Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38559095

RESUMO

Pulmonary fibrosis is a devastating disease with no effective treatments to cure, stop or reverse the unremitting, fatal fibrosis. A critical barrier to treating this disease is the lack of understanding of the pathways leading to fibrosis as well as those regulating the resolution of fibrosis. Fibrosis is the pathologic side of normal tissue repair that results when the normal wound healing programs go awry. Successful resolution of tissue injury requires several highly coordinated pathways, and this research focuses on the interplay between these overlapping pathways: immune effectors, inflammatory mediators and fibroproliferation in the resolution of fibrosis. Previously we have successfully prevented, mitigated, and even reversed established fibrosis using vaccinia vaccination immunotherapy in two models of murine lung fibrosis. The mechanism by which vaccinia reverses fibrosis is by vaccine induced lung specific Th1 skewed tissue resident memory (TRMs) in the lung. In this study, we isolated a population of vaccine induced TRMs - CD49a+ CD4+ T cells - that are both necessary and sufficient to reverse established pulmonary fibrosis. Using adoptive cellular therapy, we demonstrate that intratracheal administration of CD49a+ CD4+ TRMs into established fibrosis, reverses the fibrosis histologically, by promoting a decrease in collagen, and functionally, by improving lung function, without the need for vaccination. Furthermore, co-culture of in vitro derived CD49+ CD4+ human TRMs with human fibroblasts from individuals with idiopathic pulmonary fibrosis (IPF) results in the down regulation of IPF fibroblast collagen production. Lastly, we demonstrate in human IPF lung histologic samples that CD49a+ CD4+ TRMs, which can down regulate human IPF fibroblast function, fail to increase in the IPF lungs, thus potentially failing to promote resolution. Thus, we define a novel unappreciated role for tissue resident memory T cells in regulating established lung fibrosis to promote resolution of fibrosis and re-establish lung homeostasis. We demonstrate that immunotherapy, in the form of adoptive transfer of CD49a+ CD4+ TRMs into the lungs of mice with established fibrosis, not only stops progression of the fibrosis but more importantly reverses the fibrosis. These studies provide the insight and preclinical rationale for a novel paradigm shifting approach of using cellular immunotherapy to treat lung fibrosis.

2.
J Exp Med ; 221(6)2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38597954

RESUMO

Early stages of deadly respiratory diseases including COVID-19 are challenging to elucidate in humans. Here, we define cellular tropism and transcriptomic effects of SARS-CoV-2 virus by productively infecting healthy human lung tissue and using scRNA-seq to reconstruct the transcriptional program in "infection pseudotime" for individual lung cell types. SARS-CoV-2 predominantly infected activated interstitial macrophages (IMs), which can accumulate thousands of viral RNA molecules, taking over 60% of the cell transcriptome and forming dense viral RNA bodies while inducing host profibrotic (TGFB1, SPP1) and inflammatory (early interferon response, CCL2/7/8/13, CXCL10, and IL6/10) programs and destroying host cell architecture. Infected alveolar macrophages (AMs) showed none of these extreme responses. Spike-dependent viral entry into AMs used ACE2 and Sialoadhesin/CD169, whereas IM entry used DC-SIGN/CD209. These results identify activated IMs as a prominent site of viral takeover, the focus of inflammation and fibrosis, and suggest targeting CD209 to prevent early pathology in COVID-19 pneumonia. This approach can be generalized to any human lung infection and to evaluate therapeutics.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , Macrófagos , Inflamação , RNA Viral , Pulmão
3.
Nat Med ; 27(3): 546-559, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33654293

RESUMO

Angiotensin-converting enzyme 2 (ACE2) and accessory proteases (TMPRSS2 and CTSL) are needed for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cellular entry, and their expression may shed light on viral tropism and impact across the body. We assessed the cell-type-specific expression of ACE2, TMPRSS2 and CTSL across 107 single-cell RNA-sequencing studies from different tissues. ACE2, TMPRSS2 and CTSL are coexpressed in specific subsets of respiratory epithelial cells in the nasal passages, airways and alveoli, and in cells from other organs associated with coronavirus disease 2019 (COVID-19) transmission or pathology. We performed a meta-analysis of 31 lung single-cell RNA-sequencing studies with 1,320,896 cells from 377 nasal, airway and lung parenchyma samples from 228 individuals. This revealed cell-type-specific associations of age, sex and smoking with expression levels of ACE2, TMPRSS2 and CTSL. Expression of entry factors increased with age and in males, including in airway secretory cells and alveolar type 2 cells. Expression programs shared by ACE2+TMPRSS2+ cells in nasal, lung and gut tissues included genes that may mediate viral entry, key immune functions and epithelial-macrophage cross-talk, such as genes involved in the interleukin-6, interleukin-1, tumor necrosis factor and complement pathways. Cell-type-specific expression patterns may contribute to the pathogenesis of COVID-19, and our work highlights putative molecular pathways for therapeutic intervention.


Assuntos
COVID-19/epidemiologia , COVID-19/genética , Interações Hospedeiro-Patógeno/genética , SARS-CoV-2/fisiologia , Análise de Sequência de RNA/estatística & dados numéricos , Análise de Célula Única/estatística & dados numéricos , Internalização do Vírus , Adulto , Idoso , Idoso de 80 Anos ou mais , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/virologia , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/patologia , COVID-19/virologia , Catepsina L/genética , Catepsina L/metabolismo , Conjuntos de Dados como Assunto/estatística & dados numéricos , Demografia , Feminino , Perfilação da Expressão Gênica/estatística & dados numéricos , Humanos , Pulmão/metabolismo , Pulmão/virologia , Masculino , Pessoa de Meia-Idade , Especificidade de Órgãos/genética , Sistema Respiratório/metabolismo , Sistema Respiratório/virologia , Análise de Sequência de RNA/métodos , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Análise de Célula Única/métodos
4.
Nature ; 587(7835): 619-625, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33208946

RESUMO

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.


Assuntos
Células/classificação , Células/metabolismo , Imunidade , Pulmão/citologia , Análise de Sequência de RNA , Análise de Célula Única , Transcriptoma/genética , Idoso , Animais , Atlas como Assunto , Biomarcadores , Comunicação Celular , Células/imunologia , Quimiocinas/metabolismo , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Feminino , Humanos , Pulmão/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Receptores de Retorno de Linfócitos/metabolismo , Transdução de Sinais , Células Estromais/metabolismo
5.
Nat Genet ; 48(5): 537-43, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27019112

RESUMO

The hepatic vasculature is essential for liver development, homeostasis and regeneration, yet the developmental program of hepatic vessel formation and the embryonic origin of the liver vasculature remain unknown. Here we show in mouse that endocardial cells form a primitive vascular plexus surrounding the liver bud and subsequently contribute to a substantial portion of the liver vasculature. Using intersectional genetics, we demonstrate that the endocardium of the sinus venosus is a source for the hepatic plexus. Inhibition of endocardial angiogenesis results in reduced endocardial contribution to the liver vasculature and defects in liver organogenesis. We conclude that a substantial portion of liver vessels derives from the endocardium and shares a common developmental origin with coronary arteries.


Assuntos
Linhagem da Célula , Endocárdio/embriologia , Fígado/embriologia , Animais , Vasos Coronários/embriologia , Endocárdio/citologia , Endocárdio/metabolismo , Átrios do Coração/embriologia , Átrios do Coração/metabolismo , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Fígado/irrigação sanguínea , Circulação Hepática , Camundongos , Morfogênese , Fatores de Transcrição NFATC/metabolismo , Neovascularização Fisiológica
6.
Nat Med ; 21(8): 866-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26168292

RESUMO

Unraveling the fate specification of resident stem cells during lung regeneration is of clinical importance. It has been reported that c-kit(+) progenitor cells resident in the human lung regenerate epithelial lineages upon transplantation into injured mouse lung. Here we test the lineage potential of c-kit(+) cells by inducible genetic lineage tracing. We find that c-kit(+) cells do not contribute to lung epithelium during homeostasis and repair, and instead maintain a vascular endothelial cell fate. These findings call attention to the clinical application of c-kit(+) stem cells as lung epithelial progenitors for the treatment of pulmonary disease.


Assuntos
Células Endoteliais/fisiologia , Células Epiteliais/fisiologia , Pulmão/citologia , Proteínas Proto-Oncogênicas c-kit/análise , Animais , Linhagem da Célula , Feminino , Homeostase , Masculino , Camundongos , Células-Tronco/fisiologia
7.
Circulation ; 130(11 Suppl 1): S77-86, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25200059

RESUMO

BACKGROUND: Cell therapies offer the potential to improve cardiac function after myocardial infarction. Although injection of single-cell suspensions has proven safe, cell retention and survival rates are low. Tissue-engineered grafts allow cell delivery with minimal initial cell loss and mechanical support to the heart. However, graft performance cannot be easily compared, and optimal construct thickness, vascularization, and survival kinetics are unknown. METHODS AND RESULTS: Cardiac tissue slices (CTS) were generated by sectioning mouse hearts (n=40) expressing firefly luciferase and green fluorescent protein into slices of defined size and thickness using a vibrating blade microtome. Bioluminescence imaging of CTS transplanted onto hearts of immunodeficient mice demonstrated survival of ≤30% of transplanted cells. Cardiac slice perfusion was re-established within 3 days, likely through anastomosis of pre-existing vessels with the host vasculature and invasion of vessels from the host. Immunofluorescence showed a peak in cell death 3 days after transplantation and a gradual decline thereafter. MRI revealed preservation of contractile function and an improved ejection fraction 1 month after transplantation of CTS (28±2% CTS versus 22±2% control; P=0.05). Importantly, this effect was specific to CTS because transplantation of skeletal muscle tissue slices led to faster dilative remodeling and higher animal mortality. CONCLUSIONS: In summary, this is the first study to use CTS as a benchmark to validate and model tissue-engineered graft studies. CTS transplantation improved cell survival, established reperfusion, and enhanced cardiac function after myocardial infarction. These findings also confirm that dilative remodeling can be attenuated by topical transplantation of CTS but not skeletal muscle tissue grafts.


Assuntos
Células-Tronco Embrionárias/transplante , Ventrículos do Coração/transplante , Infarto do Miocárdio/cirurgia , Engenharia Tecidual , Transplante de Tecidos/métodos , Animais , Animais Recém-Nascidos , Feminino , Genes Reporter , Sobrevivência de Enxerto , Proteínas de Fluorescência Verde/genética , Humanos , Luciferases de Vaga-Lume/genética , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Modelos Animais , Contração Miocárdica , Tamanho do Órgão , Músculo Quadríceps , Distribuição Aleatória
8.
Methods Mol Biol ; 1074: 15-29, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23975802

RESUMO

Mouse postimplantation epiblast cultured in activin and basic fibroblast growth factor gives rise to continuously growing epiblast stem cells (EpiSCs) that share key properties with postimplantation epiblast, such as DNA methylation and an inactive X-chromosome. EpiSCs also show a distinct gene expression profile compared to embryonic stem cells (ESCs) derived from preimplantation blastocysts, and do not contribute efficiently to chimeras. EpiSCs can, however, revert to pluripotent ESC-like cells upon exposure to leukemia inhibitory factor-Stat3 signalling on feeder cells. Here we describe a protocol for the establishment of EpiSCs and their reversion to ESCs.


Assuntos
Blastocisto/citologia , Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Camadas Germinativas/citologia , Ativinas/farmacologia , Animais , Metilação de DNA/genética , Fibroblastos/citologia , Camundongos , Células-Tronco Pluripotentes/citologia , Transdução de Sinais
9.
J Cell Sci ; 125(Pt 24): 6094-104, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23077180

RESUMO

How cell fate becomes restricted during somatic cell differentiation is a long-lasting question in biology. Epigenetic mechanisms not present in pluripotent cells and acquired during embryonic development are expected to stabilize the differentiated state of somatic cells and thereby restrict their ability to convert to another fate. The histone variant macroH2A acts as a component of an epigenetic multilayer that heritably maintains the silent X chromosome and has been shown to restrict tumor development. Here we show that macroH2A marks the differentiated cell state during mouse embryogenesis. MacroH2A.1 was found to be present at low levels upon the establishment of pluripotency in the inner cell mass and epiblast, but it was highly enriched in the trophectoderm and differentiated somatic cells later in mouse development. Chromatin immunoprecipitation revealed that macroH2A.1 is incorporated in the chromatin of regulatory regions of pluripotency genes in somatic cells such as mouse embryonic fibroblasts and adult neural stem cells, but not in embryonic stem cells. Removal of macroH2A.1, macroH2A.2 or both increased the efficiency of induced pluripotency up to 25-fold. The obtained induced pluripotent stem cells reactivated pluripotency genes, silenced retroviral transgenes and contributed to chimeras. In addition, overexpression of macroH2A isoforms prevented efficient reprogramming of epiblast stem cells to naïve pluripotency. In summary, our study identifies for the first time a link between an epigenetic mark and cell fate restriction during somatic cell differentiation, which helps to maintain cell identity and antagonizes induction of a pluripotent stem cell state.


Assuntos
Células-Tronco Embrionárias/fisiologia , Histonas/metabolismo , Células-Tronco Pluripotentes/fisiologia , Animais , Diferenciação Celular/genética , Reprogramação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigenômica , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Histonas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transfecção
10.
Nucleus ; 2(6): 533-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22064467

RESUMO

How various epigenetic mechanisms restrict chromatin plasticity to determine the stability of repressed genes is poorly understood. Nuclear transfer to Xenopus oocytes induces the transcriptional reactivation of previously silenced genes. Recent work suggests that it can be used to analyze the epigenetic stability of repressed states. The notion that the epigenetic state of genes is an important determinant of the efficiency of nuclear reprogramming is supported by the differential reprogramming of given genes from different starting epigenetic configurations. After nuclear transfer, transcription from the inactive X chromosome of post-implantation-derived epiblast stem cells is reactivated. However, the same chromosome is resistant to reactivation when embryonic fibroblasts are used. Here, we discuss different kinds of evidence that link the histone variant macroH2A to the increased stability of repressed states. We focus on developmentally regulated X chromosome inactivation and repression of autosomal pluripotency genes, where macroH2A may help maintain the long-term stability of the differentiated state of somatic cells.


Assuntos
Epigênese Genética/fisiologia , Histonas/metabolismo , Técnicas de Transferência Nuclear , Oócitos/metabolismo , Proteínas Repressoras/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Histonas/genética , Oócitos/citologia , Proteínas Repressoras/genética , Transcrição Gênica/fisiologia , Cromossomo X/genética , Cromossomo X/metabolismo , Proteínas de Xenopus/genética , Xenopus laevis
11.
Nature ; 461(7268): 1292-5, 2009 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-19816418

RESUMO

The pluripotent state, which is first established in the primitive ectoderm cells of blastocysts, is lost progressively and irreversibly during subsequent development. For example, development of post-implantation epiblast cells from primitive ectoderm involves significant transcriptional and epigenetic changes, including DNA methylation and X chromosome inactivation, which create a robust epigenetic barrier and prevent their reversion to a primitive-ectoderm-like state. Epiblast cells are refractory to leukaemia inhibitory factor (LIF)-STAT3 signalling, but they respond to activin/basic fibroblast growth factor to form self-renewing epiblast stem cells (EpiSCs), which exhibit essential properties of epiblast cells and that differ from embryonic stem (ES) cells derived from primitive ectoderm. Here we show reprogramming of advanced epiblast cells from embryonic day 5.5-7.5 mouse embryos with uniform expression of N-cadherin and inactive X chromosome to ES-cell-like cells (rESCs) in response to LIF-STAT3 signalling. Cultured epiblast cells overcome the epigenetic barrier progressively as they proceed with the erasure of key properties of epiblast cells, resulting in DNA demethylation, X reactivation and expression of E-cadherin. The accompanying changes in the transcriptome result in a loss of phenotypic and epigenetic memory of epiblast cells. Using this approach, we report reversion of established EpiSCs to rESCs. Moreover, unlike epiblast and EpiSCs, rESCs contribute to somatic tissues and germ cells in chimaeras. Further studies may reveal how signalling-induced epigenetic reprogramming may promote reacquisition of pluripotency.


Assuntos
Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Camadas Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Biomarcadores/metabolismo , Caderinas/metabolismo , Células Cultivadas , Reprogramação Celular , Metilação de DNA , Ectoderma/citologia , Embrião de Mamíferos/citologia , Perfilação da Expressão Gênica , Camadas Germinativas/metabolismo , Fator Inibidor de Leucemia/metabolismo , Camundongos , Fator de Transcrição STAT3/metabolismo , Cromossomo Y/genética
12.
J Cell Biol ; 186(1): 27-40, 2009 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-19596847

RESUMO

Myc-induced SUN domain-containing protein (Misu or NSun2) is a nucleolar RNA methyltransferase important for c-Myc-induced proliferation in skin, but the mechanisms by which Misu contributes to cell cycle progression are unknown. In this study, we demonstrate that Misu translocates from the nucleoli in interphase to the spindle in mitosis as an RNA-protein complex that includes 18S ribosomal RNA. Functionally, depletion of Misu caused multiple mitotic defects, including formation of unstructured spindles, multipolar spindles, and chromosome missegregation, leading to aneuploidy and cell death. The presence of both RNA and Misu is required for correct spindle assembly, and this process is independent of active translation. Misu might mediate its function at the spindle by recruiting nucleolar and spindle-associated protein (NuSAP), an essential microtubule-stabilizing and bundling protein. We further identify NuSAP as a novel direct target gene of c-Myc. Collectively, our results suggest a novel mechanism by which c-Myc promotes proliferation by stabilizing the mitotic spindle in fast-dividing cells via Misu and NuSAP.


Assuntos
Nucléolo Celular/enzimologia , Metiltransferases/metabolismo , Fuso Acromático/enzimologia , Animais , Apoptose , Linhagem Celular Tumoral , Humanos , Imunoprecipitação , Interfase , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos , Microtúbulos/enzimologia , Mitose , Necrose , Ligação Proteica , Biossíntese de Proteínas , Transporte Proteico , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Ribossômico 18S/metabolismo , Tubulina (Proteína)/metabolismo
13.
Tissue Eng Part A ; 15(7): 1843-54, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19125642

RESUMO

Cell banking of mesenchymal stem cells (SCs) from various human tissues has significantly increased the feasibility of SC-based therapies. Sources such as adipose tissue and amnion offer outstanding possibilities for allogeneic transplantation due to their high differentiation potential and their ability to modulate immune reaction. Limitations, however, concern the reduced replicative potential as a result of progressive telomere erosion, which hampers scaleable production and long-term analysis of these cells. Here we report the establishment and characterization of two human amnion-derived and two human adipose-derived SC lines immortalized by ectopic expression of the catalytic subunit of human telomerase (hTERT). hTERT overexpression resulted in continuously growing SC lines that were largely unaltered concerning surface marker profile, morphology, karyotype, and immunosuppressive capacity with similar or enhanced differentiation potential for up to 87 population doublings. While all generated lines showed equal immunomodulation compared to the parental cells, one of the amnion-derived immortalized lines resulted in significantly increased immunogenicity. Although telomerase proves as important tool for immortalizing cells, our data emphasize the need for careful and standardized characterization of each individual cell population for cell banks.


Assuntos
Tecido Adiposo/citologia , Âmnio/citologia , Diferenciação Celular , Fatores Imunológicos/imunologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Telomerase/metabolismo , Adipogenia , Fosfatase Alcalina/metabolismo , Antígenos de Superfície/metabolismo , Contagem de Células , Linhagem Celular Transformada , Proliferação de Células , Forma Celular , Humanos , Cariotipagem , Leucócitos Mononucleares/imunologia , Células-Tronco Mesenquimais/enzimologia , Neoplasias/patologia , Osteocalcina/metabolismo , Osteogênese , PPAR gama/metabolismo , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA