Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
1.
Int J Mol Sci ; 22(7)2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33810280

RESUMO

The pathophysiology of preeclampsia (PE) is poorly understood; however, there is a large body of evidence that suggests a role of immune cells in the development of PE. Amongst these, B cells are a dominant element in the pathogenesis of PE, and they have been shown to play an important role in various immune-mediated diseases, both as pro-inflammatory and regulatory cells. Perinatal cells are defined as cells from birth-associated tissues isolated from term placentas and fetal annexes and more specifically from the amniotic membrane, chorionic membrane, chorionic villi, umbilical cord (including Wharton's jelly), the basal plate, and the amniotic fluid. They have drawn particular attention in recent years due to their ability to modulate several aspects of immunity, making them promising candidates for the prevention and treatment of various immune-mediated diseases. In this review we describe main findings regarding the multifaceted in vitro and in vivo immunomodulatory properties of perinatal cells, with a focus on B lymphocytes. Indeed, we discuss evidence on the ability of perinatal cells to inhibit B cell proliferation, impair B cell differentiation, and promote regulatory B cell formation. Therefore, the findings discussed herein unveil the possibility to modulate B cell activation and function by exploiting perinatal immunomodulatory properties, thus possibly representing a novel therapeutic strategy in PE.


Assuntos
Linfócitos B/imunologia , Células-Tronco Embrionárias/transplante , Pré-Eclâmpsia/imunologia , Animais , Células-Tronco Embrionárias/imunologia , Feminino , Humanos , Pré-Eclâmpsia/terapia , Gravidez , Transplante de Células-Tronco/métodos
2.
Reproduction ; 160(4): 547-560, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32698161

RESUMO

Recent studies have demonstrated that embryonic stem cells (ESCs) have an underdeveloped innate immune system, but the biological implications of this finding are poorly understood. In this study, we compared the responses of mouse ESCs (mESCs) and mESC differentiated fibroblasts (mESC-FBs) to tumor necrosis factor α (TNFα) and interferons (IFNs). Our data revealed that TNFα, IFNα, IFNß, or IFNγ alone do not cause apparent effects on mESCs and mESC-FBs, but the combination of TNFα and IFNγ (TNFα/IFNγ) showed toxicity to mESC-FBs as indicated by cell cycle inhibition and reduced cell viability, correlating with the expression of inducible nitric oxide synthase (iNOS). However, none of these effects were observed in mESCs that were treated with TNFα/IFNγ. Furthermore, mESC-FBs, but not mESCs, are vulnerable to cytotoxicity resulting from lipopolysaccharide (LPS)-activated macrophages. The insensitivity of mESCs to cytotoxicity in all cases is correlated with their lack of responses to TNFα and IFNγ. Similar to mESCs, human ESCs (hESCs) and iPSCs (hiPSCs) do not respond to TNFα and are not susceptible to the cytotoxicity of TNFα, IFNß, or IFNγ alone or in combination that significantly affects human foreskin fibroblast (hFBs) and Hela cells. However, unlike mESCs, hESCs and hiPSCs can respond to IFNγ, but this does not cause significant cytotoxicity in hESCs and hiPSCs. Our findings in both mouse and human PSCs together support the hypothesis that attenuated innate immune responses could be a protective mechanism that limits immunologic cytotoxicity resulting from inflammatory and immune responses.


Assuntos
Citotoxicidade Imunológica/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Interferon gama/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Fibroblastos/citologia , Fibroblastos/imunologia , Células HeLa , Humanos , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/imunologia
3.
Front Immunol ; 11: 594594, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33633725

RESUMO

The laboratory rat is widely used as a model for human diseases. Many of these diseases involve monocytes and tissue macrophages in different states of activation. Whilst methods for in vitro differentiation of mouse macrophages from embryonic stem cells (ESC) and bone marrow (BM) are well established, these are lacking for the rat. The gene expression profiles of rat macrophages have also not been characterised to the same extent as mouse. We have established the methodology for production of rat ESC-derived macrophages and compared their gene expression profiles to macrophages obtained from the lung and peritoneal cavity and those differentiated from BM and blood monocytes. We determined the gene signature of Kupffer cells in the liver using rats deficient in macrophage colony stimulating factor receptor (CSF1R). We also examined the response of BM-derived macrophages to lipopolysaccharide (LPS). The results indicate that many, but not all, tissue-specific adaptations observed in mice are conserved in the rat. Importantly, we show that unlike mice, rat macrophages express the CSF1R ligand, colony stimulating factor 1 (CSF1).


Assuntos
Células-Tronco Embrionárias/imunologia , Perfilação da Expressão Gênica , Macrófagos Alveolares/imunologia , Macrófagos Peritoneais/imunologia , Transcriptoma/imunologia , Animais , Masculino , Ratos , Ratos Mutantes
4.
J Leukoc Biol ; 107(6): 1155-1166, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31841228

RESUMO

The majority of genetically modified C57BL/6 mice contain congenic passenger DNA around the targeted gene locus as they were generated from 129-derived embryonic stem cells (ESCs) with subsequent backcrossing to the C57BL/6 genetic background. When studying the role of atypical chemokine receptor 4 (ACKR4) in the immune system, we realized that the two available Ackr4-deficient mouse strains (Ackr4-/- and Ackr4GFP/GFP ) show profoundly different phenotypes: Compared to wild-type and Ackr4GFP/GFP mice, Ackr4-/- mice show a strong accumulation of plasma blasts in mesenteric lymph node and spleen as well as increased B cell proliferation after in vitro activation. This phenotype was maintained after further backcrossing to C57BL/6 mice and was even present in heterozygous Ackr4+/- animals, suggesting that a gene variant on the targeted chromosome might cause this phenotype. Exome sequencing revealed that a region of approximately 20 Mbp around the Ackr4 locus on chromosome 9 still originates from the 129 background based on high variant density observed. In activated Ackr4-/- and Ackr4GFP/GFP B cells, transcripts of genes around the Ackr4 locus were equally deregulated compared to C57BL/6 B cells, whereas increased expression of IL-6 was selectively observed in B cells of Ackr4-/- mice. Because the gene encoding for IL-6 is placed on chromosome 5 these findings suggest that passenger DNA around the Ackr4 locus has an indirect effect on B cell activation and IL-6 production. Results of the present study should not only lead to the reinterpretation of data from earlier studies using Ackr4-/- mice but should remind the scientific community about the limitations of mouse models using mice created by gene-targeting of nonsyngeneic ESCs.


Assuntos
Linfócitos B/imunologia , Cromossomos de Mamíferos/imunologia , Interleucina-6/genética , Linfonodos/imunologia , Ativação Linfocitária , Receptores CCR/genética , Animais , Linfócitos B/citologia , Proliferação de Células , Cruzamentos Genéticos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Feminino , Genes Reporter , Loci Gênicos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Heterozigoto , Homozigoto , Interleucina-6/imunologia , Linfonodos/citologia , Masculino , Mesentério/citologia , Mesentério/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Receptores CCR/deficiência , Receptores CCR/imunologia , Baço/citologia , Baço/imunologia , Sequenciamento do Exoma
5.
Cell Stem Cell ; 25(5): 594-606, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31703770

RESUMO

Although the clinical outcomes of cell therapy trials have not met initial expectations, emerging evidence suggests that injury-mediated tissue damage might benefit from the delivery of cells or their secreted products. Pluripotent stem cells (PSCs) are promising cell sources primarily because of their capacity to generate stage- and lineage-specific differentiated derivatives. However, they carry inherent challenges for safe and efficacious clinical translation. This Review describes completed or ongoing trials of PSCs, discusses their potential mechanisms of action, and considers how to address the challenges required for them to become a major therapy, using heart repair as a case study.


Assuntos
Sobrevivência Celular/imunologia , Células-Tronco Pluripotentes Induzidas/transplante , Transplante de Células-Tronco/efeitos adversos , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/transplante , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/terapia , Humanos , Terapia de Imunossupressão , Células-Tronco Pluripotentes Induzidas/imunologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Degeneração Macular/imunologia , Degeneração Macular/terapia , Doença de Parkinson/imunologia , Doença de Parkinson/terapia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco/métodos , Transplante Autólogo/efeitos adversos , Transplante Autólogo/métodos , Transplante Homólogo/efeitos adversos , Transplante Homólogo/métodos
6.
Mol Biol (Mosk) ; 53(5): 725-740, 2019.
Artigo em Russo | MEDLINE | ID: mdl-31661474

RESUMO

Human pluripotent stem cells, which include embryonic stem cells and induced pluripotent cells (iPSCs), are capable of unlimited division and differentiation into all cells of the body. These cells are considered as a potential source of various types of cells for transplantations. The use of autologous iPSCs is not potentially associated with immune rejection and does not require immunosuppression required for allogeneic grafts. However, the high cost of this technology and the duration of obtaining iPSCs and differentiated cells may limit the use of autologous iPSCs in clinical practice. In addition, full equivalence and immunological compatibility of autologous iPSCs and their derivatives have been repeatedly questioned. One approach to solving the problem of the immunological compatibility of allogeneic derivatives of iPSCs can be the establishment of cell lines with reduced immunogenicity. Differentiated derivatives of such iPSCs may be suitable for transplantation to any patient. This review discusses the strategies for evading immune surveillance in normal and tumor processes that can be used to establish stem cell lines with reduced immunogenicity.


Assuntos
Linhagem Celular/citologia , Linhagem Celular/imunologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/imunologia , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Humanos
7.
Front Immunol ; 10: 1510, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31338094

RESUMO

Over a century ago, it was reported that immunization with embryonic/fetal tissue could lead to the rejection of transplanted tumors in animals. Subsequent studies demonstrated that vaccination of embryonic materials in animals induced cellular and humoral immunity against transplantable tumors and carcinogen-induced tumors. Therefore, it has been hypothesized that the shared antigens between tumors and embryonic/fetal tissues (oncofetal antigens) are the key to anti-tumor immune responses in these studies. However, early oncofetal antigen-based cancer vaccines usually utilize xenogeneic or allogeneic embryonic stem cells or tissues, making it difficult to tease apart the anti-tumor immunity elicited by the oncofetal antigens vs. graft-vs.-host responses. Recently, one oncofetal antigen-based cancer vaccine using autologous induced pluripotent stem cells (iPSCs) demonstrated marked prophylactic and therapeutic potential, suggesting critical roles of oncofetal antigens in inducing anti-tumor immunity. In this review, we present an overview of recent studies in the field of oncofetal antigen-based cancer vaccines, including single peptide-based cancer vaccines, embryonic stem cell (ESC)- and iPSC-based whole-cell vaccines, and provide insights on future directions.


Assuntos
Vacinas Anticâncer/imunologia , Células-Tronco Pluripotentes Induzidas/imunologia , Animais , Antígenos de Neoplasias/imunologia , Células-Tronco Embrionárias/imunologia , Humanos , Vacinação/métodos
8.
Genes (Basel) ; 10(3)2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30897824

RESUMO

The discovery of mammalian pluripotent embryonic stem cells (ESC) has revolutionised cell research and regenerative medicine. More recently discovered chicken ESC (cESC), though less intensively studied, are increasingly popular as vaccine substrates due to a dearth of avian cell lines. Information on the comparative performance of cESC with common vaccine viruses is limited. Using RNA-sequencing, we compared cESC transcriptional programmes elicited by stimulation with chicken type I interferon or infection with vaccine viruses routinely propagated in primary chicken embryo fibroblasts (CEF). We used poxviruses (fowlpox virus (FWPV) FP9, canarypox virus (CNPV), and modified vaccinia virus Ankara (MVA)) and a birnavirus (infectious bursal disease virus (IBDV) PBG98). Interferon-stimulated genes (ISGs) were induced in cESC to levels comparable to those in CEF and immortalised chicken fibroblast DF-1 cells. cESC are permissive (with distinct host transcriptional responses) to MVA, FP9, and CNPV but, surprisingly, not to PBG98. MVA, CNPV, and FP9 suppressed innate immune responses, while PBG98 induced a subset of ISGs. Dysregulation of signalling pathways (i.e., NFκB, TRAF) was observed, which might affect immune responses and viral replication. In conclusion, we show that cESC are an attractive alternative substrate to study and propagate poxvirus recombinant vaccine vectors.


Assuntos
Células-Tronco Embrionárias/virologia , Perfilação da Expressão Gênica/veterinária , Redes Reguladoras de Genes , Poxviridae/imunologia , Animais , Células Cultivadas , Embrião de Galinha , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Regulação da Expressão Gênica , Interferon Tipo I/imunologia , Poxviridae/classificação , Análise de Sequência de RNA/veterinária , Especificidade da Espécie , Vacinas Virais/classificação , Vacinas Virais/imunologia
9.
Am J Transplant ; 19(6): 1652-1662, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30549427

RESUMO

Stem cell-based approaches have the potential to address the organ shortage in transplantation. Whereas both embryonic stem cells and induced pluripotent stem cells have been utilized as cellular sources for differentiation and lineage specification, their relative ability to be recognized by immune effector cells is unclear. We determined the expression of immune recognition molecules on hepatocyte-like cells (HLC) generated from murine embryonic stem cells and induced pluripotent stem cells, compared to adult hepatocytes, and we evaluated the impact on recognition by natural killer (NK) cells. We report that HLC lack MHC class I expression, and that embryonic stem cell-derived HLC have higher expression of the NK cell activating ligands Rae1, H60, and Mult1 than induced pluripotent stem cell-derived HLC and adult hepatocytes. Moreover, the lack of MHC class I renders embryonic stem cell-derived HLC, and induced pluripotent stem cell-derived HLC, susceptible to killing by syngeneic and allogeneic NK cells. Both embryonic stem cell-derived HLC, and induced pluripotent stem cell-derived HLC, are killed by NK cells at higher levels than adult hepatocytes. Finally, we demonstrate that the NK cell activation receptor, NKG2D, plays a key role in NK cell cytotoxicity of embryonic stem cell-derived HLC, but not induced pluripotent stem cell-derived HLC.


Assuntos
Células-Tronco Embrionárias/imunologia , Hepatócitos/imunologia , Hepatócitos/transplante , Células-Tronco Pluripotentes Induzidas/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Aloenxertos , Animais , Diferenciação Celular , Transplante de Células/métodos , Citotoxicidade Imunológica , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Perfilação da Expressão Gênica , Hepatócitos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Isoenxertos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Modelos Animais , Subfamília K de Receptores Semelhantes a Lectina de Células NK/deficiência , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética
10.
Curr Opin Pharmacol ; 43: 20-26, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30071348

RESUMO

Stem cells represent a potential candidate for ß cell replacement in type 1 diabetes. Pluripotent stem cells are able to differentiate in vitro into functional insulin producing cells, that can restore normoglycemia in diabetic mice. Clinical trials with embryonic stem cell-derived pancreatic progenitors are ongoing. Besides, induced pluripotent stem cells offer the chance of personalized cell therapy. So far, transition to the clinic still needs to face critical issues, such as immunogenicity and safety of stem cell derived ß cells. To this purpose, new strategies for immunoprotection, including micro and macro-encapsulation, but also gene editing approaches, are being explored.


Assuntos
Diabetes Mellitus Tipo 1/cirurgia , Células-Tronco Embrionárias/transplante , Células-Tronco Pluripotentes Induzidas/transplante , Células Secretoras de Insulina/metabolismo , Transplante das Ilhotas Pancreáticas/métodos , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/metabolismo , Sobrevivência de Enxerto , Humanos , Células-Tronco Pluripotentes Induzidas/imunologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Transplante das Ilhotas Pancreáticas/efeitos adversos , Fenótipo , Regeneração , Fatores de Risco , Transplante de Células-Tronco/efeitos adversos , Resultado do Tratamento
11.
Adv Exp Med Biol ; 1074: 641-647, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721998

RESUMO

Transplantation of potentially therapeutic cells into the subretinal space is a promising prospective therapy for the treatment of retinal degenerative diseases including age-related macular degeneration (AMD). In rodent models with photoreceptor degeneration, subretinal transplantation of cell suspensions has repeatedly been demonstrated to rescue behaviorally measured vision, maintain electrophysiological responses from the retina and the brain, and slow the degeneration of rod and cone photoreceptors for extended periods. These studies have led to the initiation of a number of FDA-approved clinical trials for application of cell-based therapy for AMD and other retinal degenerative diseases. However, translation from rodent models directly into human clinical trials skips an important intermediary preclinical step that is needed to address critical issues for intraocular cell transplantation. These include determination of the most appropriate and least problematic surgical approach, the application of treatment in an eye with similar size and structure including the presence of a macula, and a thorough understanding of the immunological considerations regarding graft survival and the consequences of grafted cell rejection. This chapter will review these and related issues and will document current efforts to address these concerns.


Assuntos
Modelos Animais , Primatas , Degeneração Retiniana/terapia , Roedores , Transplante de Células-Tronco/métodos , Animais , Tamanho Corporal , Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/transplante , Rejeição de Enxerto/imunologia , Terapia de Imunossupressão , Degeneração Macular/terapia , Especificidade da Espécie , Imunologia de Transplantes
12.
Parasit Vectors ; 10(1): 613, 2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29258559

RESUMO

BACKGROUND: It is well known that reactive oxygen species (ROS) and reactive nitrogen species (RNS) are involved in the control of pathogens and microbiota in insects. However, the knowledge of the role of ROS and RNS in tick-pathogen and tick-microbiota interactions is limited. Here, we evaluated the immune-related redox metabolism of the embryonic cell line BME26 from the cattle tick Rhipicephalus microplus in response to Anaplasma marginale infection. METHODS: A high-throughput qPCR approach was used to determine the expression profile of 16 genes encoding proteins involved in either production or detoxification of ROS and RNS in response to different microbial challenges. In addition, the effect of RNAi-mediated gene silencing of catalase, glutathione peroxidase, thioredoxin and protein oxidation resistance 1 in the control of infection with A. marginale was evaluated. RESULTS: Infection with A. marginale resulted in downregulation of the genes encoding ROS-generating enzymes dual oxidase and endoplasmic reticulum oxidase. In contrast, the genes encoding the antioxidant enzymes superoxide dismutase, catalase, glutathione peroxidase, glutathione S-transferase, thioredoxin, thioredoxin reductase and peroxiredoxin were upregulated. The gene expression pattern in response to infection with Rickettsia rickettsii and exposure to heat-killed microorganisms, Micrococcus luteus, Enterobacter cloacae or S. cerevisiae was the opposite of that triggered by A. marginale challenge. The simultaneous silencing of three genes, catalase, glutathione peroxidase, and thioredoxin as well as the oxidation resistance 1 gene by RNAi apparently favoured the colonization of BME26 cells by A. marginale, suggesting that the antioxidant response might play a role in the control of infection. CONCLUSIONS: Taken together, our results suggest that a general response of tick cells upon microbial stimuli is to increase ROS/RNS production. In contrast, A. marginale infection triggers an opposite profile, suggesting that this pathogen might manipulate the tick redox metabolism to evade the deleterious effect of the oxidant-based innate immune response.


Assuntos
Anaplasma marginale/imunologia , Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/microbiologia , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Rhipicephalus , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Imunidade Inata , Oxirredução , Reação em Cadeia da Polimerase em Tempo Real
13.
Stem Cell Res Ther ; 8(1): 266, 2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29157299

RESUMO

BACKGROUND: This study aimed to investigate the tolerogenic mechanisms induced by semimature dendritic cells (smDCs) and regulatory dendritic cells (regDCs) after transplantation of hepatocytes differentiated from mouse embryonic stem cells (ESCs) and to confirm the low immunogenicity of hepatocytes differentiated from ESCs. METHODS: Green fluorescent protein-labeled ESCs collected from 129 mice were cultured to differentiate into hepatocytes. smDCs and regDCs were cultured in vitro. The hepatocytes were cultured after being extracted from the livers of 129 mice. After injecting smDCs or regDCs 3 days in advance, these differentiated hepatocytes and normal hepatocytes were transplanted into the livers of BALB/c mice separately. Subsequently, the histopathological features and cytokines in transplant tissues as well as the Foxp3 expression in peripheral blood CD4+ T cells of the recipients were examined. RESULTS: The morphological phenotypes of smDCs and regDCs were similar. They both expressed medium levels of MHC-II, CD40, CD80, and CD86, high levels of TGF-ß and IL-10, and low levels of IL-2. The survival of differentiated hepatocytes was prolonged and inflammatory infiltration in transplant tissues was reduced in both the smDC and regDC groups. Foxp3 expression in peripheral blood CD4+ T cells of the smDC group increased to 5.38% and that of the regDC group also rose to 3.87%. Moreover, the inflammatory infiltration in the tissues receiving transplanted hepatocytes was more obvious. CONCLUSIONS: smDCs and regDCs were similar tolerogenic dendritic cells. They both could alleviate the immune injury by inducing CD4+CD25+Foxp3+ regulatory T cells through the medium expression of MHC-II, CD40, CD80, and CD86 and the appropriate secretion of cytokines. Hepatocytes differentiated from ESCs displayed low immunogenicity.


Assuntos
Células Dendríticas/citologia , Células-Tronco Embrionárias/citologia , Hepatócitos/transplante , Tolerância Imunológica , Linfócitos T Reguladores/citologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Biomarcadores/metabolismo , Diferenciação Celular , Células Dendríticas/classificação , Células Dendríticas/imunologia , Células-Tronco Embrionárias/imunologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hepatócitos/citologia , Hepatócitos/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-2/genética , Interleucina-2/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Cultura Primária de Células , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia
14.
Sci Rep ; 7(1): 15840, 2017 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-29158489

RESUMO

Glioblastoma stem-like cells (GSC) are hypothesized to evade current therapies and cause tumor recurrence, contributing to poor patient survival. Existing cell surface markers for GSC are developed from embryonic or neural stem cell systems; however, currently available GSC markers are suboptimal in sensitivity and specificity. We hypothesized that the GSC cell surface proteome could be mined with a yeast display antibody library to reveal novel immunophenotypes. We isolated an extensive collection of antibodies that were differentially selective for GSC. A single domain antibody VH-9.7 showed selectivity for five distinct patient-derived GSC lines and visualized orthotopic GBM xenografts in vivo after conjugation with a near-infrared dye. These findings demonstrate a previously unexplored high-throughput strategy for GSC-selective antibody discovery, to aid in GSC isolation, diagnostic imaging, and therapeutic targeting.


Assuntos
Glioblastoma/imunologia , Células-Tronco Neoplásicas/imunologia , Proteoma/genética , Anticorpos de Domínio Único/imunologia , Animais , Linhagem Celular Tumoral , Células-Tronco Embrionárias/imunologia , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Imunofenotipagem/métodos , Camundongos , Células-Tronco Neoplásicas/patologia , Células-Tronco Neurais/imunologia , Proteoma/imunologia , Saccharomyces cerevisiae/genética , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Stem Cells Dev ; 26(21): 1552-1565, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28891400

RESUMO

The aim of stem cell therapy after cardiac injury is to replace damaged cardiac tissue. Human cardiac progenitor cells (CPCs) represent an interesting cell population for clinical strategies to treat cardiac disease and human CPC-specific antibodies would aid in the clinical implementation of cardiac progenitor-based cell therapy. However, the field of CPC biology suffers from the lack of human CPC-specific markers. Therefore, we raised a panel of monoclonal antibodies (mAb) against CPCs. Of this panel of antibodies, we show that mAb C1096 recognizes a progenitor-like population in the fetal and adult human heart and partially colocalize with reported CPC populations in vitro. Furthermore, mAb C1096 can be used to isolate a multipotent progenitor population from human heart tissue. Interestingly, the two lead candidates, mAb C1096 and mAb C19, recognize glycosylated residues on PECAM1 (platelet and endothelial cell adhesion molecule 1) and GRP78, respectively, and de-N-glycosylation significantly abolishes their binding. Thereby, this report describes new clinically applicable antibodies against human CPCs, and for the first time demonstrates the importance of glycosylated residues as CPCs specific markers.


Assuntos
Células-Tronco Embrionárias/citologia , Proteínas de Choque Térmico/imunologia , Mioblastos Cardíacos/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Anticorpos Monoclonais/imunologia , Antígenos de Superfície/imunologia , Células Cultivadas , Células-Tronco Embrionárias/imunologia , Chaperona BiP do Retículo Endoplasmático , Glicosilação , Humanos , Mioblastos Cardíacos/imunologia , Processamento de Proteína Pós-Traducional
16.
Toxicology ; 387: 1-9, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28645575

RESUMO

Drug-induced liver injury (DILI) is a leading cause of liver disease and a key safety factor during drug development. In addition to the initiation events of drug-specific hepatotoxicity, dysregulated immune responses have been proposed as major pathological events of DILI. Thus, there is a need for a reliable cell culture model with which to assess drug-induced immune reactions to predict hepatotoxicity for drug development. To this end, stem cell-derived hepatocytes have shown great potentials. Here we report that hepatocyte-like cells derived from human embryonic stem cells (hES-HLCs) can be used to evaluate drug-induced hepatotoxic immunological events. Treatment with acetaminophen significantly elevated the levels of inflammatory cytokines by hES-HLCs. Moreover, three human immune cell lines, Jurkat, THP-1, and NK92MI, were activated when cultured in conditioned medium obtained from acetaminophen-treated hES-HLCs. To further validate, we tested thiazolidinedione (TZD) class, antidiabetic drugs, including troglitazone withdrawn from the market because of severe idiosyncratic drug hepatotoxicity. We found that TZD drug treatment to hES-HLCs resulted in the production of pro-inflammatory cytokines and eventually associated immune cell activation. In summary, our study demonstrates for the first time the potential of hES-HLCs as an in vitro model system for assessment of drug-induced as well as immune-mediated hepatotoxicity.


Assuntos
Acetaminofen/toxicidade , Analgésicos não Narcóticos/toxicidade , Bioensaio , Diferenciação Celular , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Células-Tronco Embrionárias/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hipoglicemiantes/toxicidade , Tiazolidinedionas/toxicidade , Testes de Toxicidade/métodos , Sobrevivência Celular/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocinas/imunologia , Citocinas/metabolismo , Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/patologia , Hepatócitos/imunologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Células Jurkat , Fenótipo , Medição de Risco
17.
J Immunol ; 198(1): 239-248, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27852743

RESUMO

Application of dendritic cells (DCs) to prime responses to tumor Ags provides a promising approach to immunotherapy. However, only a limited number of DCs can be manufactured from adult precursors. In contrast, pluripotent embryonic stem (ES) cells represent an inexhaustible source for DC production, although it remains a major challenge to steer directional differentiation because ES cell-derived cells are typically immature with impaired functional capacity. Consistent with this notion, we found that mouse ES cell-derived DCs (ES-DCs) represented less mature cells compared with bone marrow-derived DCs. This finding prompted us to compare the gene expression profile of the ES cell- and adult progenitor-derived, GM-CSF-instructed, nonconventional DC subsets. We quantified the mRNA level of 17 DC-specific transcription factors and observed that 3 transcriptional regulators (Irf4, Spi-B, and Runx3) showed lower expression in ES-DCs than in bone marrow-derived DCs. In light of this altered gene expression, we probed the effects of these transcription factors in developing mouse ES-DCs with an isogenic expression screen. Our analysis revealed that forced expression of Irf4 repressed ES-DC development, whereas, in contrast, Runx3 improved the ES-DC maturation capacity. Moreover, LPS-treated and Runx3-activated ES-DCs exhibited enhanced T cell activation and migratory potential. In summary, we found that ex vivo-generated ES-DCs had a compromised maturation ability and immunogenicity. However, ectopic expression of Runx3 enhances cytokine-driven ES-DC development and acts as an instructive tool for the generation of mature DCs with enhanced immunogenicity from pluripotent stem cells.


Assuntos
Diferenciação Celular/fisiologia , Subunidade alfa 3 de Fator de Ligação ao Core/biossíntese , Células Dendríticas/citologia , Expressão Ectópica do Gene/fisiologia , Células-Tronco Embrionárias/citologia , Animais , Western Blotting , Separação Celular , Células Cultivadas , Subunidade alfa 3 de Fator de Ligação ao Core/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/imunologia , Células-Tronco Pluripotentes/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transcriptoma
18.
Stem Cell Res ; 17(3): 607-615, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27838585

RESUMO

Wnt signaling plays a central role in tumor initiation and tumor progression. Mutations in Wnt pathway components, such as the tumor suppressor APC, lead to malignant transformation. While previous studies focused on Wnt-related changes in cancer cells, the impact of aberrant Wnt signaling on the tumor microenvironment is only beginning to emerge. In order to investigate the role of increased Wnt secretion on tumor growth and the microenvironment, we generated a novel germ cell tumor model by overexpressing the Wnt secretion factor Evi/Wls in mouse embryonic stem cells. Evi-overexpressing teratoma were characterized by enhanced tumor growth in supporting a tumor-promoting role of Wnt secretion. Interestingly, enhanced Evi expression correlated with impaired immune cell recruitment. Specifically, T- and B-cell infiltration was reduced in Evi-overexpressing teratomas, which was independent of teratoma size and differentiation. Our study suggests that Wnt secretion impairs immunosurveillance. Since immune cell infiltration has been shown to have prognostic value, the levels of secreted Wnt activity might impact the efficiency of cancer immunotherapy.


Assuntos
Teratoma/imunologia , Teratoma/patologia , Proteínas Wnt/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Humanos , Camundongos , Camundongos SCID , Teratoma/metabolismo , Proteínas Wnt/imunologia
19.
PLoS One ; 11(3): e0150731, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26977925

RESUMO

Transplantation of ex vivo expanded corneal limbal stem cells (LSCs) has been the main treatment for limbal stem cell deficiency, although the shortage of donor corneal tissues remains a major concern for its wide application. Due to the development of tissue engineering, embryonic stem cells (ESCs)-derived corneal epithelial-like cells (ESC-CECs) become a new direction for this issue. However, the immunogenicity of ESC-CECs is a critical matter to be solved. In the present study, we explored the immunological properties of ESC-CECs, which were differentiated from ESCs. The results showed that ESC-CECs had a similar character and function with LSCs both in vitro and in vivo. In ESC-CECs, a large number of genes related with immune response were down-regulated. The expressions of MHC-I, MHC-II, and co-stimulatory molecules were low, but the expression of HLA-G was high. The ESC-CECs were less responsible for T cell proliferation and NK cell lysis in vitro, and there was less immune cell infiltration after transplantation in vivo compared with LSCs. Moreover, the immunological properties were not affected by interferon-γ. All these results indicated a low immunogenicity of ESC-CECs, and they can be promising in clinical use.


Assuntos
Células-Tronco Embrionárias/imunologia , Animais , Proliferação de Células , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Expressão Gênica , Humanos , Masculino , Coelhos , Transplante de Células-Tronco , Linfócitos T/citologia
20.
Stem Cell Reports ; 6(1): 137-49, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26724903

RESUMO

B cells are key components of cellular and humoral immunity and, like all lymphocytes, are thought to originate and renew from hematopoietic stem cells (HSCs). However, our recent single-HSC transfer studies demonstrate that adult bone marrow HSCs do not regenerate B-1a, a subset of tissue B cells required for protection against pneumonia, influenza, and other infections. Since B-1a are regenerated by transfers of fetal liver, the question arises as to whether B-1a derive from fetal, but not adult, HSCs. Here we show that, similar to adult HSCs, fetal HSCs selectively fail to regenerate B-1a. We also show that, in humanized mice, human fetal liver regenerates tissue B cells that are phenotypically similar to murine B-1a, raising the question of whether human HSC transplantation, the mainstay of such models, is sufficient to regenerate human B-1a. Thus, our studies overtly challenge the current paradigm that HSCs give rise to all components of the immune system.


Assuntos
Linfócitos B/imunologia , Diferenciação Celular/imunologia , Células-Tronco Fetais/transplante , Transplante de Tecido Fetal/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Animais , Animais Recém-Nascidos , Linfócitos B/citologia , Linfócitos B/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Fetais/imunologia , Células-Tronco Fetais/metabolismo , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fígado/citologia , Fígado/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...