Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
2.
J Clin Invest ; 133(15)2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37526084

RESUMEN

STAT3 deficiency (STAT3-/-) in donor T cells prevents graft-versus-host disease (GVHD), but the impact on graft-versus-leukemia (GVL) activity and mechanisms of GVHD prevention remains unclear. Here, using murine models of GVHD, we show that STAT3-/- donor T cells induced only mild reversible acute GVHD while preserving GVL effects against nonsusceptible acute lymphoblastic leukemia (ALL) cells in a donor T cell dose-dependent manner. GVHD prevention depended on programmed death ligand 1/programmed cell death protein 1 (PD-L1/PD-1) signaling. In GVHD target tissues, STAT3 deficiency amplified PD-L1/PD-1 inhibition of glutathione (GSH)/Myc pathways that regulate metabolic reprogramming in activated T cells, with decreased glycolytic and mitochondrial ATP production and increased mitochondrial ROS production and dysfunction, leading to tissue-specific deletion of host-reactive T cells and prevention of GVHD. Mitochondrial STAT3 deficiency alone did not reduce GSH expression or prevent GVHD. In lymphoid tissues, the lack of host-tissue PD-L1 interaction with PD-1 reduced the inhibition of the GSH/Myc pathway despite reduced GSH production caused by STAT3 deficiency and allowed donor T cell functions that mediate GVL activity. Therefore, STAT3 deficiency in donor T cells augments PD-1 signaling-mediated inhibition of GSH/Myc pathways and augments dysfunction of T cells in GVHD target tissues while sparing T cells in lymphoid tissues, leading to prevention of GVHD while preserving GVL effects.


Asunto(s)
Enfermedad Injerto contra Huésped , Leucemia , Ratones , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/prevención & control , Linfocitos T/metabolismo , Efecto Injerto vs Leucemia/genética , Trasplante de Médula Ósea
3.
Cell Rep Methods ; 3(5): 100465, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37323577

RESUMEN

Cellular differentiation requires global changes to DNA methylation (DNAme), where it functions to regulate transcription factor, chromatin remodeling activity, and genome interpretation. Here, we describe a simple DNAme engineering approach in pluripotent stem cells (PSCs) that stably extends DNAme across target CpG islands (CGIs). Integration of synthetic CpG-free single-stranded DNA (ssDNA) induces a target CpG island methylation response (CIMR) in multiple PSC lines, Nt2d1 embryonal carcinoma cells, and mouse PSCs but not in highly methylated CpG island hypermethylator phenotype (CIMP)+ cancer lines. MLH1 CIMR DNAme spanned the CGI, was precisely maintained through cellular differentiation, suppressed MLH1 expression, and sensitized derived cardiomyocytes and thymic epithelial cells to cisplatin. Guidelines for CIMR editing are provided, and initial CIMR DNAme is characterized at TP53 and ONECUT1 CGIs. Collectively, this resource facilitates CpG island DNAme engineering in pluripotency and the genesis of novel epigenetic models of development and disease.


Asunto(s)
Metilación de ADN , Neoplasias , Animales , Ratones , Metilación de ADN/genética , Islas de CpG/genética , ADN de Cadena Simple/metabolismo , Neoplasias/genética , Células Epiteliales/metabolismo
4.
Am J Transplant ; 23(8): 1116-1129, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37105316

RESUMEN

Induction of major histocompatibility complex (MHC) human leukocyte antigen (HLA)-mismatched mixed chimerism is a promising approach for organ transplantation tolerance; however, human leukocyte antigen-mismatched stable mixed chimerism has not been achieved in the clinic. Tolerogenic dendritic cell (DC) expression of MHC class II (MHC II) and programmed cell death 1 ligand 1 (PD-L1) is important for immune tolerance, but whether donor-MHC II or PD-L1 is required for the induction of stable MHC-mismatched mixed chimerism and transplant tolerance is unclear. Here, we show that a clinically applicable radiation-free regimen can establish stable MHC-mismatched mixed chimerism and organ transplant tolerance in murine models. Induction of MHC-mismatched mixed chimerism does not require donor cell expression of MHC II or PD-L1, but donor-type organ transplant tolerance in the mixed chimeras (MC) requires the donor hematopoietic cells and the organ transplants to express PD-L1. The PD-L1 expressed by donor hematopoietic cells and the programmed cell death 1 expressed by host cells augment host-type donor-reactive CD4+ and CD8+ T cell anergy/exhaustion and differentiation into peripheral regulatory T (pTreg) cells in association with the organ transplant tolerance in the MC. Conversely, host-type Treg cells augment the expansion of donor-type tolerogenic CD8+ DCs that express PD-L1. These results indicate that PD-L1 expressed by donor-type tolerogenic DCs and expansion of host-type pTreg cells in MHC-mismatched MCs play critical roles in mediating organ transplant tolerance.


Asunto(s)
Trasplante de Órganos , Tolerancia al Trasplante , Ratones , Humanos , Animales , Antígeno B7-H1 , Quimerismo , Antígenos de Histocompatibilidad Clase II , Complejo Mayor de Histocompatibilidad , Antígenos HLA , Tolerancia Inmunológica , Quimera por Trasplante , Trasplante de Médula Ósea/métodos
5.
Proc Natl Acad Sci U S A ; 120(16): e2205085120, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37036990

RESUMEN

PD-L1 has two receptors: PD-1 and CD80. Previous reports assumed that PD-L1 and CD80 interacted in trans, but recent reports showed that only cis PD-L1/CD80 interactions existed, and prevention of cis PD-L1/CD80 interactions on antigen-presenting cells (APCs) reduced antitumor immunity via augmenting PD-L1/PD-1 and CD80/CTLA4 interactions between T and APCs. Here, using tumor-bearing mice capable of cis and trans or trans only PD-L1/CD80 interactions, we show that trans PD-L1/CD80 interactions do exist between tumor and T cells, and the effects of trans PD-L1/CD80 interactions require tumor cell expression of MHC-I and T cell expression of CD28. The blockade of PD-L1/CD80 interactions in mice with both cis and trans interactions or with only trans interactions augments antitumor immunity by expanding IFN-γ-producing CD8+ T cells and IFN-γ-dependent NOS2-expressing tumor-associated macrophages. Our studies indicate that although cis and trans PD-L1/CD80 interactions may have opposite effects on antitumor immunity, the net effect of blocking PD-L1/CD80 interactions in vivo augments CD8+ T cell-mediated antitumor immunity.


Asunto(s)
Antígeno B7-H1 , Linfocitos T CD8-positivos , Ratones , Animales , Antígeno B7-H1/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Activación de Linfocitos , Antígeno B7-1 , Moléculas de Adhesión Celular
6.
Gastroenterology ; 164(1): 89-102, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36113570

RESUMEN

BACKGROUND & AIMS: Intestinal fibrosis is a significant complication of Crohn's disease (CD). Gut microbiota reactive Th17 cells are crucial in the pathogenesis of CD; however, how Th17 cells induce intestinal fibrosis is still not completely understood. METHODS: In this study, T-cell transfer model with wild-type (WT) and Areg-/- Th17 cells and dextran sulfate sodium (DSS)-induced chronic colitis model in WT and Areg-/- mice were used. CD4+ T-cell expression of AREG was determined by quantitative reverse-transcriptase polymerase chain reaction and enzyme-linked immunosorbent assay. The effect of AREG on proliferation/migration/collagen expression in human intestinal myofibroblasts was determined. AREG expression was assessed in healthy controls and patients with CD with or without intestinal fibrosis. RESULTS: Although Th1 and Th17 cells induced intestinal inflammation at similar levels when transferred into Tcrßxδ-/- mice, Th17 cells induced more severe intestinal fibrosis. Th17 cells expressed higher levels of AREG than Th1 cells. Areg-/- mice developed less severe intestinal fibrosis compared with WT mice on DSS insults. Transfer of Areg-/- Th17 cells induced less severe fibrosis in Tcrßxδ-/- mice compared with WT Th17 cells. Interleukin (IL)6 and IL21 promoted AREG expression in Th17 cells by activating Stat3. Stat3 inhibitor suppressed Th17-induced intestinal fibrosis. AREG promoted human intestinal myofibroblast proliferation, motility, and collagen I expression, which was mediated by activating mammalian target of rapamycin and MEK. AREG expression was increased in intestinal CD4+ T cells in fibrotic sites compared with nonfibrotic sites from patients with CD. CONCLUSIONS: These findings reveal that Th17-derived AREG promotes intestinal fibrotic responses in experimental colitis and human patients with CD. Thereby, AREG might serve as a potential therapeutic target for fibrosis in CD.


Asunto(s)
Colitis , Enfermedad de Crohn , Animales , Humanos , Ratones , Anfirregulina/genética , Anfirregulina/metabolismo , Colitis/metabolismo , Colágeno/metabolismo , Enfermedad de Crohn/patología , Sulfato de Dextran/efectos adversos , Fibrosis , Mucosa Intestinal/patología , Ratones Endogámicos C57BL , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Miofibroblastos/patología , Células Th17/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
7.
Blood Adv ; 7(17): 4886-4902, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-36322878

RESUMEN

Chronic graft-versus-host disease (cGvHD) remains a prominent barrier to allogeneic hematopoietic stem cell transplantion as the leading cause of nonrelapse mortality and significant morbidity. Tremendous progress has been achieved in both the understanding of pathophysiology and the development of new therapies for cGvHD. Although our field has historically approached treatment from an empiric position, research performed at the bedside and bench has elucidated some of the complex pathophysiology of cGvHD. From the clinical perspective, there is significant variability of disease manifestations between individual patients, pointing to diverse biological underpinnings. Capitalizing on progress made to date, the field is now focused on establishing personalized approaches to treatment. The intent of this article is to concisely review recent knowledge gained and formulate a path toward patient-specific cGvHD therapy.


Asunto(s)
Síndrome de Bronquiolitis Obliterante , Enfermedad Injerto contra Huésped , Humanos , Consenso , Medicina de Precisión , Enfermedad Injerto contra Huésped/terapia , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Biología
8.
Blood ; 140(25): 2740-2753, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36084473

RESUMEN

Chronic graft-versus-host disease (cGVHD) is an autoimmune-like syndrome. CXCR5-PD-1hi peripheral T-helper (Tph) cells have an important pathogenic role in autoimmune diseases, but the role of Tph cells in cGVHD remains unknown. We show that in patients with cGVHD, expansion of Tph cells among blood CD4+ T cells was associated with cGVHD severity. These cells augmented memory B-cell differentiation and production of immunoglobulin G via interleukin 21 (IL-21). Tph cell expansion was also observed in a murine model of cGVHD. This Tph cell expansion in the blood is associated with the expansion of pathogenic tissue-resident T-helper (Trh) cells that form lymphoid aggregates surrounded by collagen in graft-versus-host disease (GVHD) target tissues. Adoptive transfer experiments showed that Trh cells from GVHD target tissues give rise to Tph cells in the blood, and conversely, Tph cells from the blood give rise to Trh cells in GVHD target tissues. Tph cells in the blood and Trh cells in GVHD target tissues had highly overlapping T-cell receptor α and ß repertoires. Deficiency of IL-21R, B-cell lymphoma 6 (BCL6), or T-bet in donor T cells markedly reduced the proportions of Tph cells in the blood and Trh cells in GVHD target tissues and reduced T-B interaction in the lymphoid aggregates. These results indicate that clonally related pathogenic Tph cells and Trh cells traffic between the blood and cGVHD target tissues, and that IL-21R-BCL6 signaling and T-bet are required for the development and expansion of Tph and Trh cells in the pathogenesis of cGVHD.


Asunto(s)
Síndrome de Bronquiolitis Obliterante , Enfermedad Injerto contra Huésped , Humanos , Ratones , Animales , Linfocitos T Colaboradores-Inductores , Linfocitos T CD4-Positivos , Linfocitos B/patología , Enfermedad Crónica
9.
ACS Omega ; 7(37): 33482-33490, 2022 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-36157751

RESUMEN

Designing economical and nonprecious catalysts with a catalytic performance as good as that of noble metals is of great importance in future renewable bioenergy production. In this study, the metal-organic framework (MOF) was applied as a precursor template to synthesize Co3O4 nanoparticles with a carbon matrix shell (denoted as M-Co3O4). To select the synthesized optimal catalyst, stearic acid was chosen as the model reactant. The effects of catalyst dosage, methanol dosage, water dosage, temperature, and reaction time on catalytic efficiency were examined. Under the designed condition, M-Co3O4 exhibited high catalytic performance and the catalyst showed higher conversion of stearic acid (98.7%) and selectivity toward C8-C18 alkanes (92.2%) in comparison with Pt/C (95.8% conversion and 93.2% selectivity toward C8-C18). Furthermore, a series of characterization techniques such as scanning electron microscopy (SEM), high-resolution transmission electron microscopy (HRTEM), X-ray powder diffraction (XRD), X-ray photoelectron spectroscopy (XPS), nitrogen adsorption isotherms (Brunauer-Emmett-Teller (BET) method), and thermogravimetric analysis (TGA) was applied to investigate the physicochemical properties of the catalysts. Finally, we proposed that decarbonization (deCO) could be the presumably mechanistic pathway for the production of C8-C18 alkanes from the decomposition of stearic acid.

10.
Comput Math Methods Med ; 2022: 4525892, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36017154

RESUMEN

Background: Intracranial infection is a serious complication after neurosurgery. According to a survey, the incidence of intracranial infection is about 2.2%-2.6%, and patients with severe symptoms may even pose a threat to their life safety. Objective: To explore the risk factors for intracranial infection caused by Acinetobacter baumannii after surgery and the clinical effect of sequential therapy of cefoperazone/sulbactam sodium. Methods: In this study, a retrospective study was used. In this case-control study, 48 cases of intracranial Acinetobacter baumannii infection after neurosurgery in our hospital from January 2016 to December 2021 were selected as the infection group, and 96 patients without intracranial infection after surgery during the same period were selected as the control group to study all kinds of related factors and analyze the risk factors for intracranial Acinetobacter baumannii infection; in addition, in accordance with the therapeutic regimen for anti-infection, the infection group was divided into the tigecycline group (patients with tigecycline therapy in this group) and the combined group (patients with tigecycline combined with cefoperazone/sulbactam sequential therapy), with 24 cases in each group in order to compare the therapeutic effects of the two groups. Results: Logistic regression factor model results show that increasing age of patients, surgical treatment for intracranial tumor or craniocerebral trauma, postoperative drainage time (≥3 days), and postoperative hospital stay (≥10 days) were the risk factors for postoperative intracranial infection of Acinetobacter baumannii in neurosurgical patients (P < 0.05), and postoperative prophylactic antibiotic treatment can reduce the incidence of intracranial infection (P < 0.05). The cerebrospinal fluid nucleated cell count, serum CRP, and serum PCT in the combined group 72 h after treatment were lower than those in the tigecycline group, and the difference was statistically significant (P < 0.05). Compared with the clinical efficacy after 72-hour treatment, the cure rate and effective rate in the combined treatment group were 83.33% and 16.67%, respectively, and those in the tigecycline group were 54.17% and 33.33%, respectively. The invalid interest rate was 12.50%, and the combined treatment group was superior to the tigecycline group (P < 0.05). Conclusion: For patients with craniocerebral surgery, targeted preventive interventions should be carried out for the risk factors that may lead to intracranial Acinetobacter baumannii infection. The clinical effect of tigecycline combined with cefoperazone and sulbactam sodium sequentially in the treatment of intracranial Acinetobacter baumannii infection is better.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Infección Hospitalaria , Infecciones por Acinetobacter/tratamiento farmacológico , Antibacterianos/uso terapéutico , Estudios de Casos y Controles , Cefoperazona/efectos adversos , Infección Hospitalaria/tratamiento farmacológico , Humanos , Estudios Retrospectivos , Sodio , Sulbactam/farmacología , Sulbactam/uso terapéutico , Tigeciclina/farmacología , Tigeciclina/uso terapéutico
11.
Front Immunol ; 13: 907673, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35677056

RESUMEN

Allogeneic hematopoietic cell transplantation (Allo-HCT) is a curative therapy for hematological malignancies (i.e., leukemia and lymphoma) due to the graft-versus-leukemia (GVL) activity mediated by alloreactive T cells that can eliminate residual malignant cells and prevent relapse. However, the same alloreactive T cells can cause a serious side effect, known as graft-versus-host disease (GVHD). GVHD and GVL occur in distinct organ and tissues, with GVHD occurring in target organs (e.g., the gut, liver, lung, skin, etc.) and GVL in lympho-hematopoietic tissues where hematological cancer cells primarily reside. Currently used immunosuppressive drugs for the treatment of GVHD inhibit donor T cell activation and expansion, resulting in a decrease in both GVHD and GVL activity that is associated with cancer relapse. To prevent GVHD, it is important to allow full activation and expansion of alloreactive T cells in the lympho-hematopoietic tissues, as well as prevent donor T cells from migrating into the GVHD target tissues, and tolerize infiltrating T cells via protective mechanisms, such as PD-L1 interacting with PD-1, in the target tissues. In this review, we will summarize major approaches that prevent donor T cell migration into GVHD target tissues and approaches that augment tolerization of the infiltrating T cells in the GVHD target tissues while preserving strong GVL activity in the lympho-hematopoietic tissues.


Asunto(s)
Enfermedad Injerto contra Huésped , Neoplasias Hematológicas , Leucemia , Antígeno B7-H1 , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/prevención & control , Efecto Injerto vs Leucemia , Neoplasias Hematológicas/terapia , Humanos , Recurrencia , Linfocitos T/patología
12.
Front Immunol ; 13: 844271, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35251043

RESUMEN

Intestinal graft-versus-host disease (Gut-GVHD) is one of the major causes of mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). While systemic glucocorticoids (GCs) comprise the first-line treatment option, the response rate for GCs varies from 30% to 50%. The prognosis for patients with steroid-refractory acute Gut-GVHD (SR-Gut-aGVHD) remains dismal. The mechanisms underlying steroid resistance are unclear, and apart from ruxolitinib, there are no approved treatments for SR-Gut-aGVHD. In this review, we provide an overview of the current biological understanding of experimental SR-Gut-aGVHD pathogenesis, the advanced technology that can be applied to the human SR-Gut-aGVHD studies, and the potential novel therapeutic options for patients with SR-Gut-aGVHD.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Animales , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Ratones , Pronóstico , Esteroides/uso terapéutico
13.
PLoS Biol ; 20(2): e3001552, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35180231

RESUMEN

Regulatory T (Treg) cells are critical in preventing aberrant immune responses. Posttranscriptional control of gene expression by microRNA (miRNA) has recently emerged as an essential genetic element for Treg cell function. Here, we report that mice with Treg cell-specific ablation of miR-142 (hereafter Foxp3CremiR-142fl/fl mice) developed a fatal systemic autoimmune disorder due to a breakdown in peripheral T-cell tolerance. Foxp3CremiR-142fl/fl mice displayed a significant decrease in the abundance and suppressive capacity of Treg cells. Expression profiling of miR-142-deficient Treg cells revealed an up-regulation of multiple genes in the interferon gamma (IFNγ) signaling network. We identified several of these IFNγ-associated genes as direct miR-142-3p targets and observed excessive IFNγ production and signaling in miR-142-deficient Treg cells. Ifng ablation rescued the Treg cell homeostatic defect and alleviated development of autoimmunity in Foxp3CremiR-142fl/fl mice. Thus, our findings implicate miR-142 as an indispensable regulator of Treg cell homeostasis that exerts its function by attenuating IFNγ responses.


Asunto(s)
Autoinmunidad/inmunología , Regulación de la Expresión Génica/inmunología , Homeostasis/inmunología , MicroARNs/inmunología , Linfocitos T Reguladores/inmunología , Enfermedad Aguda , Animales , Autoinmunidad/genética , Trasplante de Médula Ósea/métodos , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica/métodos , Enfermedad Injerto contra Huésped/inmunología , Homeostasis/genética , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , MicroARNs/genética , RNA-Seq/métodos , Transducción de Señal/genética , Linfocitos T Reguladores/metabolismo
14.
Front Immunol ; 12: 700857, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34539630

RESUMEN

Allogeneic hematopoietic cell transplantation (allo-HCT) is a curative therapy for hematologic malignancies, but its success is complicated by graft-versus-host disease (GVHD). GVHD can be divided into acute and chronic types. Acute GVHD represents an acute alloimmune inflammatory response initiated by donor T cells that recognize recipient alloantigens. Chronic GVHD has a more complex pathophysiology involving donor-derived T cells that recognize recipient-specific antigens, donor-specific antigens, and antigens shared by the recipient and donor. Antibodies produced by donor B cells contribute to the pathogenesis of chronic GVHD but not acute GVHD. Acute GVHD can often be effectively controlled by treatment with corticosteroids or other immunosuppressant for a period of weeks, but successful control of chronic GVHD requires much longer treatment. Therefore, chronic GVHD remains the major cause of long-term morbidity and mortality after allo-HCT. Murine models of allo-HCT have made great contributions to our understanding pathogenesis of acute and chronic GVHD. In this review, we summarize new mechanistic findings from murine models of chronic GVHD, and we discuss the relevance of these insights to chronic GVHD pathogenesis in humans and their potential impact on clinical prevention and treatment.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Animales , Humanos , Ratones
15.
Diabetes ; 70(12): 2837-2849, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34556496

RESUMEN

Syntaxin 4 (STX4), a plasma membrane-localized SNARE protein, regulates human islet ß-cell insulin secretion and preservation of ß-cell mass. We found that human type 1 diabetes (T1D) and NOD mouse islets show reduced ß-cell STX4 expression, consistent with decreased STX4 expression, as a potential driver of T1D phenotypes. To test this hypothesis, we generated inducible ß-cell-specific STX4-expressing NOD mice (NOD-ißSTX4). Of NOD-ißSTX4 mice, 73% had sustained normoglycemia vs. <20% of control NOD (NOD-Ctrl) mice by 25 weeks of age. At 12 weeks of age, before diabetes conversion, NOD-ißSTX4 mice demonstrated superior whole-body glucose tolerance and ß-cell glucose responsiveness than NOD-Ctrl mice. Higher ß-cell mass and reduced ß-cell apoptosis were also detected in NOD-ißSTX4 pancreata compared with pancreata of NOD-Ctrl mice. Single-cell RNA sequencing revealed that islets from NOD-ißSTX4 had markedly reduced interferon-γ signaling and tumor necrosis factor-α signaling via nuclear factor-κB in islet ß-cells, including reduced expression of the chemokine CCL5; CD4+ regulatory T cells were also enriched in NOD-ißSTX4 islets. These results provide a deeper mechanistic understanding of STX4 function in ß-cell protection and warrant further investigation of STX4 enrichment as a strategy to reverse or prevent T1D in humans or protect ß-cell grafts.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Células Secretoras de Insulina/metabolismo , Estado Prediabético/genética , Proteínas Qa-SNARE/genética , Animales , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Especificidad de Órganos/genética , Estado Prediabético/inmunología , Proteínas Qa-SNARE/metabolismo
16.
Nat Commun ; 12(1): 805, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33547295

RESUMEN

Efforts to improve the prognosis of steroid-resistant gut acute graft-versus-host-disease (SR-Gut-aGVHD) have suffered from poor understanding of its pathogenesis. Here we show that the pathogenesis of SR-Gut-aGVHD is associated with reduction of IFN-γ+ Th/Tc1 cells and preferential expansion of IL-17-IL-22+ Th/Tc22 cells. The IL-22 from Th/Tc22 cells causes dysbiosis in a Reg3γ-dependent manner. Transplantation of IFN-γ-deficient donor CD8+ T cells in the absence of CD4+ T cells produces a phenocopy of SR-Gut-aGVHD. IFN-γ deficiency in donor CD8+ T cells also leads to a PD-1-dependent depletion of intestinal protective CX3CR1hi mononuclear phagocytes (MNP), which also augments expansion of Tc22 cells. Supporting the dual regulation, simultaneous dysbiosis induction and depletion of CX3CR1hi MNP results in full-blown Gut-aGVHD. Our results thus provide insights into SR-Gut-aGVHD pathogenesis and suggest the potential efficacy of IL-22 antagonists and IFN-γ agonists in SR-Gut-aGVHD therapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Disbiosis/inmunología , Enfermedad Injerto contra Huésped/inmunología , Interferón gamma/inmunología , Interleucinas/inmunología , Fagocitos/inmunología , Animales , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/trasplante , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/inmunología , Modelos Animales de Enfermedad , Disbiosis/genética , Disbiosis/microbiología , Disbiosis/patología , Microbioma Gastrointestinal/inmunología , Regulación de la Expresión Génica , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/microbiología , Enfermedad Injerto contra Huésped/patología , Interferón gamma/deficiencia , Interferón gamma/genética , Interleucina-17/deficiencia , Interleucina-17/genética , Interleucina-17/inmunología , Interleucinas/genética , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Depleción Linfocítica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Pancreatitis/genética , Proteínas Asociadas a Pancreatitis/inmunología , Fagocitos/citología , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal , Linfocitos T Colaboradores-Inductores , Linfocitos T Reguladores , Irradiación Corporal Total , Interleucina-22
17.
Blood ; 137(16): 2243-2255, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33511398

RESUMEN

Donor T cells mediate both graft-versus-leukemia (GVL) activity and graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (allo-HCT). Development of methods that preserve GVL activity while preventing GVHD remains a long-sought goal. Tolerogenic anti-interleukin-2 (IL-2) monoclonal antibody (JES6-1) forms anti-IL-2/IL-2 complexes that block IL-2 binding to IL-2Rß and IL-2Rγ on conventional T cells that have low expression of IL-2Rα. Here, we show that administration of JES6 early after allo-HCT in mice markedly attenuates acute GVHD while preserving GVL activity that is dramatically stronger than observed with tacrolimus (TAC) treatment. The anti-IL-2 treatment downregulated activation of the IL-2-Stat5 pathway and reduced production of granulocyte-macrophage colony-stimulating factor (GM-CSF). In GVHD target tissues, enhanced T-cell programmed cell death protein 1 (PD-1) interaction with tissue-programmed cell death-ligand 1 (PD-L1) led to reduced activation of protein kinase-mammalian target of rapamycin pathway and increased expression of eomesodermin and B-lymphocyte-induced maturation protein-1, increased T-cell anergy/exhaustion, expansion of Foxp3-IL-10-producing type 1 regulatory (Tr1) cells, and depletion of GM-CSF-producing T helper type 1 (Th1)/cytotoxic T cell type 1 (Tc1) cells. In recipient lymphoid tissues, lack of donor T-cell PD-1 interaction with tissue PD-L1 preserved donor PD-1+TCF-1+Ly108+CD8+ T memory progenitors and functional effectors that have strong GVL activity. Anti-IL-2 and TAC treatments have qualitatively distinct effects on donor T cells in the lymphoid tissues, and CD8+ T memory progenitor cells are enriched with anti-IL-2 treatment compared with TAC treatment. We conclude that administration of tolerogenic anti-IL-2 monoclonal antibody early after allo-HCT represents a novel approach for preventing acute GVHD while preserving GVL activity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedad Injerto contra Huésped/prevención & control , Efecto Injerto vs Leucemia/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Interleucina-2/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Enfermedad Injerto contra Huésped/inmunología , Inmunosupresores/uso terapéutico , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Tacrolimus/uso terapéutico , Trasplante Homólogo
18.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-32931481

RESUMEN

CD4+ T cell interactions with B cells play a critical role in the pathogenesis of systemic autoimmune diseases such as systemic lupus and chronic graft-versus-host disease (cGVHD). Extrafollicular CD44hiCD62LloPSGL1loCD4+ T cells (PSGL1loCD4+ T cells) are associated with the pathogenesis of lupus and cGVHD, but their causal role has not been established. With murine and humanized MHC-/-HLA-A2+DR4+ murine models of cGVHD, we showed that murine and human PSGL1loCD4+ T cells from GVHD target tissues have features of B cell helpers with upregulated expression of programmed cell death protein 1 (PD1) and inducible T cell costimulator (ICOS) and production of IL-21. They reside in nonlymphoid tissues without circulating in the blood and have features of tissue-resident memory T cells with upregulated expression of CD69. Murine PSGL1loCD4+ T cells from GVHD target tissues augmented B cell differentiation into plasma cells and production of autoantibodies via their PD1 interaction with PD-L2 on B cells. Human PSGL1loCD4+ T cells were apposed with memory B cells in the liver tissues of humanized mice and cGVHD patients. Human PSGL1loCD4+ T cells from humanized GVHD target tissues also augmented autologous memory B cell differentiation into plasma cells and antibody production in a PD1/PD-L2-dependent manner. Further preclinical studies targeting tissue-resident T cells to treat antibody-mediated features of autoimmune diseases are warranted.


Asunto(s)
Autoinmunidad , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Enfermedad Injerto contra Huésped/inmunología , Glicoproteínas de Membrana/inmunología , Animales , Linfocitos B/patología , Linfocitos T CD4-Positivos/patología , Diferenciación Celular/genética , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Humanos , Memoria Inmunológica/genética , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología
19.
Proc Natl Acad Sci U S A ; 117(49): 31219-31230, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33229527

RESUMEN

Type 1 diabetes (T1D) results from the autoimmune destruction of ß cells, so cure of firmly established T1D requires both reversal of autoimmunity and restoration of ß cells. It is known that ß cell regeneration in nonautoimmune diabetic mice can come from differentiation of progenitors and/or transdifferentiation of α cells. However, the source of ß cell regeneration in autoimmune nonobese diabetic (NOD) mice remains unclear. Here, we show that, after reversal of autoimmunity by induction of haploidentical mixed chimerism, administration of gastrin plus epidermal growth factor augments ß cell regeneration and normalizes blood glucose in the firmly established diabetic NOD mice. Using transgenic NOD mice with inducible lineage-tracing markers for insulin-producing ß cells, Sox9+ ductal progenitors, Nestin+ mesenchymal stem cells, and glucagon-producing α cells, we have found that both reactivation of dysfunctional low-level insulin expression (insulinlo) ß cells and neogenesis contribute to the regeneration, with the latter predominantly coming from transdifferentiation of α cells. These results indicate that, after reversal of autoimmunity, reactivation of ß cells and transdifferentiation of α cells can provide sufficient new functional ß cells to reach euglycemia in firmly established T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Regeneración/genética , Animales , Autoinmunidad/genética , Glucemia/efectos de los fármacos , Transdiferenciación Celular/genética , Quimerismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Factor de Crecimiento Epidérmico/farmacología , Femenino , Gastrinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucagón/biosíntesis , Células Secretoras de Glucagón/metabolismo , Insulina/genética , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Endogámicos NOD/genética , Células Precursoras de Linfocitos B/efectos de los fármacos
20.
J Clin Invest ; 130(12): 6457-6476, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32817590

RESUMEN

Clinical trials are currently testing whether induction of haploidentical mixed chimerism (Haplo-MC) induces organ transplantation tolerance. Whether Haplo-MC can be used to treat established autoimmune diseases remains unknown. Here, we show that established autoimmunity in euthymic and adult-thymectomized NOD (H-2g7) mice was cured by induction of Haplo-MC under a non-myeloablative anti-thymocyte globulin-based conditioning regimen and infusion of CD4+ T cell-depleted hematopoietic graft from H-2b/g7 F1 donors that expressed autoimmune-resistant H-2b or from H-2s/g7 F1 donors that expressed autoimmune-susceptible H-2s. The cure was associated with enhanced thymic negative selection, increased thymic Treg (tTreg) production, and anergy or exhaustion of residual host-type autoreactive T cells in the periphery. The peripheral tolerance was accompanied by expansion of donor- and host-type CD62L-Helios+ tTregs as well as host-type Helios-Nrp1+ peripheral Tregs (pTregs) and PD-L1hi plasmacytoid DCs (pDCs). Depletion of donor- or host-type Tregs led to reduction of host-type PD-L1hi pDCs and recurrence of autoimmunity, whereas PD-L1 deficiency in host-type DCs led to reduction of host-type pDCs and Helios-Nrp1+ pTregs. Thus, induction of Haplo-MC reestablished both central and peripheral tolerance through mechanisms that depend on allo-MHC+ donor-type DCs, PD-L1hi host-type DCs, and the generation and persistence of donor- and host-type tTregs and pTregs.


Asunto(s)
Trasplante de Médula Ósea , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Quimera por Trasplante/inmunología , Aloinjertos , Animales , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/terapia , Ratones , Ratones Endogámicos NOD
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...