Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
Viruses ; 16(4)2024 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-38675895

RESUMO

Macrophages play multiple roles in innate immunity including phagocytosing pathogens, modulating the inflammatory response, presenting antigens, and recruiting other immune cells. Tissue-resident macrophages (TRMs) adapt to the local microenvironment and can exhibit different immune responses upon encountering distinct pathogens. In this study, we generated induced macrophages (iMACs) derived from human pluripotent stem cells (hPSCs) to investigate the interactions between the macrophages and various human pathogens, including the hepatitis C virus (HCV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and Streptococcus pneumoniae. iMACs can engulf all three pathogens. A comparison of the RNA-seq data of the iMACs encountering these pathogens revealed that the pathogens activated distinct gene networks related to viral response and inflammation in iMACs. Interestingly, in the presence of both HCV and host cells, iMACs upregulated different sets of genes involved in immune cell migration and chemotaxis. Finally, we constructed an image-based high-content analysis system consisting of iMACs, recombinant GFP-HCV, and hepatic cells to evaluate the effect of a chemical inhibitor on HCV infection. In summary, we developed a human cell-based in vitro model to study the macrophage response to human viral and bacterial infections; the results of the transcriptome analysis indicated that the iMACs were a useful resource for modeling pathogen-macrophage-tissue microenvironment interactions.


Assuntos
Hepacivirus , Macrófagos , Células-Tronco Pluripotentes , SARS-CoV-2 , Humanos , Macrófagos/imunologia , Macrófagos/virologia , Hepacivirus/imunologia , Hepacivirus/fisiologia , SARS-CoV-2/imunologia , Células-Tronco Pluripotentes/imunologia , Streptococcus pneumoniae/imunologia , COVID-19/imunologia , COVID-19/virologia , Hepatite C/imunologia , Hepatite C/virologia , Fagocitose , Viroses/imunologia , Imunidade Inata
2.
Prog Mol Biol Transl Sci ; 199: 271-296, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37678974

RESUMO

It is urgent to prepare and store large numbers of clinical trial grade human pluripotent stem (hPS) cells for off-the-shelf use in stem cell therapies. However, stem cell banks, which store off-the-shelf stem cells, need financial support and large amounts of technicians for daily cell maintenance. Therefore, it is valuable to create "universal" or "hypoimmunogenic" hPS cells with genome editing engineering by knocking in or out immune-related genes. Only a small number of universal or hypoimmunogenic hPS cell lines should be needed to store for off-the-shelf usage and reduce the large amounts of instruments, consumables and technicians. In this article, we consider how to create hypoimmunogenic or universal hPS cells as well as the demerits of the technology. ß2-Microglobulin-knockout hPS cells did not harbor human leukocyte antigen (HLA)-expressing class I cells but led to the activation of natural killer cells. To escape the activities of macrophages and natural killer cells, homozygous hPS cells having a single allele of an HLA class I gene, such as HLA-C, were proposed. Major HLA class Ia molecules were knocked out, and CD47, HLA-G and PD-L1 were knocked in hPS cells utilizing CRISPR/Cas9 genome editing. Finally, some researchers are trying to generate universal hPS cells without genome editing. The cells evaded the activation of not only T cells but also macrophages and natural killer cells. These universal hPS cells have high potential for application in cell therapy.


Assuntos
Células-Tronco Pluripotentes , Transplante de Células-Tronco , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/imunologia , Células-Tronco Pluripotentes/metabolismo , Antígenos HLA , Humanos , Técnicas de Silenciamento de Genes , Técnicas de Inativação de Genes , Edição de Genes , Técnicas de Introdução de Genes , Animais , Imunologia de Transplantes , Bancos de Espécimes Biológicos
3.
Viruses ; 13(10)2021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34696391

RESUMO

Respiratory Syncytial Virus (RSV) is the leading cause of acute lower respiratory infections in young children and infection has been linked to the development of persistent lung disease in the form of wheezing and asthma. Despite substantial research efforts, there are no RSV vaccines currently available and an effective monoclonal antibody targeting the RSV fusion protein (palivizumab) is of limited general use given the associated expense. Therefore, the development of novel approaches to prevent RSV infection is highly desirable to improve pediatric health globally. We have developed a method to generate alveolar-like macrophages (ALMs) from pluripotent stem cells. These ALMs have shown potential to promote airway innate immunity and tissue repair and so we hypothesized that ALMs could be used as a strategy to prevent RSV infection. Here, we demonstrate that ALMs are not productively infected by RSV and prevent the infection of epithelial cells. Prevention of epithelial infection was mediated by two different mechanisms: phagocytosis of RSV particles and release of an antiviral soluble factor different from type I interferon. Furthermore, intratracheal administration of ALMs protected mice from subsequent virus-induced weight loss and decreased lung viral titres and inflammation, indicating that ALMs can impair the pathogenesis of RSV infection. Our results support a prophylactic role for ALMs in the setting of RSV infection and warrant further studies on stem cell-derived ALMs as a novel cell-based therapy for pulmonary viral infections.


Assuntos
Imunidade Inata , Macrófagos/imunologia , Macrófagos/virologia , Células-Tronco Pluripotentes/fisiologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Animais , Anticorpos Antivirais/sangue , Linhagem Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Epiteliais/virologia , Sangue Fetal/citologia , Humanos , Inflamação/virologia , Macrófagos/classificação , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco Pluripotentes/imunologia , Infecções por Vírus Respiratório Sincicial/terapia
4.
Nat Commun ; 12(1): 5023, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34408144

RESUMO

T cells are pivotal effectors of the immune system and can be harnessed as therapeutics for regenerative medicine and cancer immunotherapy. An unmet challenge in the field is the development of a clinically relevant system that is readily scalable to generate large numbers of T-lineage cells from hematopoietic stem/progenitor cells (HSPCs). Here, we report a stromal cell-free, microbead-based approach that supports the efficient in vitro development of both human progenitor T (proT) cells and T-lineage cells from CD34+cells sourced from cord blood, GCSF-mobilized peripheral blood, and pluripotent stem cells (PSCs). DL4-µbeads, along with lymphopoietic cytokines, induce an ordered sequence of differentiation from CD34+ cells to CD34+CD7+CD5+ proT cells to CD3+αß T cells. Single-cell RNA sequencing of human PSC-derived proT cells reveals a transcriptional profile similar to the earliest thymocytes found in the embryonic and fetal thymus. Furthermore, the adoptive transfer of CD34+CD7+ proT cells into immunodeficient mice demonstrates efficient thymic engraftment and functional maturation of peripheral T cells. DL4-µbeads provide a simple and robust platform to both study human T cell development and facilitate the development of engineered T cell therapies from renewable sources.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas de Ligação ao Cálcio/imunologia , Células-Tronco Hematopoéticas/citologia , Linfopoese , Doenças da Imunodeficiência Primária/terapia , Linfócitos T/citologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Antígenos CD34/genética , Antígenos CD34/imunologia , Proteínas de Ligação ao Cálcio/genética , Linhagem da Célula , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/imunologia , Doenças da Imunodeficiência Primária/genética , Doenças da Imunodeficiência Primária/imunologia , Doenças da Imunodeficiência Primária/fisiopatologia , Linfócitos T/imunologia , Linfócitos T/transplante
5.
Stem Cell Rev Rep ; 17(6): 2107-2119, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34365591

RESUMO

The virus responsible for coronavirus disease 2019 (COVID-19), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has infected over 190 million people to date, causing a global pandemic. SARS-CoV-2 relies on binding of its spike glycoprotein to angiotensin-converting enzyme 2 (ACE2) for infection. In addition to fever, cough, and shortness of breath, severe cases of SARS-CoV-2 infection may result in the rapid overproduction of pro-inflammatory cytokines. This overactive immune response is known as a cytokine storm, which leads to several serious clinical manifestations such as acute respiratory distress syndrome and myocardial injury. Cardiovascular disorders such as acute coronary syndrome (ACS) and heart failure not only enhance disease progression at the onset of infection, but also arise in hospitalized patients with COVID-19. Tissue-specific differentiated cells and organoids derived from human pluripotent stem cells (hPSCs) serve as an excellent model to address how SARS-CoV-2 damages the lungs and the heart. In this review, we summarize the molecular basis of SARS-CoV-2 infection and the current clinical perspectives of the bidirectional relationship between the cardiovascular system and viral progression. Furthermore, we also address the utility of hPSCs as a dynamic model for SARS-CoV-2 research and clinical translation.


Assuntos
COVID-19/virologia , Sistema Cardiovascular/virologia , Células-Tronco Pluripotentes/virologia , COVID-19/imunologia , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/virologia , Sistema Cardiovascular/imunologia , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/virologia , Humanos , Pulmão/imunologia , Pulmão/virologia , Pandemias/prevenção & controle , Células-Tronco Pluripotentes/imunologia , SARS-CoV-2/patogenicidade
6.
STAR Protoc ; 2(2): 100563, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34151293

RESUMO

We describe the extended endothelial cell culture method (EECM) for the differentiation of human pluripotent stem cells (hPSCs) into brain microvascular endothelial cell (BMEC)-like cells. EECM-BMEC-like cells resemble primary human BMECs in morphology, molecular junctional architecture, and diffusion barrier characteristics. A mature immune phenotype with proper endothelial adhesion molecule expression makes this model distinct from any other hPSC-derived in vitro blood-brain barrier (BBB) model and suitable to study immune cell migration across the BBB in a disease relevant and personalized fashion. For complete details on the use and execution of this protocol, please refer to Lian et al. (2014), Nishihara et al. (2020a).


Assuntos
Encéfalo/irrigação sanguínea , Diferenciação Celular , Endotélio Vascular/citologia , Microvasos/citologia , Células-Tronco Pluripotentes/citologia , Células Cultivadas , Endotélio Vascular/imunologia , Humanos , Microvasos/imunologia , Células-Tronco Pluripotentes/imunologia
8.
Nat Rev Immunol ; 21(7): 441-453, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33398129

RESUMO

Advancements in human pluripotent stem cell technology offer a unique opportunity for the neuroimmunology field to study host-virus interactions directly in disease-relevant cells of the human central nervous system (CNS). Viral encephalitis is most commonly caused by herpesviruses, arboviruses and enteroviruses targeting distinct CNS cell types and often leading to severe neurological damage with poor clinical outcomes. Furthermore, different neurotropic viruses will affect the CNS at distinct developmental stages, from early prenatal brain development to the aged brain. With the unique flexibility and scalability of human pluripotent stem cell technology, it is now possible to examine the molecular mechanisms underlying acute infection and latency, determine which CNS subpopulations are specifically infected, study temporal aspects of viral susceptibility, perform high-throughput chemical or genetic screens for viral restriction factors and explore complex cell-non-autonomous disease mechanisms. Therefore, human pluripotent stem cell technology has the potential to address key unanswered questions about antiviral immunity in the CNS, including emerging questions on the potential CNS tropism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).


Assuntos
Sistema Nervoso Central/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Células-Tronco Pluripotentes/imunologia , Tropismo Viral , COVID-19 , Humanos , Microglia , Neuroglia , Neurônios , SARS-CoV-2
9.
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
10.
Curr Protoc Stem Cell Biol ; 54(1): e113, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32588980

RESUMO

New human pluripotent stem cell (hPSC)-derived therapies are advancing to clinical trials at an increasingly rapid pace. In addition to ensuring that the therapies function properly, there is a critical need to investigate the human immune response to these cell products. A robust allogeneic (or autologous) immune response could swiftly eliminate an otherwise promising cell therapy, even in immunosuppressed patients. In coming years, researchers in the regenerative medicine field will need to utilize a number of in vitro and in vivo assays and models to evaluate and better understand hPSC immunogenicity. Humanized mouse models-mice engrafted with functional human immune cell types-are an important research tool for investigating the mechanisms of the adaptive immune response to hPSC therapies. This article provides an overview of humanized mouse models relevant to the study of hPSC immunogenicity and explores central considerations for investigators seeking to utilize these powerful models in their research. © 2020 Wiley Periodicals LLC.


Assuntos
Células-Tronco Pluripotentes/imunologia , Animais , Quimerismo , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Modelos Animais , Transplante de Células-Tronco
11.
Front Immunol ; 10: 2580, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31787975

RESUMO

The safe utilization of induced pluripotent stem cell (iPSC) derivatives in clinical use is attributed to the complete elimination of the risk of forming teratomas after transplantation. The extent by which such a risk exists in immune-competent hosts is mostly unknown. Here, using humanized mice reconstituted with fetal hematopoietic stem cells and autologous thymus tissue (bone-liver-thymus humanized mice [Hu-BLT]) or following the adoptive transfer of peripheral blood mononuclear cells(PBMCs) (Hu-AT), we evaluated the capacity of immune cells to prevent or eliminate teratomas derived from human iPSCs (hiPSCs). Our results showed that the injection of hiPSCs failed to form teratomas in Hu-AT mice reconstituted with allogeneic or autologous PBMCs or purified natural killer (NK) cells alone. However, teratomas were observed in Hu-AT mice reconstituted with autologous PBMCs depleted from NK cells. In line with these results, Hu-BLT, which do not have functional NK cells, could not prevent the growth of teratomas. Finally, we found that established teratomas were not targeted by NK cells and instead were efficiently rejected by allogeneic but not autologous T cells in Hu-AT mice. Overall, our findings suggest that autologous hiPSC-derived therapies are unlikely to form teratomas in the presence of NK cells.


Assuntos
Células Matadoras Naturais/imunologia , Células-Tronco Pluripotentes/imunologia , Teratoma/prevenção & controle , Transferência Adotiva/efeitos adversos , Adulto , Animais , Humanos , Leucócitos Mononucleares/transplante , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Linfócitos T/imunologia , Teratoma/etiologia , Teratoma/imunologia , Transplante Heterólogo
12.
Cell Tissue Bank ; 20(4): 467-488, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31606767

RESUMO

Breastmilk is a dynamic, multi-faceted, and complex fluid containing a plethora of biochemical and cellular components that execute developmental effects or differentiation program, providing nourishment and immunity to newborns. Recently, it was reported that breastmilk contains a heterogeneous population of naïve cells, including pluripotent stem cells, multipotent stem cells, immune cells, and non-immune cells. The stem cells derived from breastmilk possess immune privilege and non-tumorigenic properties. Thus, breastmilk may represent an ideal source of stem cells collected by non-perceive procedure than other available sources. Thus, this "maternally originating natural regenerative medicine" may have innumerable applications in clinical biology, cosmetics, and pharmacokinetics. This review describes the efficient integrated cellular system of mammary glands, the impressive stem cell hierarchy of breastmilk, and their possible implications in translational research and therapeutics.


Assuntos
Leite Humano/citologia , Células-Tronco Multipotentes/citologia , Células-Tronco Pluripotentes/citologia , Pesquisa com Células-Tronco , Diferenciação Celular , Humanos , Imunidade Celular , Recém-Nascido , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/imunologia , Leite Humano/imunologia , Células-Tronco Multipotentes/imunologia , Células-Tronco Pluripotentes/imunologia , Medicina Regenerativa/métodos
13.
Trends Mol Med ; 25(11): 935-937, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31624020

RESUMO

Immune checkpoint inhibitors (ICIs) have revolutionised cancer immunotherapy but their success is wholly dependent on amplifying an existing immune response directed against the tumour. A recent study by Tsuchiya et al. suggests how the properties of induced pluripotent stem cells (iPSCs) may be exploited for the targeted delivery of interferon-α (IFNα) to elicit an appropriate response.


Assuntos
Imunoterapia , Interferon-alfa , Terapia de Alvo Molecular , Neoplasias/imunologia , Humanos , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , Neoplasias/terapia , Células-Tronco Pluripotentes/imunologia , Células-Tronco Pluripotentes/metabolismo
14.
Am J Physiol Heart Circ Physiol ; 317(5): H954-H957, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31559827

RESUMO

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) can be exploited as models for a wide range of research applications and numerous protocols for generating hPSC-CMs have been described. However, it is currently not possible to direct differentiation to a single, homogeneous end point, and the resulting heterogeneity may be variable among laboratories, cell lines, and protocols. Consequently, the ability to assess phenotypic heterogeneity of the cell population is critical to the interpretation, repeatability, and reproduction of hPSC-CM studies. While flow cytometry is well suited for this purpose, a review of published literature reveals there is currently no consensus regarding which marker, antibody, or protocol is best suited to enable comparisons of hPSC-CM culture heterogeneity. Moreover, the lack of available experimental detail, combined with the variability in the approaches used for hPSC-CM evaluation, makes it challenging to reproduce, interpret, and compare published data. Consequently, this article calls for an alignment of the way researchers approach the routine use and documentation of the antibodies and controls used during flow cytometry-based assessment of hPSC-CM cultures. We advocate for the adoption of a "fit for purpose" validation mindset, whereby antibodies and experimental conditions are demonstrated as specific within a defined experimental design and biological context. Overall, we expect that by adhering to rigorous standards for antibody validation and use, reporting of experimental details, and presentation of data, the concepts emphasized here will promote enhanced utility and dialogue regarding hPSC-CM for a variety of research and translational applications by enabling more accurate comparisons of results among studies.


Assuntos
Anticorpos/imunologia , Diferenciação Celular , Linhagem da Célula , Citometria de Fluxo/normas , Miócitos Cardíacos/imunologia , Células-Tronco Pluripotentes/imunologia , Especificidade de Anticorpos , Biomarcadores/metabolismo , Células Cultivadas , Consenso , Guias como Assunto/normas , Humanos , Miócitos Cardíacos/metabolismo , Fenótipo , Células-Tronco Pluripotentes/metabolismo , Reprodutibilidade dos Testes
15.
Proc Natl Acad Sci U S A ; 116(21): 10441-10446, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31040209

RESUMO

Polymorphic HLAs form the primary immune barrier to cell therapy. In addition, innate immune surveillance impacts cell engraftment, yet a strategy to control both, adaptive and innate immunity, is lacking. Here we employed multiplex genome editing to specifically ablate the expression of the highly polymorphic HLA-A/-B/-C and HLA class II in human pluripotent stem cells. Furthermore, to prevent innate immune rejection and further suppress adaptive immune responses, we expressed the immunomodulatory factors PD-L1, HLA-G, and the macrophage "don't-eat me" signal CD47 from the AAVS1 safe harbor locus. Utilizing in vitro and in vivo immunoassays, we found that T cell responses were blunted. Moreover, NK cell killing and macrophage engulfment of our engineered cells were minimal. Our results describe an approach that effectively targets adaptive as well as innate immune responses and may therefore enable cell therapy on a broader scale.


Assuntos
Engenharia Genética/métodos , Células-Tronco Pluripotentes/imunologia , Sistemas CRISPR-Cas , Linhagem Celular , Técnicas de Inativação de Genes , Genes MHC Classe I , Genes MHC da Classe II , Humanos
16.
Cell Stem Cell ; 24(3): 376-389.e8, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30661959

RESUMO

The ability to generate T cells from pluripotent stem cells (PSCs) has the potential to transform autologous T cell immunotherapy by facilitating universal, off-the-shelf cell products. However, differentiation of human PSCs into mature, conventional T cells has been challenging with existing methods. We report that a continuous 3D organoid system induced an orderly sequence of commitment and differentiation from PSC-derived embryonic mesoderm through hematopoietic specification and efficient terminal differentiation to naive CD3+CD8αß+ and CD3+CD4+ conventional T cells with a diverse T cell receptor (TCR) repertoire. Introduction of an MHC class I-restricted TCR in PSCs produced naive, antigen-specific CD8αß+ T cells that lacked endogenous TCR expression and showed anti-tumor efficacy in vitro and in vivo. Functional assays and RNA sequencing aligned PSC-derived T cells with primary naive CD8+ T cells. The PSC-artificial thymic organoid (ATO) system presented here is an efficient platform for generating functional, mature T cells from human PSCs.


Assuntos
Diferenciação Celular , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Linfócitos T/citologia , Animais , Células Cultivadas , Humanos , Células K562 , Camundongos , Camundongos Endogâmicos NOD , Organoides/imunologia , Células-Tronco Pluripotentes/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia
17.
Cancer Sci ; 110(1): 16-22, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30485606

RESUMO

Recent outstanding clinical results produced by engineered T cells, including chimeric antigen receptors, have already facilitated further research that broadens their applicability. One such direction is to explore new T cell sources for allogeneic "off-the-shelf" adoptive immunotherapy. Human pluripotent stem cells could serve as an alternative cell source for this purpose due to their unique features of infinite propagation ability and pluripotency. Here, we describe the current state of engineered T cell transfer with the focus on cell manufacturing processes and the potentials and challenges of induced pluripotent stem cell-derived T cells as a starting material to construct off-the-shelf T-cell banks.


Assuntos
Engenharia Celular/métodos , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Linfócitos T/transplante , Diferenciação Celular/imunologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/imunologia , Neoplasias/imunologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transplante Homólogo
18.
Stem Cells Dev ; 28(4): 268-277, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30572796

RESUMO

In diabetes, multipotent stromal cells (MSCs) are functionally deficient. It is unknown, however, whether their antibacterial function is compromised. In this study, MSCs were isolated from the bone marrow samples provided by nine diabetic and six nondiabetic donors and treated with or without Escherichia coli lipopolysaccharides (LPS). The supernatant of diabetic MSCs (MSCs-dia) and nondiabetic control MSCs (MSCs-c) was added into the cultures of E. coli for evaluation of the effect of MSCs-dia and MSCs-c on bacterial growth. The number of E. coli colonies increased when they were cultured with the supernatant of MSCs-dia, with or without LPS stimulation, compared with the E. coli cultured with the supernatant of MSCs-c. Human macrophages were co-cultured with either MSCs-dia or MSCs-c, for 24 h, and then cultured with heat-inactivated E. coli. Bacterial phagocytosis was reduced after macrophages were co-cultured with MSCs-dia. Gene expression of antibacterial peptide LL-37 and indoleamine 2,3-dioxygenase (IDO) by MSCs-dia was reduced compared with MSCs-c. The supernatant of MSCs-dia and MSCs-c was applied to a 42-cytokine antibody array. While the cytokine profiles of MSCs-dia and MSCs-c were largely similar, the productions of MCP-1 and interleukin-6 distinguished MSCs-dia from MSCs-c in response to LPS treatment. In conclusion, MSCs-dia were less inhibitive of the growth of bacteria and compromised in regulation of macrophages for bacterial phagocytosis. The reduced expression of IDO and LL-37 and an altered cytokine profile in MSCs-dia should be taken into consideration in developing cell therapies for diabetic infection.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Citocinas/metabolismo , Diabetes Mellitus/imunologia , Fagocitose , Células-Tronco Pluripotentes/imunologia , Idoso , Peptídeos Catiônicos Antimicrobianos/genética , Células da Medula Óssea/imunologia , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Citocinas/genética , Escherichia coli/efeitos dos fármacos , Feminino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Macrófagos/imunologia , Masculino , Pessoa de Meia-Idade , Catelicidinas
19.
Differentiation ; 104: 42-49, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30453197

RESUMO

Macrophages are phagocytic immune cells resident in every tissue that are not only important for host defence, but are also involved in tissue homeostasis, injury, and disease. Despite increasingly sophisticated methods for in vitro macrophage isolation, expansion and activation over the past three decades, these have largely been restricted to modelling bone-marrow or blood-derived cells. The in vitro derivation of macrophages from human pluripotent stem cells provides new opportunities to study macrophage biology, including the factors that impact human myeloid development and those that induce macrophage activation. While sharing many of the functional characteristics of monocyte-derived macrophages, stem cell-derived macrophages may offer new opportunities to understand the role of development or tissue context in innate immune cell function. Immune responsiveness to pathogenic challenge is known to be impacted by a macrophage's history of prior exposure, as well as ontogeny and tissue context. Therefore, we explore the factors of in vitro derivation likely to influence macrophage phenotype and function.


Assuntos
Diferenciação Celular/genética , Células-Tronco Pluripotentes Induzidas/citologia , Macrófagos/citologia , Células-Tronco Pluripotentes/citologia , Humanos , Imunidade Inata/genética , Células-Tronco Pluripotentes Induzidas/imunologia , Macrófagos/imunologia , Monócitos/citologia , Monócitos/imunologia , Células-Tronco Pluripotentes/imunologia
20.
Sci Rep ; 8(1): 16550, 2018 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-30410112

RESUMO

The in vitro induction of corneal epithelial cells (CECs) from human induced pluripotent stem cells (iPSCs) represents a new strategy for obtaining CE stem/progenitor cells for the surgical reconstruction of a diseased or injured ocular surface. The clinical promise of this strategy is considerable, but if the approaches' potential is to be realised, robust methods for the purification of iPSC-derived CE lineage cells need to be developed to avoid contamination with other cells that may carry the risk of unwanted side effects, such as tumorigenesis. Experiments conducted here revealed that during CEC isolation, CD200-negative selection using a cell sorter considerably reduced the contamination of the cell population with various non-CECs compared with what could be achieved using TRA-1-60, a conventional negative marker for CECs. Furthermore, CD200-negative sorting did not affect the yield of CECs nor that of their stem/progenitor cells. Single-cell gene expression analysis for CEC sheets obtained using CD200-negative sorting showed that all analysed cells were CE-lineage cells, expressing PAX6, delta-N p63, and E-cadherin. Non-CECs, on the other hand, expressed non-CEC genes such as FGFR1 and RPE65. CD200, thus, represents a robust negative marker for purification of induced CE lineage cells, which is expressed by undifferentiated iPSCs and non-CECs, including iPSC-derived neural and retinal cells.


Assuntos
Antígenos CD/metabolismo , Epitélio Corneano/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Antígenos CD/genética , Antígenos de Superfície/genética , Caderinas/genética , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Epitélio Corneano/imunologia , Humanos , Células-Tronco Pluripotentes Induzidas/imunologia , Fator de Transcrição PAX6/genética , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/imunologia , Proteoglicanas/genética , Análise de Célula Única
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...