource for understanding the T cell response in cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Mapeo Epitopo/métodos , Epítopos de Linfocito T/genética , Neoplasias Pulmonares/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T/inmunología , Algoritmos , Presentación de Antígeno , Antígenos de Neoplasias/metabolismo , Células Cultivadas , Reacciones Cruzadas , Epítopos de Linfocito T/metabolismo , Antígeno HLA-A2/metabolismo , Humanos , Unión Proteica , Especificidad del Receptor de Antígeno de Linfocitos T
8.
Cell ; 160(1-2): 285-98, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25594184

RESUMEN

How are skeletal tissues derived from skeletal stem cells? Here, we map bone, cartilage, and stromal development from a population of highly pure, postnatal skeletal stem cells (mouse skeletal stem cells, mSSCs) to their downstream progenitors of bone, cartilage, and stromal tissue. We then investigated the transcriptome of the stem/progenitor cells for unique gene-expression patterns that would indicate potential regulators of mSSC lineage commitment. We demonstrate that mSSC niche factors can be potent inducers of osteogenesis, and several specific combinations of recombinant mSSC niche factors can activate mSSC genetic programs in situ, even in nonskeletal tissues, resulting in de novo formation of cartilage or bone and bone marrow stroma. Inducing mSSC formation with soluble factors and subsequently regulating the mSSC niche to specify its differentiation toward bone, cartilage, or stromal cells could represent a paradigm shift in the therapeutic regeneration of skeletal tissues.


Asunto(s)
Huesos/citología , Células Madre Mesenquimatosas/citología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Cartílago/citología , Linaje de la Célula , Cruzamientos Genéticos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
9.
Cell ; 160(6): 1196-208, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25728669

RESUMEN

Most cell-surface receptors for cytokines and growth factors signal as dimers, but it is unclear whether remodeling receptor dimer topology is a viable strategy to "tune" signaling output. We utilized diabodies (DA) as surrogate ligands in a prototypical dimeric receptor-ligand system, the cytokine Erythropoietin (EPO) and its receptor (EpoR), to dimerize EpoR ectodomains in non-native architectures. Diabody-induced signaling amplitudes varied from full to minimal agonism, and structures of these DA/EpoR complexes differed in EpoR dimer orientation and proximity. Diabodies also elicited biased or differential activation of signaling pathways and gene expression profiles compared to EPO. Non-signaling diabodies inhibited proliferation of erythroid precursors from patients with a myeloproliferative neoplasm due to a constitutively active JAK2V617F mutation. Thus, intracellular oncogenic mutations causing ligand-independent receptor activation can be counteracted by extracellular ligands that re-orient receptors into inactive dimer topologies. This approach has broad applications for tuning signaling output for many dimeric receptor systems.


Asunto(s)
Receptores de Eritropoyetina/química , Receptores de Eritropoyetina/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Línea Celular , Cristalografía por Rayos X , Dimerización , Eritropoyetina/metabolismo , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Mutación Puntual , Ingeniería de Proteínas , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/antagonistas & inhibidores , Alineación de Secuencia
10.
Nature ; 628(8006): 162-170, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38538791

RESUMEN

Ageing of the immune system is characterized by decreased lymphopoiesis and adaptive immunity, and increased inflammation and myeloid pathologies1,2. Age-related changes in populations of self-renewing haematopoietic stem cells (HSCs) are thought to underlie these phenomena3. During youth, HSCs with balanced output of lymphoid and myeloid cells (bal-HSCs) predominate over HSCs with myeloid-biased output (my-HSCs), thereby promoting the lymphopoiesis required for initiating adaptive immune responses, while limiting the production of myeloid cells, which can be pro-inflammatory4. Ageing is associated with increased proportions of my-HSCs, resulting in decreased lymphopoiesis and increased myelopoiesis3,5,6. Transfer of bal-HSCs results in abundant lymphoid and myeloid cells, a stable phenotype that is retained after secondary transfer; my-HSCs also retain their patterns of production after secondary transfer5. The origin and potential interconversion of these two subsets is still unclear. If they are separate subsets postnatally, it might be possible to reverse the ageing phenotype by eliminating my-HSCs in aged mice. Here we demonstrate that antibody-mediated depletion of my-HSCs in aged mice restores characteristic features of a more youthful immune system, including increasing common lymphocyte progenitors, naive T cells and B cells, while decreasing age-related markers of immune decline. Depletion of my-HSCs in aged mice improves primary and secondary adaptive immune responses to viral infection. These findings may have relevance to the understanding and intervention of diseases exacerbated or caused by dominance of the haematopoietic system by my-HSCs.


Asunto(s)
Inmunidad Adaptativa , Envejecimiento , Linaje de la Célula , Células Madre Hematopoyéticas , Linfocitos , Células Mieloides , Rejuvenecimiento , Animales , Femenino , Masculino , Ratones , Inmunidad Adaptativa/inmunología , Envejecimiento/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Inflamación/inmunología , Inflamación/patología , Linfocitos/citología , Linfocitos/inmunología , Linfopoyesis , Células Mieloides/citología , Células Mieloides/inmunología , Mielopoyesis , Fenotipo , Linfocitos T/citología , Linfocitos T/inmunología , Virus/inmunología
11.
Nature ; 630(8016): 457-465, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38750365

RESUMEN

Adoptively transferred T cells and agents designed to block the CD47-SIRPα axis are promising cancer therapeutics that activate distinct arms of the immune system1,2. Here we administered anti-CD47 antibodies in combination with adoptively transferred T cells with the goal of enhancing antitumour efficacy but observed abrogated therapeutic benefit due to rapid macrophage-mediated clearance of T cells expressing chimeric antigen receptors (CARs) or engineered T cell receptors. Anti-CD47-antibody-mediated CAR T cell clearance was potent and rapid enough to serve as an effective safety switch. To overcome this challenge, we engineered the CD47 variant CD47(Q31P) (47E), which engages SIRPα and provides a 'don't eat me' signal that is not blocked by anti-CD47 antibodies. TCR or CAR T cells expressing 47E are resistant to clearance by macrophages after treatment with anti-CD47 antibodies, and mediate substantial, sustained macrophage recruitment to the tumour microenvironment. Although many of the recruited macrophages manifested an M2-like profile3, the combined therapy synergistically enhanced antitumour efficacy. Our study identifies macrophages as major regulators of T cell persistence and illustrates the fundamental challenge of combining T-cell-directed therapeutics with those designed to activate macrophages. It delivers a therapeutic approach that is capable of simultaneously harnessing the antitumour effects of T cells and macrophages, offering enhanced potency against solid tumours.


Asunto(s)
Antígeno CD47 , Inmunoterapia Adoptiva , Neoplasias , Linfocitos T , Animales , Femenino , Humanos , Masculino , Ratones , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/metabolismo , Antígeno CD47/genética , Antígeno CD47/inmunología , Antígeno CD47/metabolismo , Línea Celular Tumoral , Inmunoterapia Adoptiva/métodos , Macrófagos/citología , Macrófagos/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Microambiente Tumoral/inmunología , Anticuerpos/inmunología , Anticuerpos/uso terapéutico , Activación de Macrófagos
12.
Nature ; 590(7844): 122-128, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33473210

RESUMEN

Ageing is characterized by the development of persistent pro-inflammatory responses that contribute to atherosclerosis, metabolic syndrome, cancer and frailty1-3. The ageing brain is also vulnerable to inflammation, as demonstrated by the high prevalence of age-associated cognitive decline and Alzheimer's disease4-6. Systemically, circulating pro-inflammatory factors can promote cognitive decline7,8, and in the brain, microglia lose the ability to clear misfolded proteins that are associated with neurodegeneration9,10. However, the underlying mechanisms that initiate and sustain maladaptive inflammation with ageing are not well defined. Here we show that in ageing mice myeloid cell bioenergetics are suppressed in response to increased signalling by the lipid messenger prostaglandin E2 (PGE2), a major modulator of inflammation11. In ageing macrophages and microglia, PGE2 signalling through its EP2 receptor promotes the sequestration of glucose into glycogen, reducing glucose flux and mitochondrial respiration. This energy-deficient state, which drives maladaptive pro-inflammatory responses, is further augmented by a dependence of aged myeloid cells on glucose as a principal fuel source. In aged mice, inhibition of myeloid EP2 signalling rejuvenates cellular bioenergetics, systemic and brain inflammatory states, hippocampal synaptic plasticity and spatial memory. Moreover, blockade of peripheral myeloid EP2 signalling is sufficient to restore cognition in aged mice. Our study suggests that cognitive ageing is not a static or irrevocable condition but can be reversed by reprogramming myeloid glucose metabolism to restore youthful immune functions.


Asunto(s)
Envejecimiento/metabolismo , Disfunción Cognitiva/prevención & control , Células Mieloides/metabolismo , Adulto , Anciano , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Animales , Respiración de la Célula , Células Cultivadas , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/genética , Dinoprostona/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Glucógeno/biosíntesis , Glucógeno/metabolismo , Humanos , Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Trastornos de la Memoria/tratamiento farmacológico , Ratones , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Mitocondrias/metabolismo , Células Mieloides/inmunología , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP2 de Receptores de Prostaglandina E/deficiencia , Subtipo EP2 de Receptores de Prostaglandina E/genética , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal/efectos de los fármacos , Memoria Espacial/efectos de los fármacos
13.
Nature ; 597(7877): 549-554, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34497417

RESUMEN

Monoclonal antibody therapies targeting tumour antigens drive cancer cell elimination in large part by triggering macrophage phagocytosis of cancer cells1-7. However, cancer cells evade phagocytosis using mechanisms that are incompletely understood. Here we develop a platform for unbiased identification of factors that impede antibody-dependent cellular phagocytosis (ADCP) using complementary genome-wide CRISPR knockout and overexpression screens in both cancer cells and macrophages. In cancer cells, beyond known factors such as CD47, we identify many regulators of susceptibility to ADCP, including the poorly characterized enzyme adipocyte plasma membrane-associated protein (APMAP). We find that loss of APMAP synergizes with tumour antigen-targeting monoclonal antibodies and/or CD47-blocking monoclonal antibodies to drive markedly increased phagocytosis across a wide range of cancer cell types, including those that are otherwise resistant to ADCP. Additionally, we show that APMAP loss synergizes with several different tumour-targeting monoclonal antibodies to inhibit tumour growth in mice. Using genome-wide counterscreens in macrophages, we find that the G-protein-coupled receptor GPR84 mediates enhanced phagocytosis of APMAP-deficient cancer cells. This work reveals a cancer-intrinsic regulator of susceptibility to antibody-driven phagocytosis and, more broadly, expands our knowledge of the mechanisms governing cancer resistance to macrophage phagocytosis.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/genética , Sistemas CRISPR-Cas , Citofagocitosis/genética , Macrófagos/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos de Neoplasias/inmunología , Antígeno CD47/antagonistas & inhibidores , Línea Celular Tumoral , Células Cultivadas , Femenino , Edición Génica , Técnicas de Inactivación de Genes , Humanos , Linfoma de Células T/inmunología , Linfoma de Células T/patología , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Receptores Acoplados a Proteínas G/metabolismo
14.
Nature ; 597(7875): 256-262, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34381212

RESUMEN

Loss of skeletal integrity during ageing and disease is associated with an imbalance in the opposing actions of osteoblasts and osteoclasts1. Here we show that intrinsic ageing of skeletal stem cells (SSCs)2 in mice alters signalling in the bone marrow niche and skews the differentiation of bone and blood lineages, leading to fragile bones that regenerate poorly. Functionally, aged SSCs have a decreased bone- and cartilage-forming potential but produce more stromal lineages that express high levels of pro-inflammatory and pro-resorptive cytokines. Single-cell RNA-sequencing studies link the functional loss to a diminished transcriptomic diversity of SSCs in aged mice, which thereby contributes to the transformation of the bone marrow niche. Exposure to a youthful circulation through heterochronic parabiosis or systemic reconstitution with young haematopoietic stem cells did not reverse the diminished osteochondrogenic activity of aged SSCs, or improve bone mass or skeletal healing parameters in aged mice. Conversely, the aged SSC lineage promoted osteoclastic activity and myeloid skewing by haematopoietic stem and progenitor cells, suggesting that the ageing of SSCs is a driver of haematopoietic ageing. Deficient bone regeneration in aged mice could only be returned to youthful levels by applying a combinatorial treatment of BMP2 and a CSF1 antagonist locally to fractures, which reactivated aged SSCs and simultaneously ablated the inflammatory, pro-osteoclastic milieu. Our findings provide mechanistic insights into the complex, multifactorial mechanisms that underlie skeletal ageing and offer prospects for rejuvenating the aged skeletal system.


Asunto(s)
Envejecimiento/patología , Huesos/patología , Senescencia Celular , Inflamación/patología , Nicho de Células Madre , Células Madre/patología , Animales , Proteína Morfogenética Ósea 2/metabolismo , Regeneración Ósea , Linaje de la Célula , Femenino , Curación de Fractura , Hematopoyesis , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Células Mieloides/citología , Osteoclastos/citología , Rejuvenecimiento
15.
Proc Natl Acad Sci U S A ; 121(5): e2318534121, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38261615

RESUMEN

The use of colony-stimulating factor-1 receptor (CSF1R) inhibitors has been widely explored as a strategy for cancer immunotherapy due to their robust depletion of tumor-associated macrophages (TAMs). While CSF1R blockade effectively eliminates TAMs from the solid tumor microenvironment, its clinical efficacy is limited. Here, we use an inducible CSF1R knockout model to investigate the persistence of tumor progression in the absence of TAMs. We find increased frequencies of granulocytic myeloid-derived suppressor cells (G-MDSCs) in the bone marrow, throughout circulation, and in the tumor following CSF1R deletion and loss of TAMs. We find that G-MDSCs are capable of suppressing macrophage phagocytosis, and the elimination of G-MDSCs through CXCR2 inhibition increases macrophage capacity for tumor cell clearance. Further, we find that combination therapy of CXCR2 inhibition and CD47 blockade synergize to elicit a significant anti-tumor response. These findings reveal G-MDSCs as key drivers of tumor immunosuppression and demonstrate their inhibition as a potent strategy to increase macrophage phagocytosis and enhance the anti-tumor efficacy of CD47 blockade in B16-F10 melanoma.


Asunto(s)
Melanoma Experimental , Células Supresoras de Origen Mieloide , Animales , Antígeno CD47 , Granulocitos , Macrófagos , Microambiente Tumoral , Ratones
16.
Nature ; 587(7835): 619-625, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33208946

RESUMEN

Although single-cell RNA sequencing studies have begun to provide compendia of cell expression profiles1-9, it has been difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here, using droplet- and plate-based single-cell RNA sequencing of approximately 75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, we create an extensive cell atlas of the human lung. We define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 out of 45 previously known cell types and 14 previously unknown ones. This comprehensive molecular atlas identifies the biochemical functions of lung cells and the transcription factors and markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signalling interactions and immune cell homing; and identifies cell types that are directly affected by lung disease genes and respiratory viruses. By comparing human and mouse data, we identified 17 molecular cell types that have been gained or lost during lung evolution and others with substantially altered expression profiles, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This atlas provides the molecular foundation for investigating how lung cell identities, functions and interactions are achieved in development and tissue engineering and altered in disease and evolution.


Asunto(s)
Células/clasificación , Células/metabolismo , Inmunidad , Pulmón/citología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcriptoma/genética , Anciano , Animales , Atlas como Asunto , Biomarcadores , Comunicación Celular , Células/inmunología , Quimiocinas/metabolismo , Células Endoteliales/metabolismo , Células Epiteliales/metabolismo , Femenino , Humanos , Pulmón/inmunología , Masculino , Ratones , Persona de Mediana Edad , Receptores Mensajeros de Linfocitos/metabolismo , Transducción de Señal , Células del Estroma/metabolismo
17.
Cell ; 142(5): 699-713, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20813259

RESUMEN

Monoclonal antibodies are standard therapeutics for several cancers including the anti-CD20 antibody rituximab for B cell non-Hodgkin lymphoma (NHL). Rituximab and other antibodies are not curative and must be combined with cytotoxic chemotherapy for clinical benefit. Here we report the eradication of human NHL solely with a monoclonal antibody therapy combining rituximab with a blocking anti-CD47 antibody. We identified increased expression of CD47 on human NHL cells and determined that higher CD47 expression independently predicted adverse clinical outcomes in multiple NHL subtypes. Blocking anti-CD47 antibodies preferentially enabled phagocytosis of NHL cells and synergized with rituximab. Treatment of human NHL-engrafted mice with anti-CD47 antibody reduced lymphoma burden and improved survival, while combination treatment with rituximab led to elimination of lymphoma and cure. These antibodies synergized through a mechanism combining Fc receptor (FcR)-dependent and FcR-independent stimulation of phagocytosis that might be applicable to many other cancers.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígeno CD47/inmunología , Linfoma no Hodgkin/inmunología , Linfoma no Hodgkin/terapia , Fagocitosis , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales de Origen Murino , Linfocitos B/inmunología , Línea Celular Tumoral , Humanos , Linfoma no Hodgkin/diagnóstico , Ratones , Receptores Fc/inmunología , Rituximab , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nature ; 572(7769): 392-396, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31367043

RESUMEN

Ovarian cancer and triple-negative breast cancer are among the most lethal diseases affecting women, with few targeted therapies and high rates of metastasis. Cancer cells are capable of evading clearance by macrophages through the overexpression of anti-phagocytic surface proteins called 'don't eat me' signals-including CD471, programmed cell death ligand 1 (PD-L1)2 and the beta-2 microglobulin subunit of the major histocompatibility class I complex (B2M)3. Monoclonal antibodies that antagonize the interaction of 'don't eat me' signals with their macrophage-expressed receptors have demonstrated therapeutic potential in several cancers4,5. However, variability in the magnitude and durability of the response to these agents has suggested the presence of additional, as yet unknown 'don't eat me' signals. Here we show that CD24 can be the dominant innate immune checkpoint in ovarian cancer and breast cancer, and is a promising target for cancer immunotherapy. We demonstrate a role for tumour-expressed CD24 in promoting immune evasion through its interaction with the inhibitory receptor sialic-acid-binding Ig-like lectin 10 (Siglec-10), which is expressed by tumour-associated macrophages. We find that many tumours overexpress CD24 and that tumour-associated macrophages express high levels of Siglec-10. Genetic ablation of either CD24 or Siglec-10, as well as blockade of the CD24-Siglec-10 interaction using monoclonal antibodies, robustly augment the phagocytosis of all CD24-expressing human tumours that we tested. Genetic ablation and therapeutic blockade of CD24 resulted in a macrophage-dependent reduction of tumour growth in vivo and an increase in survival time. These data reveal CD24 as a highly expressed, anti-phagocytic signal in several cancers and demonstrate the therapeutic potential for CD24 blockade in cancer immunotherapy.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Antígeno CD24/antagonistas & inhibidores , Inmunoterapia/métodos , Lectinas/metabolismo , Macrófagos/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Antígeno CD24/deficiencia , Antígeno CD24/genética , Antígeno CD24/inmunología , Línea Celular Tumoral , Humanos , Lectinas/antagonistas & inhibidores , Lectinas/genética , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Neoplasias/inmunología , Neoplasias/patología , Fagocitosis/efectos de los fármacos , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/genética , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología
19.
Nature ; 571(7764): 205-210, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31270459

RESUMEN

The mammalian brain contains neurogenic niches that comprise neural stem cells and other cell types. Neurogenic niches become less functional with age, but how they change during ageing remains unclear. Here we perform single-cell RNA sequencing of young and old neurogenic niches in mice. The analysis of 14,685 single-cell transcriptomes reveals a decrease in activated neural stem cells, changes in endothelial cells and microglia, and an infiltration of T cells in old neurogenic niches. T cells in old brains are clonally expanded and are generally distinct from those in old blood, which suggests that they may experience specific antigens. T cells in old brains also express interferon-γ, and the subset of neural stem cells that has a high interferon response shows decreased proliferation in vivo. We find that T cells can inhibit the proliferation of neural stem cells in co-cultures and in vivo, in part by secreting interferon-γ. Our study reveals an interaction between T cells and neural stem cells in old brains, opening potential avenues through which to counteract age-related decline in brain function.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/citología , Movimiento Celular , Células-Madre Neurales/citología , Neurogénesis , Análisis de la Célula Individual , Nicho de Células Madre/fisiología , Linfocitos T/citología , Animales , Sangre , Proliferación Celular , Células Clonales/citología , Técnicas de Cocultivo , Células Endoteliales/citología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Análisis de Secuencia de ARN , Transducción de Señal , Linfocitos T/metabolismo , Transcriptoma/genética
20.
Proc Natl Acad Sci U S A ; 119(29): e2203032119, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35858312

RESUMEN

Colonial tunicates are marine organisms that possess multiple brains simultaneously during their colonial phase. While the cyclical processes of neurogenesis and neurodegeneration characterizing their life cycle have been documented previously, the cellular and molecular changes associated with such processes and their relationship with variation in brain morphology and individual (zooid) behavior throughout adult life remains unknown. Here, we introduce Botryllus schlosseri as an invertebrate model for neurogenesis, neural degeneration, and evolutionary neuroscience. Our analysis reveals that during the weekly colony budding (i.e., asexual reproduction), prior to programmed cell death and removal by phagocytes, decreases in the number of neurons in the adult brain are associated with reduced behavioral response and significant change in the expression of 73 mammalian homologous genes associated with neurodegenerative disease. Similarly, when comparing young colonies (1 to 2 y of age) to those reared in a laboratory for ∼20 y, we found that older colonies contained significantly fewer neurons and exhibited reduced behavioral response alongside changes in the expression of 148 such genes (35 of which were differentially expressed across both timescales). The existence of two distinct yet apparently related neurodegenerative pathways represents a novel platform to study the gene products governing the relationship between aging, neural regeneration and degeneration, and loss of nervous system function. Indeed, as a member of an evolutionary clade considered to be a sister group of vertebrates, this organism may be a fundamental resource in understanding how evolution has shaped these processes across phylogeny and obtaining mechanistic insight.


Asunto(s)
Evolución Biológica , Enfermedades Neurodegenerativas , Urocordados , Animales , Expresión Génica , Enfermedades Neurodegenerativas/genética , Reproducción Asexuada , Urocordados/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA