Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.550
Filtrar
1.
Front Immunol ; 15: 1447536, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39224602

RESUMO

Mesenchymal stem cell derived extracellular vesicles (MSC EVs) are paracrine modulators of macrophage function. Scientific research has primarily focused on the immunomodulatory and regenerative properties MSC EVs derived from bone marrow. The dental pulp is also a source for MSCs, and their anatomical location and evolutionary function has primed them to be potent immunomodulators. In this study, we demonstrate that extracellular vesicles derived from dental pulp stem cells (DPSC EVs) have pronounced immunomodulatory effect on primary macrophages by regulating the NFκb pathway. Notably, the anti-inflammatory activity of DPSC-EVs is enhanced following exposure to an inflammatory stimulus (LPS). These inhibitory effects were also observed in vivo. Sequencing of the naïve and LPS preconditioned DPSC-EVs and comparison with our published results from marrow MSC EVs revealed that Naïve and LPS preconditioned DPSC-EVs are enriched with anti-inflammatory miRNAs, particularly miR-320a-3p, which appears to be unique to DPSC-EVs and regulates the NFκb pathway. Overall, our findings highlight the immunomodulatory properties of DPSC-EVs and provide vital clues that can stimulate future research into miRNA-based EV engineering as well as therapeutic approaches to inflammation control and disease treatment.


Assuntos
Polpa Dentária , Vesículas Extracelulares , Imunomodulação , Inflamação , NF-kappa B , Polpa Dentária/citologia , Polpa Dentária/imunologia , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/imunologia , Humanos , Animais , Inflamação/imunologia , Inflamação/metabolismo , NF-kappa B/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , MicroRNAs/genética , Lipopolissacarídeos/farmacologia , Camundongos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/imunologia , Células Cultivadas , Transdução de Sinais , Células-Tronco/imunologia , Células-Tronco/metabolismo , Masculino
2.
Sci Adv ; 10(34): eado2048, 2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39178248

RESUMO

Adoptive T cell therapy (ACT), the therapeutic transfer of defined T cell immunity to patients, offers great potential in the fight against different human diseases including difficult-to-treat viral infections, but persistence and longevity of the cells are areas of concern. Very-early-differentiated stem cell memory T cells (TSCMs) have superior self-renewal, engraftment, persistence, and anticancer efficacy, but their potential for antiviral ACT remains unknown. Here, we developed a clinically scalable protocol for expanding Epstein-Barr virus (EBV)-specific TSCM-enriched T cells with high proportions of CD4+ T cells and broad EBV antigen coverage. These cells showed tumor control in a xenograft model of EBV-induced lymphoma and were superior to previous ACT protocols in terms of tumor infiltration, in vivo proliferation, persistence, proportion of functional CD4+ T cells, and diversity of EBV antigen specificity. Thus, our protocol may pave the way for the next generation of potent unmodified antigen-specific cell therapies for EBV-associated diseases, including tumors, and other indications.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Células T de Memória , Herpesvirus Humano 4/imunologia , Animais , Humanos , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/terapia , Camundongos , Células T de Memória/imunologia , Linfócitos T CD4-Positivos/imunologia , Imunoterapia Adotiva/métodos , Ensaios Antitumorais Modelo de Xenoenxerto , Células-Tronco/imunologia , Células-Tronco/virologia , Memória Imunológica , Linfoma/terapia , Linfoma/imunologia , Linfoma/patologia , Modelos Animais de Doenças
3.
J Clin Invest ; 134(18)2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39052353

RESUMO

Epithelial barriers are programmed for defense and repair but are also the site of long-term structural remodeling and disease. In general, this paradigm features epithelial stem cells (ESCs) that are called on to regenerate damaged tissues but can also be reprogrammed for detrimental remodeling. Here we identified a Wfdc21-dependent monocyte-derived dendritic cell (moDC) population that functioned as an early sentinel niche for basal ESC reprogramming in mouse models of epithelial injury after respiratory viral infection. Niche function depended on moDC delivery of ligand GPNMB to the basal ESC receptor CD44 so that properly timed antibody blockade of ligand or receptor provided long-lasting correction of reprogramming and broad disease phenotypes. These same control points worked directly in mouse and human basal ESC organoids. Together, the findings identify a mechanism to explain and modify what is otherwise a stereotyped but sometimes detrimental response to epithelial injury.


Assuntos
Reprogramação Celular , Animais , Camundongos , Humanos , Reprogramação Celular/imunologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Dendríticas/imunologia , Receptores de Hialuronatos/metabolismo , Receptores de Hialuronatos/imunologia , Receptores de Hialuronatos/genética , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Nicho de Células-Tronco/imunologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Pneumopatias/imunologia , Pneumopatias/patologia , Pneumopatias/metabolismo
4.
Cancer Gene Ther ; 31(7): 995-1006, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38858535

RESUMO

Herein, we present human adipose-derived stem cells (ADSCs) inserted with the receptor-interacting protein kinase-3 (RIP3) gene (RP@ADSCs), which induces cell necroptosis, for tumor immunotherapy. Necroptosis has characteristics of both apoptosis, such as programmed cell death, and necrosis, such as swelling and plasma membrane rupture, during which damage-related molecular patterns are released, triggering an immune response. Therefore, necroptosis has the potential to be used as an effective anticancer immunotherapy. RP@ADSCs were programmed to necroptosis after a particular time after being injected in vivo, and various pro-inflammatory cytokines secreted during the stem cell death process stimulated the immune system, showing local and sustained anticancer effects. It was confirmed that RIP3 protein expression increased in ADSCs after RP transfection. RP@ADSCs continued to induce ADSCs death for 7 days, and various pro-inflammatory cytokines were secreted through ADSCs death. The efficacy of RP@ADSCs-mediated immunotherapy was evaluated in mouse models bearing GL-26 (glioblastoma) and K1735 (melanoma), and it was found that RP resulted in an increase in the population of long-term cytotoxic T cells and a decrease in the population of regulatory T cells. This shows that RP@ADSCs have potential and applicability as an excellent anticancer immunotherapy agent in clinical practice.


Assuntos
Imunoterapia , Necroptose , Humanos , Animais , Camundongos , Necroptose/genética , Imunoterapia/métodos , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Células-Tronco/metabolismo , Células-Tronco/imunologia , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/genética , Neoplasias/patologia , Linhagem Celular Tumoral
5.
Nat Immunol ; 25(7): 1231-1244, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38898157

RESUMO

To understand the role of T cells in the pathogenesis of ulcerative colitis (UC), we analyzed colonic T cells isolated from patients with UC and controls. Here we identified colonic CD4+ and CD8+ T lymphocyte subsets with gene expression profiles resembling stem-like progenitors, previously reported in several mouse models of autoimmune disease. Stem-like T cells were increased in inflamed areas compared to non-inflamed regions from the same patients. Furthermore, TCR sequence analysis indicated stem-like T cells were clonally related to proinflammatory T cells, suggesting their involvement in sustaining effectors that drive inflammation. Using an adoptive transfer colitis model in mice, we demonstrated that CD4+ T cells deficient in either BCL-6 or TCF1, transcription factors that promote T cell stemness, had decreased colon T cells and diminished pathogenicity. Our results establish a strong association between stem-like T cell populations and UC pathogenesis, highlighting the potential of targeting this population to improve clinical outcomes.


Assuntos
Colite Ulcerativa , Fator 1-alfa Nuclear de Hepatócito , Colite Ulcerativa/imunologia , Colite Ulcerativa/patologia , Humanos , Animais , Camundongos , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 1-alfa Nuclear de Hepatócito/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD4-Positivos/imunologia , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/genética , Células-Tronco/imunologia , Células-Tronco/metabolismo , Feminino , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Camundongos Knockout , Colo/imunologia , Colo/patologia , Masculino , Camundongos Endogâmicos C57BL , Transferência Adotiva , Modelos Animais de Doenças , Adulto , Pessoa de Meia-Idade
8.
Nat Commun ; 15(1): 5176, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38890279

RESUMO

The longevity of grafts remains a major challenge in allogeneic transplantation due to immune rejection. Systemic immunosuppression can impair graft function and can also cause severe adverse effects. Here, we report a local immuno-protective strategy to enhance post-transplant persistence of allografts using a mesenchymal stem cell membrane-derived vesicle (MMV)-crosslinked hydrogel (MMV-Gel). MMVs are engineered to upregulate expression of Fas ligand (FasL) and programmed death ligand 1 (PD-L1). The MMVs are retained within the hydrogel by crosslinking. The immuno-protective microenvironment of the hydrogel protects allografts by presenting FasL and PD-L1. The binding of these ligands to T effector cells, the dominant contributors to graft destruction and rejection, results in apoptosis of T effector cells and generation of regulatory T cells. We demonstrate that implantation with MMV-Gel prolongs the survival and function of grafts in mouse models of allogeneic pancreatic islet cells and skin transplantation.


Assuntos
Membrana Celular , Reagentes de Ligações Cruzadas , Hidrogéis , Transplante das Ilhotas Pancreáticas , Transplante de Pele , Reagentes de Ligações Cruzadas/química , Hidrogéis/química , Aloenxertos , Células-Tronco/imunologia , Membrana Celular/imunologia , Proteína Ligante Fas/imunologia , Antígeno B7-H1/imunologia , Proliferação de Células , Linfócitos T/imunologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/cirurgia , Pele/imunologia , Ativação Linfocitária , Feminino , Animais , Camundongos , Sobrevivência de Enxerto , Transplante Homólogo
9.
Immunity ; 57(5): 933-935, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38749394

RESUMO

Stem cells heal wounds. In this issue of Immunity, Luan et al. demonstrate that epidermal stem cells orchestrate the recruitment of regulatory T (Treg) cells and neutrophils during wound healing. Treg cells facilitate a tolerogenic environment to protect epithelial regeneration while neutrophils promote inflammation to ward off infection.


Assuntos
Neutrófilos , Células-Tronco , Linfócitos T Reguladores , Cicatrização , Cicatrização/imunologia , Humanos , Células-Tronco/imunologia , Linfócitos T Reguladores/imunologia , Animais , Neutrófilos/imunologia
10.
Immunity ; 57(7): 1567-1585.e5, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38821051

RESUMO

Conventional dendritic cells (cDC) are antigen-presenting cells comprising cDC1 and cDC2, responsible for priming naive CD8+ and CD4+ T cells, respectively. Recent studies have unveiled cDC2 heterogeneity and identified various cDC2 progenitors beyond the common DC progenitor (CDP), hinting at distinct cDC2 lineages. By generating Cd300ciCre-hCD2R26tdTomato reporter mice, we identified a bone marrow pro-cDC2 progenitor exclusively generating cDC2 in vitro and in vivo. Single-cell analyses and multiparametric flow cytometry demonstrated that pro-cDC2 encompasses myeloid-derived pre-cDC2 and lymphoid-derived plasmacytoid DC (pDC)-like precursors differentiating into a transcriptionally convergent cDC2 phenotype. Cd300c-traced cDC2 had distinct transcriptomic profiles, phenotypes, and tissue distributions compared with Ms4a3CreR26tdTomato lineage-traced DC3, a monocyte-DC progenitor (MDP)-derived subset that bypasses CDP. Mice with reduced Cd300c-traced cDC2 showed impaired humoral responses to T cell-dependent antigens. We conclude that progenitors of distinct lineages shape the diversity of mature cDC2 across tissues. Thus, ontogenesis may impact tissue immune responses.


Assuntos
Diferenciação Celular , Linhagem da Célula , Células Dendríticas , Animais , Células Dendríticas/imunologia , Camundongos , Diferenciação Celular/imunologia , Camundongos Endogâmicos C57BL , Análise de Célula Única , Células-Tronco/citologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Camundongos Transgênicos
11.
Immunol Rev ; 325(1): 9-22, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38804499

RESUMO

Stem-like T cells are characterized by their ability to self-renew, survive long-term, and give rise to a heterogeneous pool of effector and memory T cells. Recent advances in single-cell RNA-sequencing (scRNA-seq) and lineage tracing technologies revealed an important role for stem-like T cells in both autoimmunity and cancer. In cancer, stem-like T cells constitute an important arm of the anti-tumor immune response by giving rise to effector T cells that mediate tumor control. In contrast, in autoimmunity stem-like T cells perform an unfavorable role by forming a reservoir of long-lived autoreactive cells that replenish the pathogenic, effector T-cell pool and thereby driving disease pathology. This review provides background on the discovery of stem-like T cells and their function in cancer and autoimmunity. Moreover, the influence of the microbiota and metabolism on the stem-like T-cell pool is summarized. Lastly, the implications of our knowledge about stem-like T cells for clinical treatment strategies for cancer and autoimmunity will be discussed.


Assuntos
Autoimunidade , Neoplasias , Linfócitos T , Humanos , Neoplasias/imunologia , Animais , Linfócitos T/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Doenças Autoimunes/imunologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Diferenciação Celular
12.
Nature ; 629(8011): 417-425, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38658748

RESUMO

Cancer-specific TCF1+ stem-like CD8+ T cells can drive protective anticancer immunity through expansion and effector cell differentiation1-4; however, this response is dysfunctional in tumours. Current cancer immunotherapies2,5-9 can promote anticancer responses through TCF1+ stem-like CD8+ T cells in some but not all patients. This variation points towards currently ill-defined mechanisms that limit TCF1+CD8+ T cell-mediated anticancer immunity. Here we demonstrate that tumour-derived prostaglandin E2 (PGE2) restricts the proliferative expansion and effector differentiation of TCF1+CD8+ T cells within tumours, which promotes cancer immune escape. PGE2 does not affect the priming of TCF1+CD8+ T cells in draining lymph nodes. PGE2 acts through EP2 and EP4 (EP2/EP4) receptor signalling in CD8+ T cells to limit the intratumoural generation of early and late effector T cell populations that originate from TCF1+ tumour-infiltrating CD8+ T lymphocytes (TILs). Ablation of EP2/EP4 signalling in cancer-specific CD8+ T cells rescues their expansion and effector differentiation within tumours and leads to tumour elimination in multiple mouse cancer models. Mechanistically, suppression of the interleukin-2 (IL-2) signalling pathway underlies the PGE2-mediated inhibition of TCF1+ TIL responses. Altogether, we uncover a key mechanism that restricts the IL-2 responsiveness of TCF1+ TILs and prevents anticancer T cell responses that originate from these cells. This study identifies the PGE2-EP2/EP4 axis as a molecular target to restore IL-2 responsiveness in anticancer TILs to achieve cancer immune control.


Assuntos
Linfócitos T CD8-Positivos , Proliferação de Células , Dinoprostona , Linfócitos do Interstício Tumoral , Neoplasias , Células-Tronco , Evasão Tumoral , Animais , Feminino , Humanos , Masculino , Camundongos , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Dinoprostona/metabolismo , Modelos Animais de Doenças , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Interleucina-2 , Linfonodos/citologia , Linfonodos/imunologia , Linfócitos do Interstício Tumoral/citologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Neoplasias/prevenção & controle , Receptores de Prostaglandina E Subtipo EP2/deficiência , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Receptores de Prostaglandina E Subtipo EP4/deficiência , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Evasão Tumoral/imunologia
14.
Immunity ; 57(5): 1071-1086.e7, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38677291

RESUMO

Following tissue damage, epithelial stem cells (SCs) are mobilized to enter the wound, where they confront harsh inflammatory environments that can impede their ability to repair the injury. Here, we investigated the mechanisms that protect skin SCs within this inflammatory environment. Characterization of gene expression profiles of hair follicle SCs (HFSCs) that migrated into the wound site revealed activation of an immune-modulatory program, including expression of CD80, major histocompatibility complex class II (MHCII), and CXC motif chemokine ligand 5 (CXCL5). Deletion of CD80 in HFSCs impaired re-epithelialization, reduced accumulation of peripherally generated Treg (pTreg) cells, and increased infiltration of neutrophils in wounded skin. Importantly, similar wound healing defects were also observed in mice lacking pTreg cells. Our findings suggest that upon skin injury, HFSCs establish a temporary protective network by promoting local expansion of Treg cells, thereby enabling re-epithelialization while still kindling inflammation outside this niche until the barrier is restored.


Assuntos
Antígeno B7-1 , Folículo Piloso , Inflamação , Pele , Células-Tronco , Linfócitos T Reguladores , Cicatrização , Animais , Linfócitos T Reguladores/imunologia , Camundongos , Cicatrização/imunologia , Pele/imunologia , Pele/lesões , Pele/patologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Inflamação/imunologia , Folículo Piloso/imunologia , Antígeno B7-1/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reepitelização/imunologia , Movimento Celular/imunologia , Proliferação de Células
15.
Nature ; 629(8010): 201-210, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38600376

RESUMO

Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment of haematological malignancies such as acute lymphoblastic leukaemia, B cell lymphoma and multiple myeloma1-4, but the efficacy of CAR T cell therapy in solid tumours has been limited5. This is owing to a number of factors, including the immunosuppressive tumour microenvironment that gives rise to poorly persisting and metabolically dysfunctional T cells. Analysis of anti-CD19 CAR T cells used clinically has shown that positive treatment outcomes are associated with a more 'stem-like' phenotype and increased mitochondrial mass6-8. We therefore sought to identify transcription factors that could enhance CAR T cell fitness and efficacy against solid tumours. Here we show that overexpression of FOXO1 promotes a stem-like phenotype in CAR T cells derived from either healthy human donors or patients, which correlates with improved mitochondrial fitness, persistence and therapeutic efficacy in vivo. This work thus reveals an engineering approach to genetically enforce a favourable metabolic phenotype that has high translational potential to improve the efficacy of CAR T cells against solid tumours.


Assuntos
Proteína Forkhead Box O1 , Imunoterapia Adotiva , Neoplasias , Receptores de Antígenos Quiméricos , Células-Tronco , Linfócitos T , Humanos , Camundongos , Linhagem Celular Tumoral , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Mitocôndrias/metabolismo , Fenótipo , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/citologia , Microambiente Tumoral/imunologia , Células-Tronco/citologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia
16.
Cell Stem Cell ; 31(5): 597-616, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38593798

RESUMO

Advances in modern medicine have enabled a rapid increase in lifespan and, consequently, have highlighted the immune system as a key driver of age-related disease. Immune regeneration therapies present exciting strategies to address age-related diseases by rebooting the host's primary lymphoid tissues or rebuilding the immune system directly via biomaterials or artificial tissue. Here, we identify important, unanswered questions regarding the safety and feasibility of these therapies. Further, we identify key design parameters that should be primary considerations guiding technology design, including timing of application, interaction with the host immune system, and functional characterization of the target patient population.


Assuntos
Células-Tronco , Humanos , Células-Tronco/imunologia , Células-Tronco/citologia , Animais , Transplante de Células-Tronco , Imunidade , Sistema Imunitário
17.
Adv Sci (Weinh) ; 11(28): e2307545, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38666393

RESUMO

Adapted immune cells are known to develop memory functions that increase resistance to subsequent infections after initial pathogen exposure, however, it is unclear whether non-immune cells, like tissue-resident stem cells, have similar memory functions. Here, it is found that tissue-resident stem cells crucial for tissue regeneration show diminished adverse effects on diverse stem cell functions against successive exposure to foreign antigen (ß-glucan) to maintain tissue homeostasis and stability both in vitro and in vivo. These data suggest that endometrial stem cells may possess a robust memory function, in contrast, fully differentiated cells like fibroblasts and vesicular cells do not show these memory mechanisms upon consecutive antigen exposure. Moreover, the pivotal role of Angiopoietin-like 4 (ANGPTL4) in regulating the memory functions of endometrial stem cells is identified through specific shRNA knockdown in vitro and knockout mice in vivo experiments. ANGPTL4 is associated with the alteration of diverse stem cell functions and epigenetic modifications, notably through histone H3 methylation changes and two pathways (i.e., PI3K/Akt and FAK/ERK1/2 signaling) upon consecutive antigen exposure. These findings imply the existence of inherent self-defense mechanisms through which local stem cells can adapt and protect themselves from recurrent antigenic challenges, ultimately mitigating adverse consequences.


Assuntos
Proteína 4 Semelhante a Angiopoietina , Camundongos Knockout , Células-Tronco , Animais , Camundongos , Proteína 4 Semelhante a Angiopoietina/genética , Proteína 4 Semelhante a Angiopoietina/metabolismo , Proteína 4 Semelhante a Angiopoietina/imunologia , Células-Tronco/metabolismo , Células-Tronco/imunologia , Feminino , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Memória Imunológica/imunologia , Diferenciação Celular/imunologia
18.
FEBS Lett ; 598(11): 1354-1365, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38594179

RESUMO

Chronic infection with the hepatitis B virus (HBV) induces progressive hepatic impairment. Achieving complete eradication of the virus remains a formidable challenge. Cytotoxic T lymphocytes, specific to viral antigens, either exhibit a numerical deficiency or succumb to an exhausted state in individuals chronically afflicted with HBV. The comprehension of the genesis and dissemination of stem cell memory T cells (TSCMs) targeting HBV remains inadequately elucidated. We identified TSCMs in subjects with chronic HBV infection and scrutinized their efficacy in a murine model with human hepatocyte transplants, specifically the TK-NOG mice. TSCMs were discerned in all subjects under examination. Introduction of TSCMs into the HBV mouse model precipitated a severe necro-inflammatory response, resulting in the elimination of human hepatocytes. TSCMs may constitute a valuable tool in the pursuit of a remedial therapy for HBV infection.


Assuntos
Diferenciação Celular , Vírus da Hepatite B , Hepatócitos , Células T de Memória , Linfócitos T Citotóxicos , Animais , Humanos , Hepatócitos/virologia , Hepatócitos/imunologia , Hepatócitos/transplante , Vírus da Hepatite B/imunologia , Vírus da Hepatite B/fisiologia , Linfócitos T Citotóxicos/imunologia , Camundongos , Diferenciação Celular/imunologia , Células T de Memória/imunologia , Hepatite B Crônica/imunologia , Hepatite B Crônica/virologia , Masculino , Feminino , Modelos Animais de Doenças , Células-Tronco/virologia , Células-Tronco/imunologia , Células-Tronco/citologia , Adulto
19.
Trends Plant Sci ; 29(7): 715-717, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38331684

RESUMO

Pathogenic viruses are a constant threat to all organisms, including plants. However, in plants, a small group of cells (stem cells) protect themselves from viral invasion. Recently, Incarbone et al. uncovered a novel salicylic acid (SA) and RNAi mechanism of stem cell resistance, broadening our understanding of RNAi-mediated antiviral plant immunity.


Assuntos
Doenças das Plantas , Imunidade Vegetal , Ácido Salicílico , Ácido Salicílico/metabolismo , Doenças das Plantas/virologia , Doenças das Plantas/imunologia , Imunidade Inata , Interferência de RNA , Plantas/imunologia , Plantas/virologia , Vírus de Plantas/fisiologia , Células-Tronco/imunologia
20.
J Allergy Clin Immunol ; 153(4): 1125-1139, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38072195

RESUMO

BACKGROUND: Inborn errors of immunity (IEI) often lack specific disease models and personalized management. Signal transducer and activator of transcription (STAT)-1 gain of function (GoF) is such example of an IEI with diverse clinical phenotype with unclear pathomechanisms and unpredictable response to therapy. Limitations in obtaining fresh samples for functional testing and research further highlights the need for patient-specific ex vivo platforms. OBJECTIVE: Using STAT1-GoF as an example IEI, we investigated the potential of patient-derived expanded potential stem cells (EPSC) as an ex vivo platform for disease modeling and personalized treatment. METHODS: We generated EPSC derived from individual STAT1-GoF patients. STAT1 mutations were confirmed with Sanger sequencing. Functional testing including STAT1 phosphorylation/dephosphorylation and gene expression with or without Janus activating kinase inhibitors were performed. Functional tests were repeated on EPSC lines with GoF mutations repaired by CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) editing. RESULTS: EPSC were successfully reprogrammed from STAT1-GoF patients and expressed the same pluripotent makers as controls, with distinct morphologic differences. Patient-derived EPSC recapitulated the functional abnormalities of index STAT1-GoF patients with STAT1 hyperphosphorylation and increased expression of STAT1 and its downstream genes (IRF1, APOL6, and OAS1) after IFN-γ stimulation. Addition of ruxolitinib and baricitinib inhibited STAT1 hyperactivation in STAT1-GoF EPSC in a dose-dependent manner, which was not observed with tofacitinib. Corrected STAT1 phosphorylation and downstream gene expression were observed among repaired STAT1-GoF EPSC cell lines. CONCLUSION: This proof-of-concept study demonstrates the potential of our patient-derived EPSC platform to model STAT1-GoF. We propose this platform when researching, recapitulating, and repairing other IEI in the future.


Assuntos
Mutação com Ganho de Função , Fator de Transcrição STAT1 , Células-Tronco , Humanos , Mutação , Fosforilação , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Células-Tronco/imunologia , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...