Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 53(3): 581-596.e5, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32707034

RESUMEN

Glucocorticoids (GC) are the mainstay treatment option for inflammatory conditions. Despite the broad usage of GC, the mechanisms by which GC exerts its effects remain elusive. Here, utilizing murine autoimmune and allergic inflammation models, we report that Foxp3+ regulatory T (Treg) cells are irreplaceable GC target cells in vivo. Dexamethasone (Dex) administered in the absence of Treg cells completely lost its ability to control inflammation, and the lack of glucocorticoid receptor in Treg cells alone resulted in the loss of therapeutic ability of Dex. Mechanistically, Dex induced miR-342-3p specifically in Treg cells and miR-342-3p directly targeted the mTORC2 component, Rictor. Altering miRNA-342-3p or Rictor expression in Treg cells dysregulated metabolic programming in Treg cells, controlling their regulatory functions in vivo. Our results uncover a previously unknown contribution of Treg cells during glucocorticoid-mediated treatment of inflammation and the underlying mechanisms operated via the Dex-miR-342-Rictor axis.


Asunto(s)
Dexametasona/farmacología , Glucocorticoides/farmacología , Inflamación/tratamiento farmacológico , MicroARNs/genética , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Antiinflamatorios/farmacología , Factores de Transcripción Forkhead/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/biosíntesis , Receptores de Glucocorticoides/genética , Linfocitos T Reguladores/metabolismo
2.
J Immunol ; 212(12): 1867-1876, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38647384

RESUMEN

Allergic airway inflammation results from uncontrolled immune responses to environmental Ags. Although it is well established that allergic immune responses exhibit a high degree of diversity, driven by primary effector cell types such as eosinophils, neutrophils, or CD4 T cells with distinct effector signatures, the mechanisms responsible for such pathogenesis remain elusive. Foxp3+ regulatory T cells (Tregs) are essential immune regulators during chronic inflammation, including allergic airway inflammation. Emerging evidence suggests that Tregs infiltrating inflamed tissues exhibit distinct phenotypes dependent on the specific tissue sites and can display heterogeneity and tissue residency. Whether diverse allergic airway inflammatory responses influence infiltrating Treg heterogeneity or Treg lung residency has not been explored. We employed an unbiased single-cell RNA sequencing approach to investigate lung-infiltrating Tregs in models of eosinophilic and neutrophilic airway inflammation. We found that lung-infiltrating Tregs are highly heterogeneous, and that Tregs displaying lung-resident phenotypes are significantly different depending on the types of inflammation. Treg expression of ST2, a receptor for alarmin IL-33, was predominantly associated with eosinophilic inflammation and tissue residency. Nevertheless, Treg-specific ST2 deficiency did not affect the development of eosinophilic allergic inflammation or the generation of lung-resident Tregs. These results uncover a stark heterogeneity among Tregs infiltrating the lungs during allergic airway inflammation. The results indicate that varying types of inflammation may give rise to phenotypically distinct lung-resident Tregs, underscoring a (to our knowledge) novel mechanism by which inflammatory cues may shape the composition of infiltrating Tregs, allowing them to regulate inflammatory responses through tissue-adapted mechanisms.


Asunto(s)
Eosinófilos , Pulmón , Neutrófilos , Análisis de la Célula Individual , Linfocitos T Reguladores , Linfocitos T Reguladores/inmunología , Animales , Ratones , Neutrófilos/inmunología , Eosinófilos/inmunología , Pulmón/inmunología , Pulmón/patología , Ratones Endogámicos C57BL , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Ratones Noqueados , Inflamación/inmunología , Modelos Animales de Enfermedad , Interleucina-33/inmunología , Eosinofilia/inmunología , Eosinofilia/patología
3.
Am J Transplant ; 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39278625

RESUMEN

Antibody-mediated rejection (AMR) is among the leading causes of graft failure in solid organ transplantation. However, AMR treatment options are limited by incomplete understanding the mechanisms underlying de novo donor specific antibody (DSA) generation. The development of pathogenic isotype-switched DSA in response to transplanted allografts is typically attributed to follicular B cells undergoing germinal center reaction whereas the contribution of other B cell subsets have not been previously addressed. The current study investigated the role of recipient marginal zone (MZ) B cells in DSA responses using a mouse models of heart and renal allotransplantation. MZ B cells rapidly differentiate into antibody-secreting cells in response to allotransplantation. Despite selective depletion of FO B cells in heart allograft recipients, MZ B cells are sufficient for T-dependent IgM and early IgG DSA production. Furthermore, the presence of intact MZ B cell subset is required to support generation of pathogenic isotype-switched DSA in renal allograft recipients containing donor-reactive memory helper T cells. These findings are the first demonstration for the role of MZ B cells in humoral alloimmune responses following solid organ transplantation and identify MZ B cells as a potential therapeutic target for minimizing de novo DSA production and AMR in transplant recipients.

4.
Am J Transplant ; 24(7): 1146-1160, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38219867

RESUMEN

Cardiac allograft vasculopathy (CAV) causes late graft failure and mortality after heart transplantation. Donor-specific antibodies (DSAs) lead to chronic endothelial cell injury, inflammation, and arterial intimal thickening. In this study, GeoMx digital spatial profiling was used to analyze arterial areas of interest (AOIs) from CAV+DSA+ rejected cardiac allografts (N = 3; 22 AOIs total). AOIs were categorized based on CAV neointimal thickening and underwent whole transcriptome and protein profiling. By comparing our transcriptomic data with that of healthy control vessels of rapid autopsy myocardial tissue, we pinpointed specific pathways and transcripts indicative of heightened inflammatory profiles in CAV lesions. Moreover, we identified protein and transcriptomic signatures distinguishing CAV lesions exhibiting low and high neointimal lesions. AOIs with low neointima showed increased markers for activated inflammatory infiltrates, endothelial cell activation transcripts, and gene modules involved in metalloproteinase activation and TP53 regulation of caspases. Inflammatory and apoptotic proteins correlated with inflammatory modules in low neointima AOIs. High neointima AOIs exhibited elevated TGFß-regulated transcripts and modules enriched for platelet activation/aggregation. Proteins associated with growth factors/survival correlated with modules enriched for proliferation/repair in high neointima AOIs. Our findings reveal novel insight into immunological mechanisms mediating CAV pathogenesis.


Asunto(s)
Rechazo de Injerto , Trasplante de Corazón , Trasplante de Corazón/efectos adversos , Rechazo de Injerto/etiología , Rechazo de Injerto/patología , Rechazo de Injerto/inmunología , Humanos , Masculino , Aloinjertos , Isoanticuerpos/inmunología , Persona de Mediana Edad , Femenino , Transcriptoma , Neointima/patología , Neointima/inmunología , Neointima/etiología , Supervivencia de Injerto/inmunología , Pronóstico , Estudios de Seguimiento , Perfilación de la Expresión Génica , Biomarcadores/metabolismo , Donantes de Tejidos , Enfermedades Vasculares/etiología , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/patología , Multiómica
5.
Am J Transplant ; 24(3): 406-418, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38379280

RESUMEN

HLA donor-specific antibodies (DSA) elicit alloimmune responses against the graft vasculature, leading to endothelial cell (EC) activation and monocyte infiltration during antibody-mediated rejection (AMR). AMR promotes chronic inflammation and remodeling, leading to thickening of the arterial intima termed transplant vasculopathy or cardiac allograft vasculopathy (CAV) in heart transplants. Intragraft-recipient macrophages serve as a diagnostic marker in AMR; however, their polarization and function remain unclear. In this study, we utilized an in vitro Transwell coculture system to explore the mechanisms of monocyte-to-macrophage polarization induced by HLA I DSA-activated ECs. Anti-HLA I (IgG or F(ab')2) antibody-activated ECs induced the polarization of M2 macrophages with increased CD206 expression and MMP9 secretion. However, inhibition of TLR4 signaling or PSGL-1-P-selectin interactions significantly decreased both CD206 and MMP9. Monocyte adherence to Fc-P-selectin coated plates induced M2 macrophages with increased CD206 and MMP9. Moreover, Fc-receptor and IgG interactions synergistically enhanced active-MMP9 in conjunction with P-selectin. Transcriptomic analysis of arteries from DSA+CAV+ rejected cardiac allografts and multiplex-immunofluorescent staining illustrated the expression of CD68+CD206+CD163+MMP9+ M2 macrophages within the neointima of CAV-affected lesions. These findings reveal a novel mechanism linking HLA I antibody-activated endothelium to the generation of M2 macrophages which secrete vascular remodeling proteins contributing to AMR and CAV pathogenesis.


Asunto(s)
Receptor Toll-Like 4 , Enfermedades Vasculares , Humanos , Metaloproteinasa 9 de la Matriz , Selectina-P , Macrófagos , Endotelio , Antígenos HLA , Aloinjertos , Inmunoglobulina G
6.
Am J Transplant ; 23(9): 1307-1318, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37084848

RESUMEN

Antibodies reactive to self-antigens are an important component of posttransplant immune responses. The generation requirements and functions of autoantibodies, as well as the mechanisms of their influence on alloimmune responses, still remain to be determined. Our study investigated the contribution of autoimmunity during rejection of renal allografts. We have previously characterized a mouse model in which the acute rejection of a life-supporting kidney allograft is mediated by antibodies. At rejection, recipient sera screening against >4000 potential autoantigens revealed DNA topoisomerase I peptide 205-219 (TI-I205-219) as the most prominent epitope. Subsequent analysis showed TI-I205-219-reactive autoantibodies are induced in nonsensitized recipients of major histocompatibility complex-mismatched kidney allografts in a T cell-dependent manner. Immunization with TI-I205-219 broke self-tolerance, elicited TI-I205-219 immunoglobin G autoantibodies, and resulted in acute rejection of allogeneic but not syngeneic renal transplants. The graft loss was associated with increased priming of donor-reactive T cells but not with donor-specific alloantibodies elevation. Similarly, passive transfer of anti-TI-I205-219 sera following transplantation increased donor-reactive T cell activation with minimal effects on donor-specific alloantibody levels. The results identify DNA topoisomerase I as a novel self-antigen in transplant settings and demonstrate that autoantibodies enhance activation of donor-reactive T cells following renal transplantation.


Asunto(s)
Trasplante de Riñón , Linfocitos T , Ratones , Animales , Trasplante de Riñón/efectos adversos , ADN-Topoisomerasas de Tipo I , Autoanticuerpos , Rechazo de Injerto , Aloinjertos , Riñón
7.
Am J Pathol ; 192(7): 1053-1065, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35490714

RESUMEN

Cardiac allograft vasculopathy (CAV) limits the long-term success of heart transplants. Generation of donor-specific antibodies (DSAs) is associated with increased incidence of CAV clinically, but mechanisms underlying development of this pathology remain poorly understood. Major histocompatibility complex-mismatched A/J cardiac allografts in B6.CCR5-/- recipients have been reported to undergo acute rejection with little T-cell infiltration, but intense deposition of C4d in large vessels and capillaries of the graft accompanied by high titers of DSA. This model was modified to investigate mechanisms of antibody-mediated CAV by transplanting A/J hearts to B6.CCR5-/- CD8-/- mice that were treated with low doses of anti-CD4 monoclonal antibody to decrease T-cell-mediated graft injury and promote antibody-mediated injury. Although the mild inhibition of CD4 T cells extended allograft survival, the grafts developed CAV with intense C4d deposition and macrophage infiltration by 14 days after transplantation. Development of CAV correlated with recipient DSA titers. Transcriptomic analysis of microdissected allograft arteries identified the Notch ligand Dll4 as the most elevated transcript in CAV, associated with high versus low titers of DSA. More importantly, these analyses revealed a differential expression of transcripts in the CAV lesions compared with the matched apical tissue that lacks large arteries. In conclusion, these findings report a novel model of antibody-mediated CAV with the potential to facilitate further understanding of the molecular mechanisms promoting development of CAV.


Asunto(s)
Rechazo de Injerto , Trasplante de Corazón , Aloinjertos , Animales , Anticuerpos Monoclonales , Modelos Animales de Enfermedad , Trasplante de Corazón/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ápice del Diente
8.
J Immunol ; 206(11): 2521-2526, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34001658

RESUMEN

We investigated the function of the newly discovered myosin family protein myosin 18A (Myo18A) in Ab-mediated immunity by generating B cell-conditional Myo18A-deficient mice. Myo18A deficiency led to expansion of bone marrow progenitor B cells and mature B cells in secondary lymphoid organs. Myo18A-deficient mice displayed serum IgM hyperglobulinemia and increased splenic IgM-secreting cells, with older mice switching to IgG1 hyperglobulinemia and autoantibody development. Immunization of Myo18A-deficient mice with inactivated influenza virus led to development of more potent neutralizing Abs against the major Ag hemagglutinin, associated with persistent accumulation of Ag-specific germinal center B cells and more Ag-specific bone marrow plasma cells. In vitro stimulation with TLR7 and BCR ligands revealed a greater ability of Myo18A-deficient B cells to differentiate into Ab-secreting cells, associated with higher AID and Blimp-1 expression. Overall, our study demonstrates that Myo18A is a novel negative regulator of B cell homeostasis, differentiation, and humoral immunity.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Linfocitos B/inmunología , Inmunidad Humoral/inmunología , Miosinas/inmunología , Animales , Diferenciación Celular/inmunología , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Miosinas/deficiencia
9.
Environ Res ; 222: 115351, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36709030

RESUMEN

Wastewater surveillance has proven to be a useful tool for evidence-based epidemiology in the fight against the SARS-CoV-2 virus. It is particularly useful at the population level where acquisition of individual test samples may be time or cost-prohibitive. Wastewater surveillance for SARS-CoV-2 has typically been performed at wastewater treatment plants; however, this study was designed to sample on a local level to monitor the spread of the virus among three communities with distinct social vulnerability indices in Shreveport, Louisiana, located in a socially vulnerable region of the United States. Twice-monthly grab samples were collected from September 30, 2020, to March 23, 2021, during the Beta wave of the pandemic. The goals of the study were to examine whether: 1) concentrations of SARS-CoV-2 RNA in wastewater varied with social vulnerability indices and, 2) the time lag of spikes differed during wastewater monitoring in the distinct communities. The size of the population contributing to each sample was assessed via the quantification of the pepper mild mottle virus (PMMoV), which was significantly higher in the less socially vulnerable community. We found that the communities with higher social vulnerability exhibited greater viral loads as assessed by wastewater when normalized with PMMoV (Kruskal-Wallis, p < 0.05). The timing of the spread of the virus through the three communities appeared to be similar. These results suggest that interconnected communities within a municipality experienced the spread of the SARS-CoV-2 virus at similar times, but areas of high social vulnerability experienced more intense wastewater viral loads.


Asunto(s)
COVID-19 , Humanos , ARN Viral , SARS-CoV-2 , Carga Viral , Aguas Residuales , Monitoreo Epidemiológico Basado en Aguas Residuales
10.
FASEB J ; 35(11): e21990, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34665898

RESUMEN

Eukaryotic initiation factor 2A (eIF2A) is a 65 kDa protein that functions in minor initiation pathways, which affect the translation of only a subset of messenger ribonucleic acid (mRNAs), such as internal ribosome entry site (IRES)-containing mRNAs and/or mRNAs harboring upstream near cognate/non-AUG start codons. These non-canonical initiation events are important for regulation of protein synthesis during cellular development and/or the integrated stress response. Selective eIF2A knockdown in cellular systems significantly inhibits translation of such mRNAs, which rely on alternative initiation mechanisms for their translation. However, there exists a gap in our understanding of how eIF2A functions in mammalian systems in vivo (on the organismal level) and ex vivo (in cells). Here, using an eIF2A-knockout (KO) mouse model, we present evidence implicating eIF2A in the biology of aging, metabolic syndrome and central tolerance. We discovered that eIF2A-KO mice have reduced life span and that eIF2A plays an important role in maintenance of lipid homeostasis, the control of glucose tolerance, insulin resistance and also reduces the abundance of B lymphocytes and dendritic cells in the thymic medulla of mice. We also show the eIF2A KO affects male and female mice differently, suggesting that eIF2A may affect sex-specific pathways. Interestingly, our experiments involving pharmacological induction of endoplasmic reticulum (ER) stress with tunicamycin did not reveal any substantial difference between the response to ER stress in eIF2A-KO and wild-type mice. The identification of eIF2A function in the development of metabolic syndrome bears promise for the further identification of specific eIF2A targets responsible for these changes.


Asunto(s)
Metabolismo de los Lípidos , Longevidad , Síndrome Metabólico/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores Sexuales
11.
Am J Transplant ; 21(11): 3519-3523, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34058061

RESUMEN

In 1963, Lepow and colleagues resolved C1, the first component of the classical pathway, into three components, which they named C1q, C1r, and C1s. All three of these components were demonstrated to be involved in causing hemolysis in vitro. For over 30 years after that seminal discovery, the primary function attributed to C1q was as part of the C1 complex that initiated the classical pathway of the complement cascade. Then, a series of papers reported that isolated C1q could bind to apoptotic cells and facilitate their clearance by macrophages. Since then, rheumatologists have recognized that C1q is an important pattern recognition receptor (PRR) that diverts autoantigen containing extracellular vesicles from immune recognition. This critical function of C1q as a regulator of immune recognition has been largely overlooked in transplantation. Now that extracellular vesicles released from transplants have been identified as a major agent of immune recognition, it is logical to consider the potential impact of C1q on modulating the delivery of allogeneic extracellular vesicles to antigen presenting cells. This concept has clinical implications in the possible use of C1q or a derivative as a biological therapeutic to down-modulate immune responses to transplants.


Asunto(s)
Complemento C1r , Complemento C1s , Activación de Complemento , Complemento C1q
12.
FASEB J ; 34(5): 6888-6906, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32232901

RESUMEN

Previously, we identified a mechanism of inflammation control directed by ribosomal protein L13a and "GAIT" (Gamma Activated Inhibitor of Translation) elements in target mRNAs and showed that its elimination in myeloid cell-specific L13a knockout mice (L13a KO) increased atherosclerosis susceptibility and severity. Here, we investigated the mechanistic basis of this endogenous defense against atherosclerosis. We compared molecular and cellular aspects of atherosclerosis in high-fat diet (HFD)-fed L13a KO and intact (control) mice. HFD treatment of control mice induced release of L13a from 60S ribosome, formation of RNA-binding complex, and subsequent GAIT element-mediated translational silencing. Atherosclerotic plaques from HFD-treated KO mice showed increased infiltration of M1 type inflammatory macrophages. Macrophages from KO mice showed increased phagocytic activity and elevated expression of LDL receptor and pro-inflammatory mediators. NanoString analysis of the plaques from KO mice showed upregulation of a number of mRNAs encoding inflammatory proteins. Bioinformatics analysis suggests the presence of the potential GAIT elements in the 3'UTRs of several of these mRNAs. Macrophage induces L13a/GAIT-dependent translational silencing of inflammatory genes in response to HFD as an endogenous defense against atherosclerosis in ApoE-/- model.


Asunto(s)
Aterosclerosis/prevención & control , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Proteínas Ribosómicas/deficiencia , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Diferenciación Celular , Colesterol/metabolismo , Dieta Alta en Grasa/efectos adversos , Femenino , Macrófagos/clasificación , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Ratones Noqueados para ApoE , Células Mieloides/metabolismo , Células Mieloides/patología , Fagocitosis , Placa Aterosclerótica/etiología , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de LDL/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo
13.
J Immunol ; 202(4): 1045-1056, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30617225

RESUMEN

Contact hypersensitivity (CHS) is a CD8 T cell-mediated response to hapten skin sensitization and challenge. Sensitization of wild-type (WT) mice induces hapten-reactive effector CD8 T cells producing IFN-γ and IL-17- and IL-4-producing CD4 T cells that cannot mediate CHS. Although CXCR2-dependent Ly6G+ (neutrophil) cell recruitment into hapten-challenged skin is required to direct effector CD8 T cell infiltration into the challenge site to elicit CHS, 2,4-dinitrofluorobenezene (DNFB) sensitization of CXCR2-/- mice and neutrophil-depleted WT mice induced both hapten-reactive CD4 and CD8 T cells producing IFN-γ and IL-17. CD4 T cell-mediated CHS responses were not generated during DNFB sensitization of neutrophil-depleted WT mice treated with anti-IL-12 mAb or neutrophil-depleted IL-12-/- mice. Neutrophil depletion during DNFB sensitization of WT mice markedly increased IL-12-producing hapten-primed dendritic cell numbers in the skin-draining lymph nodes. Sensitization of mice lacking the neutrophil serine protease cathepsin G (CG)-induced hapten-reactive CD4 and CD8 T cells producing IFN-γ and IL-17 with elevated and elongated CHS responses to DNFB challenge. Induction of CHS effector CD4 T cells producing IFN-γ in neutrophil-depleted WT mice was eliminated by s.c. injection of active, but not inactivated, CG during sensitization. Thus, hapten skin sensitization induces neutrophil release of CG that systemically inhibits hapten-presenting dendritic cell production of IL-12 and the development of hapten-reactive CD4 T cells to IFN-γ-producing CHS effector cells.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Catepsina G/metabolismo , Células Dendríticas/metabolismo , Haptenos/metabolismo , Interleucina-12/biosíntesis , Neutrófilos/enzimología , Piel/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Dermatitis por Contacto/inmunología , Dermatitis por Contacto/metabolismo , Femenino , Haptenos/inmunología , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Receptores de Interleucina-8B/deficiencia , Receptores de Interleucina-8B/inmunología , Receptores de Interleucina-8B/metabolismo , Piel/inmunología
14.
J Cutan Pathol ; 48(9): 1166-1172, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33931901

RESUMEN

Retiform purpura has been described as a relatively frequent cutaneous finding in patients with coronavirus disease 2019 (COVID-19). The etiology is hypothesized to be related to thrombotic vasculopathy based on lesional biopsy specimen findings, but the pathogenesis of the vasculopathy is not completely understood. Here, we present a case of a retiform purpuric patch on the sacrum/buttocks in a hospitalized patient prior to subsequent diagnosis of COVID-19 and an eventual fatal disease course. Two lesional biopsy specimens at different time points in the disease course revealed thrombotic vasculopathy, despite therapeutic anticoagulation. Detailed histopathologic evaluation using immunohistochemical markers suggest the etiology of the vasculopathy involves both persistent complement activation and platelet aggregation, which possibly promote ongoing thrombus formation. This case highlights that sacral/buttock retiform purpuric patches may be a presenting sign of infection with SARS-CoV-2 virus and may represent an ominous sign supporting a future severe disease course. In addition, biopsy specimen findings at separate time points demonstrate that cutaneous vasculopathy may persist despite adequate systemic anticoagulation, possibly due to the combination of persistent complement and platelet activation. Finally, occlusive thrombi in sacral/buttock retiform purpuric patches may contribute to future ulceration and significant cutaneous morbidity in patients who survive COVID-19.


Asunto(s)
Nalgas/patología , COVID-19/complicaciones , COVID-19/patología , Púrpura/diagnóstico , Sacro/patología , Anciano , Anticoagulantes/uso terapéutico , Biopsia/métodos , Nalgas/virología , COVID-19/diagnóstico , COVID-19/inmunología , Calcifilaxia/diagnóstico , Activación de Complemento/inmunología , Diagnóstico Diferencial , Progresión de la Enfermedad , Resultado Fatal , Femenino , Humanos , Pacientes Internos , Agregación Plaquetaria/inmunología , Púrpura/virología , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Sacro/virología , Piel/patología , Enfermedades Cutáneas Vasculares/etiología , Enfermedades Cutáneas Vasculares/patología
15.
Kidney Int ; 98(4): 897-905, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32763116

RESUMEN

Allogeneic transplants elicit dynamic T cell responses that are modulated by positive and negative co-stimulatory receptors. Understanding mechanisms that intrinsically modulate the immune responses to transplants is vital to develop rational treatment for rejection. Here, we have investigated the impact of programed cell death-1 (PD-1) protein, a negative co-stimulatory receptor, on the rejection of MHC incompatible kidney transplants in mice. T cells were found to rapidly infiltrate the kidneys of A/J mice transplanted to C57BL/6 mice, which peaked at six days and decline by day 14. The T cells primarily encircled tubules with limited infiltration of the tubular epithelium. Lipocalin 2 (LCN2), a marker of tubular injury, also peaked in the urine at day six and then declined. Notably, flow cytometry demonstrated that most of the T cells expressed PD-1 (over 90% of CD8 and about 75% of CD4 cells) at day six. Administration of blocking antibody to PD-L1, the ligand for PD-1, before day six increased T cell infiltrates and urinary LCN2, causing terminal acute rejection. In contrast, blocking PD-1/PD-L1 interactions after day six caused only a transient increase in urinary LCN2. Depleting CD4 and CD8 T cells virtually eliminated LCN2 in the urine in support of T cells injuring tubules. Thus, our data indicate that PD-1/PD-L1 interactions are not just related to chronic antigenic stimulation of T cells but are critical for the regulation of acute T cell responses to renal transplants.


Asunto(s)
Trasplante de Riñón , Receptor de Muerte Celular Programada 1 , Animales , Antígeno B7-H1 , Linfocitos T CD8-positivos , Muerte Celular , Ligandos , Ratones , Ratones Endogámicos C57BL
16.
Am J Transplant ; 20(10): 2686-2702, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32320528

RESUMEN

HLA donor-specific antibodies (DSAs) binding to vascular endothelial cells of the allograft trigger inflammation, vessel injury, and antibody-mediated rejection (AMR). Accumulation of intragraft-recipient macrophages is a histological characteristic of AMR, which portends worse outcome. HLA class I (HLA I) DSAs enhance monocyte recruitment by activating endothelial cells and engaging FcγRs, but the DSA-activated donor endothelial influence on macrophage differentiation is unknown. In this study, we explored the consequence of DSA-activated endothelium on infiltrating monocyte differentiation. Here we show that cardiac allografts from murine recipients treated with MHC I DSA upregulated genes related to monocyte transmigration and Fc receptor stimulation. Human monocytes co-cultured with HLA I IgG-stimulated primary human endothelium promoted monocyte differentiation into CD68+ CD206+ CD163+ macrophages (M(HLA I IgG)), whereas HLA I F(ab')2 stimulated endothelium solely induced higher CD206 (M(HLA I F(ab')2 )). Both macrophage subtypes exhibited significant changes in discrete cytokines/chemokines and unique gene expression profiles. Cross-comparison of gene transcripts between murine DSA-treated cardiac allografts and human co-cultured macrophages identified overlapping genes. These findings uncover the role of HLA I DSA-activated endothelium in monocyte differentiation, and point to a novel, remodeling phenotype of infiltrating macrophages that may contribute to vascular injury.


Asunto(s)
Células Endoteliales , Rechazo de Injerto , Aloinjertos , Animales , Rechazo de Injerto/etiología , Antígenos HLA , Humanos , Inflamación/etiología , Isoanticuerpos , Macrófagos , Ratones , Fenotipo , Donantes de Tejidos
17.
Proc Natl Acad Sci U S A ; 114(38): 10190-10195, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28874534

RESUMEN

Dysregulated Foxp3+ Treg functions result in uncontrolled immune activation and autoimmunity. Therefore, identifying cellular factors modulating Treg functions is an area of great importance. Here, using Treg-specific Il27ra-/- mice, we report that IL-27 signaling in Foxp3+ Tregs is essential for Tregs to control autoimmune inflammation in the central nervous system (CNS). Following experimental autoimmune encephalomyelitis (EAE) induction, Treg-specific Il27ra-/- mice develop more severe EAE. Consistent with the severe disease, the numbers of IFNγ- and IL-17-producing CD4 T cells infiltrating the CNS tissues are greater in these mice. Treg accumulation in the inflamed CNS tissues is not affected by the lack of IL-27 signaling in Tregs, suggesting a functional defect of Il27ra-/- Tregs. IL-10 production by conventional CD4 T cells and their CNS accumulation are rather elevated in Treg-specific Il27ra-/- mice. Analysis with Treg fate-mapping reporter mice further demonstrates that IL-27 signaling in Tregs may control stability of Foxp3 expression. Finally, systemic administration of recombinant IL-27 in Treg-specific Il27ra-/- mice fails to ameliorate the disease even in the presence of IL-27-responsive conventional CD4 T cells. These findings uncover a previously unknown role of IL-27 in regulating Treg function to control autoimmune inflammation.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Encefalomielitis/inmunología , Receptores de Citocinas/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Sistema Nervioso Central/inmunología , Evaluación Preclínica de Medicamentos , Encefalomielitis/tratamiento farmacológico , Factores de Transcripción Forkhead/metabolismo , Interleucinas/metabolismo , Interleucinas/uso terapéutico , Ratones Transgénicos , Receptores de Citocinas/genética , Receptores de Interleucina
18.
J Am Soc Nephrol ; 30(12): 2413-2425, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597715

RESUMEN

BACKGROUND: The mechanisms underlying the effects of prolonged cold-ischemia storage on kidney allografts are poorly understood. METHODS: To investigate effects of cold ischemia on donor-reactive immune responses and graft pathology, we used a mouse kidney transplantation model that subjected MHC-mismatched BALB/c kidney allografts to cold-ischemia storage for 0.5 or 6 hours before transplant into C57BL/6 mice. RESULTS: At day 14 post-transplant, recipients of allografts subjected to 6 versus 0.5 hours of cold-ischemia storage had increased levels of anti-MHC class II (but not class I) donor-specific antibodies, increased donor-reactive T cells, and a significantly higher proportion of transplant glomeruli infiltrated with macrophages. By day 60 post-transplant, allografts with a 6 hour cold-ischemia time developed extensive glomerular injury compared with moderate pathology in allografts with 0.5 hour of cold-ischemia time. Pathology was associated with increased serum levels of anti-class 2 but not anti-class 1 donor-specific antibodies. Recipient B cell depletion abrogated early macrophage recruitment, suggesting augmented donor-specific antibodies, rather than T cells, increase glomerular pathology after prolonged cold ischemia. Lymphocyte sequestration with sphingosine-1-phosphate receptor 1 antagonist FTY720 specifically inhibited anti-MHC class II antibody production and abrogated macrophage infiltration into glomeruli. Adoptive transfer of sera containing anti-donor MHC class II antibodies or mAbs against donor MHC class II restored early glomerular macrophage infiltration in FTY720-treated recipients. CONCLUSIONS: Post-transplant inflammation augments generation of donor-specific antibodies against MHC class II antigens. Resulting MHC class II-reactive donor-specific antibodies are essential mediators of kidney allograft glomerular injury caused by prolonged cold ischemia.


Asunto(s)
Isquemia Fría/efectos adversos , Antígenos de Histocompatibilidad Clase II/inmunología , Isoanticuerpos/inmunología , Glomérulos Renales/patología , Trasplante de Riñón , Animales , Anticuerpos Monoclonales/inmunología , Clorhidrato de Fingolimod/uso terapéutico , Histocompatibilidad , Inmunidad Celular , Inmunidad Humoral , Inmunoglobulina G/inmunología , Inmunoglobulina G/uso terapéutico , Isoanticuerpos/biosíntesis , Glomérulos Renales/inmunología , Depleción Linfocítica , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especificidad del Receptor de Antígeno de Linfocitos T , Trasplante Homólogo
19.
Kidney Int ; 95(2): 350-362, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30503624

RESUMEN

Antibody mediated rejection (ABMR) is a major barrier to long-term kidney graft survival. Dysregulated donor-specific antibody (DSA) responses are induced in CCR5-deficient mice transplanted with complete major histocompatibility complex (MHC)-mismatched kidney allografts, and natural killer (NK) cells play a critical role in graft injury and rejection. We investigated the consequence of high DSA titers on kidney graft outcomes in the presence or absence of NK cell activation within the graft. Equivalent serum DSA titers were induced in CCR5-deficient B6 recipients of complete MHC mismatched A/J allografts and semi-allogeneic (A/J x B6) F1 kidney grafts, peaking by day 14 post-transplant. A/J allografts were rejected between days 16-28, whereas B6 isografts and semi-allogeneic grafts survived past day 65. On day 7 post-transplant, NK cell infiltration into A/J allografts was composed of distinct populations expressing high and low levels of the surface antigen NK1.1, with NK1.1low cells reflecting the highest level of activation. These NK cell populations increased with time post-transplant. In contrast, NK cell infiltration into semi-allogeneic grafts on day 7 was composed entirely of NK1.1high cells that decreased thereafter. On day 65 post-transplant the semi-allogeneic grafts had severe interstitial fibrosis, glomerulopathy, and arteriopathy, accompanied by expression of pro-fibrogenic genes. These results suggest that NK cells synergize with DSA to cause acute kidney allograft rejection, whereas high DSA titers in the absence of NK cell activation cannot provoke acute ABMR but instead induce the indolent development of interstitial fibrosis and glomerular injury that leads to late graft failure.


Asunto(s)
Aloinjertos/patología , Rechazo de Injerto/inmunología , Isoanticuerpos/inmunología , Trasplante de Riñón/efectos adversos , Riñón/patología , Enfermedad Aguda , Aloinjertos/citología , Aloinjertos/inmunología , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Rechazo de Injerto/patología , Supervivencia de Injerto/inmunología , Humanos , Riñón/citología , Riñón/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR5/genética , Receptores CCR5/inmunología , Trasplante Homólogo
20.
Am J Transplant ; 19(7): 1881-1887, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30903736

RESUMEN

IgG and albumin are the most abundant proteins in the circulation and have the longest half-lives. These properties are due to a unique receptor, the neonatal Fc receptor (FcRn). Although FcRn is named for its function of transferring IgG across the placenta from maternal to fetal circulation, FcRn functions throughout life to maintain IgG and albumin concentrations. FcRn protects IgG and albumin from intracellular degradation and recycles them back into the circulation. Clinical trials have confirmed that pathogenic antibodies can be depleted by blocking this homeostatic function of FcRn. Moreover, understanding the molecular interactions between IgG and FcRn has resulted in the design of therapeutic monoclonal antibodies with more efficacious pharmacokinetics. As a result of genetic engineering these monoclonals can be delivered at lower doses and at longer intervals. More recent findings have demonstrated that FcRn enhances phagocytosis by neutrophils, immune complex clearance by podocytes and antigen presentation by dendritic cells, macrophages, and B cells. This minireview highlights the relevance of FcRn to transplantation.


Asunto(s)
Productos Biológicos/farmacología , Antígenos de Histocompatibilidad Clase I/metabolismo , Homeostasis , Inmunoglobulina G/metabolismo , Receptores Fc/metabolismo , Presentación de Antígeno , Antígenos de Histocompatibilidad Clase I/efectos de los fármacos , Humanos , Fagocitosis , Receptores Fc/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA