Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Annu Rev Immunol ; 34: 449-78, 2016 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-27168243

RESUMO

Hematopoietic stem cells (HSCs) and downstream progenitors have long been studied based on phenotype, cell purification, proliferation, and transplantation into myeloablated recipients. These experiments, complemented by data on expression profiles, mouse mutants, and humans with hematopoietic defects, are the foundation for the current hematopoietic differentiation tree. However, there are fundamental gaps in our knowledge of the quantitative and qualitative operation of the HSC/progenitor system under physiological and pathological conditions in vivo. The hallmarks of HSCs, self-renewal and multipotency, are observed in in vitro assays and cell transplantation experiments; however, the extent to which these features occur naturally in HSCs and progenitors remains uncertain. We focus here on work that strives to address these unresolved questions, with emphasis on fate mapping and modeling of the hematopoietic flow from stem cells toward myeloid and lymphoid lineages during development and adult life.


Assuntos
Envelhecimento/imunologia , Diferenciação Celular , Hematopoese , Células-Tronco Hematopoéticas/fisiologia , Células Progenitoras Linfoides/fisiologia , Animais , Linhagem da Célula , Autorrenovação Celular , Humanos , Camundongos , Modelos Teóricos , Transcriptoma
3.
Nature ; 548(7668): 456-460, 2017 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-28813413

RESUMO

Developmental deconvolution of complex organs and tissues at the level of individual cells remains challenging. Non-invasive genetic fate mapping has been widely used, but the low number of distinct fluorescent marker proteins limits its resolution. Much higher numbers of cell markers have been generated using viral integration sites, viral barcodes, and strategies based on transposons and CRISPR-Cas9 genome editing; however, temporal and tissue-specific induction of barcodes in situ has not been achieved. Here we report the development of an artificial DNA recombination locus (termed Polylox) that enables broadly applicable endogenous barcoding based on the Cre-loxP recombination system. Polylox recombination in situ reaches a practical diversity of several hundred thousand barcodes, allowing tagging of single cells. We have used this experimental system, combined with fate mapping, to assess haematopoietic stem cell (HSC) fates in vivo. Classical models of haematopoietic lineage specification assume a tree with few major branches. More recently, driven in part by the development of more efficient single-cell assays and improved transplantation efficiencies, different models have been proposed, in which unilineage priming may occur in mice and humans at the level of HSCs. We have introduced barcodes into HSC progenitors in embryonic mice, and found that the adult HSC compartment is a mosaic of embryo-derived HSC clones, some of which are unexpectedly large. Most HSC clones gave rise to multilineage or oligolineage fates, arguing against unilineage priming, and suggesting coherent usage of the potential of cells in a clone. The spreading of barcodes, both after induction in embryos and in adult mice, revealed a basic split between common myeloid-erythroid development and common lymphocyte development, supporting the long-held but contested view of a tree-like haematopoietic structure.


Assuntos
Sítios de Ligação Microbiológicos/genética , Linhagem da Célula/genética , Rastreamento de Células/métodos , Código de Barras de DNA Taxonômico/métodos , Células-Tronco Hematopoéticas/citologia , Recombinação Genética/genética , Análise de Célula Única/métodos , Animais , Células Clonais/citologia , Células Clonais/metabolismo , Embrião de Mamíferos/citologia , Células Eritroides/citologia , Células Eritroides/metabolismo , Feminino , Células-Tronco Hematopoéticas/metabolismo , Integrases/metabolismo , Linfócitos/citologia , Linfócitos/metabolismo , Masculino , Camundongos , Mosaicismo , Células Mieloides/citologia , Células Mieloides/metabolismo
4.
Nature ; 518(7540): 542-6, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25686605

RESUMO

Haematopoietic stem cells (HSCs) are widely studied by HSC transplantation into immune- and blood-cell-depleted recipients. Single HSCs can rebuild the system after transplantation. Chromosomal marking, viral integration and barcoding of transplanted HSCs suggest that very low numbers of HSCs perpetuate a continuous stream of differentiating cells. However, the numbers of productive HSCs during normal haematopoiesis, and the flux of differentiating progeny remain unknown. Here we devise a mouse model allowing inducible genetic labelling of the most primitive Tie2(+) HSCs in bone marrow, and quantify label progression along haematopoietic development by limiting dilution analysis and data-driven modelling. During maintenance of the haematopoietic system, at least 30% or ∼5,000 HSCs are productive in the adult mouse after label induction. However, the time to approach equilibrium between labelled HSCs and their progeny is surprisingly long, a time scale that would exceed the mouse's life. Indeed, we find that adult haematopoiesis is largely sustained by previously designated 'short-term' stem cells downstream of HSCs that nearly fully self-renew, and receive rare but polyclonal HSC input. By contrast, in fetal and early postnatal life, HSCs are rapidly used to establish the immune and blood system. In the adult mouse, 5-fluoruracil-induced leukopenia enhances the output of HSCs and of downstream compartments, thus accelerating haematopoietic flux. Label tracing also identifies a strong lineage bias in adult mice, with several-hundred-fold larger myeloid than lymphoid output, which is only marginally accentuated with age. Finally, we show that transplantation imposes severe constraints on HSC engraftment, consistent with the previously observed oligoclonal HSC activity under these conditions. Thus, we uncover fundamental differences between the normal maintenance of the haematopoietic system, its regulation by challenge, and its re-establishment after transplantation. HSC fate mapping and its linked modelling provide a quantitative framework for studying in situ the regulation of haematopoiesis in health and disease.


Assuntos
Linhagem da Célula/fisiologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Células-Tronco/citologia , Envelhecimento , Animais , Animais Recém-Nascidos , Transplante de Medula Óssea , Proliferação de Células , Rastreamento de Células , Feminino , Feto/citologia , Feto/embriologia , Fluoruracila , Células-Tronco Hematopoéticas/metabolismo , Masculino , Camundongos , Receptor TIE-2/metabolismo , Células-Tronco/metabolismo
5.
Nature ; 518(7540): 547-51, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25470051

RESUMO

Most haematopoietic cells renew from adult haematopoietic stem cells (HSCs), however, macrophages in adult tissues can self-maintain independently of HSCs. Progenitors with macrophage potential in vitro have been described in the yolk sac before emergence of HSCs, and fetal macrophages can develop independently of Myb, a transcription factor required for HSC, and can persist in adult tissues. Nevertheless, the origin of adult macrophages and the qualitative and quantitative contributions of HSC and putative non-HSC-derived progenitors are still unclear. Here we show in mice that the vast majority of adult tissue-resident macrophages in liver (Kupffer cells), brain (microglia), epidermis (Langerhans cells) and lung (alveolar macrophages) originate from a Tie2(+) (also known as Tek) cellular pathway generating Csf1r(+) erythro-myeloid progenitors (EMPs) distinct from HSCs. EMPs develop in the yolk sac at embryonic day (E) 8.5, migrate and colonize the nascent fetal liver before E10.5, and give rise to fetal erythrocytes, macrophages, granulocytes and monocytes until at least E16.5. Subsequently, HSC-derived cells replace erythrocytes, granulocytes and monocytes. Kupffer cells, microglia and Langerhans cells are only marginally replaced in one-year-old mice, whereas alveolar macrophages may be progressively replaced in ageing mice. Our fate-mapping experiments identify, in the fetal liver, a sequence of yolk sac EMP-derived and HSC-derived haematopoiesis, and identify yolk sac EMPs as a common origin for tissue macrophages.


Assuntos
Linhagem da Célula , Eritrócitos/citologia , Hematopoese , Macrófagos/citologia , Células-Tronco/citologia , Saco Vitelino/citologia , Animais , Proliferação de Células , Rastreamento de Células , Feminino , Feto/citologia , Granulócitos/citologia , Células de Kupffer/citologia , Células de Langerhans/citologia , Fígado/citologia , Fígado/embriologia , Macrófagos Alveolares/citologia , Masculino , Camundongos , Microglia/citologia , Monócitos/citologia , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Receptor TIE-2/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo
6.
Blood ; 129(4): 415-419, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-27903529

RESUMO

Microvascular endothelial cells (ECs) display a high degree of phenotypic and functional heterogeneity among different organs. Organ-specific ECs control their tissue microenvironment by angiocrine factors in health and disease. Liver sinusoidal endothelial cells (LSECs) are uniquely differentiated to fulfill important organ-specific functions in development, under homeostatic conditions, and in regeneration and liver pathology. Recently, Bmp2 has been identified by us as an organ-specific angiokine derived from LSECs. To study angiocrine Bmp2 signaling in the liver, we conditionally deleted Bmp2 in LSECs using EC subtype-specific Stab2-Cre mice. Genetic inactivation of hepatic angiocrine Bmp2 signaling in Stab2-Cre;Bmp2fl/fl (Bmp2LSECKO) mice caused massive iron overload in the liver and increased serum iron levels and iron deposition in several organs similar to classic hereditary hemochromatosis. Iron overload was mediated by decreased hepatic expression of hepcidin, a key regulator of iron homeostasis. Thus, angiocrine Bmp2 signaling within the hepatic vascular niche represents a constitutive pathway indispensable for iron homeostasis in vivo that is nonredundant with Bmp6. Notably, we demonstrate that organ-specific angiocrine signaling is essential not only for the homeostasis of the respective organ but also for the homeostasis of the whole organism.


Assuntos
Proteína Morfogenética Óssea 2/genética , Células Endoteliais/metabolismo , Hemocromatose/genética , Hepcidinas/genética , Homeostase/genética , Ferro/metabolismo , Fígado/metabolismo , Animais , Proteína Morfogenética Óssea 2/deficiência , Proteína Morfogenética Óssea 6/genética , Proteína Morfogenética Óssea 6/metabolismo , Capilares/metabolismo , Capilares/patologia , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Células Endoteliais/patologia , Feminino , Regulação da Expressão Gênica , Hemocromatose/metabolismo , Hemocromatose/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepcidinas/metabolismo , Integrases/genética , Integrases/metabolismo , Fígado/irrigação sanguínea , Fígado/patologia , Masculino , Camundongos , Camundongos Transgênicos , Comunicação Parácrina , Transdução de Sinais , Transcrição Gênica
8.
J Pathol ; 229(5): 775-83, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23341364

RESUMO

Burkitt lymphoma (BL) is caused by translocation of the MYC gene to an immunoglobulin locus resulting in its constitutive expression depending on the activity of the immunoglobulin (Ig) enhancer elements. Treatment of BL cell lines with epigenetic modifiers is known to repress B-cell-specific genes and to up-regulate B-cell-inappropriate genes including the transcription repressor ID2 expression. We found that the DNA methyltransferase inhibitor decitabine/5-aza-2-deoxycytidine (5-aza-dC) represses the MYC oncogene on RNA and protein levels by inducing ID2. Down-regulation of MYC was associated with repression of transcriptional activity of the Ig locus and with inhibition of proliferation. The induction of ID2 can be in part explained by activation of the transcription factor NF-κB. We conclude that up-regulation of ID2 contributes to anti-tumour activity of 5-aza-dC via repression of Ig locus activity and consequently MYC expression.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Linfoma de Burkitt/genética , Metilases de Modificação do DNA/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/genética , Translocação Genética/efeitos dos fármacos , Azacitidina/farmacologia , Linfoma de Burkitt/enzimologia , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Decitabina , Relação Dose-Resposta a Droga , Repressão Epigenética , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoglobulina M/genética , Imunoglobulina M/metabolismo , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transcrição Gênica , Transfecção , Regulação para Cima
9.
Blood ; 114(12): 2448-58, 2009 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-19628709

RESUMO

Deregulated c-MYC is found in a variety of cancers where it promotes proliferation as well as apoptosis. In many hematologic malignancies, enhanced NF-kappaB exerts prosurvival functions. Here we investigated the role of NF-kappaB in mouse and human c-MYC-transformed lymphomas. The NF-kappaB pathway is extinguished in murine lymphoma cells, and extrinsic stimuli typically inducing NF-kappaB activity fail to activate this pathway. Genetic activation of the NF-kappaB pathway induces apoptosis in these cells, whereas inhibition of NF-kappaB by an IkappaBalpha superrepressor provides a selective advantage in vivo. Furthermore, in human Burkitt lymphoma cells we find that NF-kappaB activation induces apoptosis. NF-kappaB up-regulates Fas and predisposes to Fas-induced cell death, which is caspase-8 mediated and can be prevented by CFLAR overexpression. We conclude that c-MYC overexpression sensitizes cells to NF-kappaB-induced apoptosis, and persistent inactivity of NF-kappaB signaling is a prerequisite for MYC-mediated tumorigenesis. We could also show that low immunogenicity and Fas insensitivity of MYC-driven lymphoma cells are reversed by activation of NF-kappaB. Our observations provide a molecular explanation for the described absence of the NF-kappaB signaling in Burkitt lymphoma and question the applicability of NF-kappaB inhibitors as candidates for treatment of this cancer.


Assuntos
Apoptose , Linfoma de Burkitt/patologia , Quinase I-kappa B/fisiologia , Linfoma de Células B/patologia , NF-kappa B/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Animais , Western Blotting , Linfoma de Burkitt/metabolismo , Caspases/metabolismo , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Ensaio de Desvio de Mobilidade Eletroforética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Linfoma de Células B/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Transfecção
10.
Oncoimmunology ; 10(1): 1906500, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-34026331

RESUMO

Only a small subset of colorectal cancer (CRC) patients benefits from immunotherapies, comprising blocking antibodies (Abs) against checkpoint receptor "programmed-cell-death-1" (PD1) and its ligand (PD-L1), because most cases lack the required mutational burden and neo-antigen load caused by microsatellite instability (MSI) and/or an inflamed, immune cell-infiltrated PD-L1+ tumor microenvironment. Peroxisome proliferator-activated-receptor-gamma (PPARγ), a metabolic transcription factor stimulated by anti-diabetic drugs, has been previously implicated in pre/clinical responses to immunotherapy. We therefore raised the hypothesis that PPARγ induces PD-L1 on microsatellite stable (MSS) tumor cells to enhance Ab-target engagement and responsiveness to PD-L1 blockage. We found that PPARγ-agonists upregulate PD-L1 mRNA/protein expression in human gastrointestinal cancer cell lines and MSS+ patient-derived tumor organoids (PDOs). Mechanistically, PPARγ bound to and activated DNA-motifs similar to cognate PPARγ-responsive-elements (PPREs) in the proximal -2 kb promoter of the human PD-L1 gene. PPARγ-agonist reduced proliferation and viability of tumor cells in co-cultures with PD-L1 blocking Ab and lymphokine-activated killer cells (LAK) derived from the peripheral blood of CRC patients or healthy donors. Thus, metabolic modifiers improved the antitumoral response of immune checkpoint Ab, proposing novel therapeutic strategies for CRC.


Assuntos
Neoplasias Colorretais , PPAR gama , Antígeno B7-H1/genética , Neoplasias Colorretais/tratamento farmacológico , Humanos , Instabilidade de Microssatélites , PPAR gama/genética , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA