Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
FASEB J ; 33(4): 5220-5236, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30620626

RESUMEN

Human leukocyte antigen G (HLA-G), a nonclassic HLA class Ib molecule involved in the maintenance of maternal tolerance to semiallogeneic fetal tissues during pregnancy, has emerged as a potential therapeutic target to control allograft rejection. We demonstrate here that the level of soluble HLA-G dimer was higher in a group of 90 patients with a functioning renal allograft compared with 40 patients who rejected (RJ) their transplants. The HLA-G dimer level was not affected by demographic status. One of the potential mechanisms in tissue-organ allograft rejection involves the induction of granzymes and perforin, which are the main effector molecules expressed by CD8+ cytotoxic T lymphocytes and function to destroy allogeneic transplants. Using genomics and molecular and cellular analyses of cells from T-cell-mediated RJ and nonrejected kidney transplant patients, cells from leukocyte Ig-like receptor B1 (LILRB1) transgenic mice, humanized mice, and genetically engineered HLA-G dimer, we demonstrated a novel mechanism by which HLA-G dimer inhibits activation and cytotoxic capabilities of human CD8+ T cells. This mechanism implicated the down-regulation of Granzyme B expression and the essential involvement of LILRB1. Thus, HLA-G dimer has the potential to be a specific and effective therapy for prevention of allograft rejection and prolongation of graft survival.-Ajith, A., Portik-Dobos, V., Nguyen-Lefebvre, A. T., Callaway, C., Horuzsko, D. D., Kapoor, R., Zayas, C., Maenaka, K., Mulloy, L. L., Horuzsko, A. HLA-G dimer targets Granzyme B pathway to prolong human renal allograft survival.


Asunto(s)
Granzimas/metabolismo , Antígenos HLA-G/metabolismo , Adulto , Animales , Antígenos CD/metabolismo , Linfocitos T CD8-positivos/metabolismo , Concanavalina A/farmacología , Femenino , Citometría de Flujo , Rechazo de Injerto , Supervivencia de Injerto , Humanos , Trasplante de Riñón , Receptor Leucocitario Tipo Inmunoglobulina B1/antagonistas & inhibidores , Receptor Leucocitario Tipo Inmunoglobulina B1/metabolismo , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/metabolismo
2.
Artículo en Inglés | MEDLINE | ID: mdl-37855709

RESUMEN

This study investigates the effects of peroxisome proliferator-activated receptor gamma (PPARγ) inhibition on bone and immune cell profiles in aged female mice, as well as in vitro stromal stem cell osteogenic differentiation and inflammation gene expression. The hypothesis was that inhibition of PPARγ would increase bone mass and alter immune and other cellular functions. Our results showed that treatment with PPARγ antagonist GW9662 for 6 weeks reduced bone volume and trabecular number and increased trabecular spacing. However, inhibition of PPARγ had no significant effect on marrow and spleen immune cell composition in aged female mice. In vitro experiments indicated that GW9662 treatment increased the expression of osteogenic genes but did not affect adipogenic genes. Additionally, GW9662 treatment decreased the expression of several inflammation-related genes. Overall, these findings suggest that PPARγ inhibition may have adverse effects on bone in aged female mice.


Asunto(s)
Anilidas , Osteogénesis , PPAR gamma , Animales , Femenino , Ratones , Adipogénesis , Anilidas/administración & dosificación , Inflamación , Osteogénesis/efectos de los fármacos , PPAR gamma/antagonistas & inhibidores , Huesos/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Envejecimiento/patología
3.
Hum Immunol ; 84(8): 366-373, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36934068

RESUMEN

Despite the growing interest in the role of regulatory B cells (Bregs) in autoimmunity, their distinct role and function in kidney transplant outcomes remain elusive. Here, we retrospectively analyzed the proportion of Bregs, transitional Bregs (tBregs) and memory Bregs (mBregs) and their capacity to produce IL-10 in non-rejected (NR) versus rejected (RJ) kidney transplant recipients. In the NR group, we observed a significant increase in the proportion of mBregs (CD19+CD24hiCD27+) but no difference in tBregs (CD19+CD24hiCD38+), as compared to the RJ group. We also observed a significant increase in IL-10-producing mBregs (CD19+CD24hiCD27+IL-10+) in the NR group. As our group and others have previously reported a potential role of the human leukocyte antigen G (HLA-G) in human renal allograft survival, notably through IL-10, we then investigated possible crosstalk between HLA-G and IL-10+ mBregs. Our ex vivo data suggest a role of HLA-G in enhancing IL-10+ mBreg expansion upon stimulation, which further decreased CD3+ T cell proliferation capability. Using RNA-sequencing (RNA-seq), we identified potential key signaling pathways involved in HLA-G-driven IL-10+ mBreg expansion, such as the MAPK, TNF and chemokine signaling pathways. Together, our study highlights a novel HLA-G-mediated IL-10-producing mBreg pathway that may serve as a therapeutic target to improve kidney allograft survival.


Asunto(s)
Linfocitos B Reguladores , Trasplante de Riñón , Humanos , Antígenos HLA-G/metabolismo , Interleucina-10/metabolismo , Estudios Retrospectivos , Riñón , Aloinjertos
4.
J Clin Invest ; 133(21)2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37651197

RESUMEN

The triggering receptor expressed on myeloid cell 1 (TREM1) plays a critical role in development of chronic inflammatory disorders and the inflamed tumor microenvironment (TME) associated with most solid tumors. We examined whether loss of TREM1 signaling can abrogate the immunosuppressive TME and enhance cancer immunity. To investigate the therapeutic potential of TREM1 in cancer, we used mice deficient in Trem1 and developed a novel small molecule TREM1 inhibitor, VJDT. We demonstrated that genetic or pharmacological TREM1 silencing significantly delayed tumor growth in murine melanoma (B16F10) and fibrosarcoma (MCA205) models. Single-cell RNA-Seq combined with functional assays during TREM1 deficiency revealed decreased immunosuppressive capacity of myeloid-derived suppressor cells (MDSCs) accompanied by expansion in cytotoxic CD8+ T cells and increased PD-1 expression. Furthermore, TREM1 inhibition enhanced the antitumorigenic effect of anti-PD-1 treatment, in part, by limiting MDSC frequency and abrogating T cell exhaustion. In patient-derived melanoma xenograft tumors, treatment with VJDT downregulated key oncogenic signaling pathways involved in cell proliferation, migration, and survival. Our work highlights the role of TREM1 in cancer progression, both intrinsically expressed in cancer cells and extrinsically in the TME. Thus, targeting TREM1 to modify an immunosuppressive TME and improve efficacy of immune checkpoint therapy represents what we believe to be a promising therapeutic approach to cancer.


Asunto(s)
Melanoma , Células Supresoras de Origen Mieloide , Humanos , Animales , Ratones , Receptor Activador Expresado en Células Mieloides 1/genética , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Células Mieloides/patología , Línea Celular Tumoral , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Linfocitos T Citotóxicos/patología , Modelos Animales de Enfermedad , Microambiente Tumoral
5.
Front Immunol ; 12: 687715, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34177940

RESUMEN

The outcome of organ transplantation is largely dictated by selection of a well-matched donor, which results in less chance of graft rejection. An allogeneic immune response is the main immunological barrier for successful organ transplantation. Donor and recipient human leukocyte antigen (HLA) mismatching diminishes outcomes after solid organ transplantation. The current evaluation of HLA incompatibility does not provide information on the immunogenicity of individual HLA mismatches and impact of non-HLA-related alloantigens, especially in vivo. Here we demonstrate a new method for analysis of alloimmune responsiveness between donor and recipient in vivo by introducing a humanized mouse model. Using molecular, cellular, and genomic analyses, we demonstrated that a recipient's personalized humanized mouse provided the most sensitive assessment of allogeneic responsiveness to potential donors. In our study, HLA typing provided a better recipient-donor match for one donor among two related donors. In contrast, assessment of an allogeneic response by mixed lymphocyte reaction (MLR) was indistinguishable between these donors. We determined that, in the recipient's humanized mouse model, the donor selected by HLA typing induced the strongest allogeneic response with markedly increased allograft rejection markers, including activated cytotoxic Granzyme B-expressing CD8+ T cells. Moreover, the same donor induced stronger upregulation of genes involved in the allograft rejection pathway as determined by transcriptome analysis of isolated human CD45+cells. Thus, the humanized mouse model determined the lowest degree of recipient-donor alloimmune response, allowing for better selection of donor and minimized immunological risk of allograft rejection in organ transplantation. In addition, this approach could be used to evaluate the level of alloresponse in allogeneic cell-based therapies that include cell products derived from pluripotent embryonic stem cells or adult stem cells, both undifferentiated and differentiated, all of which will produce allogeneic immune responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Rechazo de Injerto/prevención & control , Antígenos HLA/inmunología , Prueba de Histocompatibilidad , Histocompatibilidad , Leucocitos Mononucleares/trasplante , Trasplante de Órganos , Bazo/inmunología , Tolerancia al Trasplante , Animales , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Bases de Datos Genéticas , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Supervivencia de Injerto , Antígenos HLA/genética , Humanos , Isoanticuerpos/metabolismo , Leucocitos Mononucleares/inmunología , Prueba de Cultivo Mixto de Linfocitos , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Órganos/efectos adversos , Fenotipo , Valor Predictivo de las Pruebas , Bazo/metabolismo , Transcriptoma , Trasplante Homólogo
6.
Hum Immunol ; 81(4): 178-185, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32093884

RESUMEN

HLA-G is a nonclassical MHC-Class I molecule whose expression, along the feto-maternal barrier contributes towards tolerance of the semiallogeneic fetus during pregnancy. In light of its inhibitory properties, recent research has established HLA-G involvement in mechanisms responsible for directing allogeneic immune responses towards tolerance during allogeneic situations such as organ transplantation. Here, we critically review the data supporting the tolerogenic role of HLA-G in organ transplantation, the various factors influencing its expression, and the introduction of novel humanized mouse models that are one of the best approaches to assess the utility of HLA-G as a therapeutic tool in organ transplantation.


Asunto(s)
Antígenos HLA-G/genética , Antígenos HLA-G/inmunología , Modelos Animales , Trasplante de Órganos , Animales , Regulación de la Expresión Génica/inmunología , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Antígenos HLA-G/química , Antígenos HLA-G/metabolismo , Humanos , Tolerancia Inmunológica , Ratones , Ratones Transgénicos , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
7.
J Clin Invest ; 128(11): 4870-4883, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30137027

RESUMEN

Inflammation occurs in all tissues in response to injury or stress and is the key process underlying hepatic fibrogenesis. Targeting chronic and uncontrolled inflammation is one strategy to prevent liver injury and fibrosis progression. Here, we demonstrate that triggering receptor expressed on myeloid cells 1 (TREM-1), an amplifier of inflammation, promotes liver disease by intensifying hepatic inflammation and fibrosis. In the liver, TREM-1 expression was limited to liver macrophages and monocytes and was highly upregulated on Kupffer cells, circulating monocytes, and monocyte-derived macrophages in a mouse model of chronic liver injury and fibrosis induced by carbon tetrachloride (CCl4) administration. TREM-1 signaling promoted proinflammatory cytokine production and mobilization of inflammatory cells to the site of injury. Deletion of Trem1 reduced liver injury, inflammatory cell infiltration, and fibrogenesis. Reconstitution of Trem1-deficient mice with Trem1-sufficient Kupffer cells restored the recruitment of inflammatory monocytes and the severity of liver injury. Markedly increased infiltration of liver fibrotic areas with TREM-1-positive Kupffer cells and monocytes/macrophages was found in patients with hepatic fibrosis. Our data support a role of TREM-1 in liver injury and hepatic fibrogenesis and suggest that TREM-1 is a master regulator of Kupffer cell activation, which escalates chronic liver inflammatory responses, activates hepatic stellate cells, and reveals a mechanism of promotion of liver fibrosis.


Asunto(s)
Macrófagos del Hígado/inmunología , Cirrosis Hepática/inmunología , Hígado/inmunología , Hígado/lesiones , Receptor Activador Expresado en Células Mieloides 1/inmunología , Animales , Enfermedad Crónica , Citocinas/genética , Citocinas/inmunología , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Macrófagos del Hígado/patología , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Mutantes , Monocitos/inmunología , Monocitos/patología , Receptor Activador Expresado en Células Mieloides 1/genética
8.
Hum Immunol ; 77(9): 711-9, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27085792

RESUMEN

HLA-G was described originally as a tolerogenic molecule that allows the semiallogeneic fetus to escape from recognition by the maternal immune response. This review will discuss different steps in the study of HLA-G expression and functions in vivo, starting with analyses of expression of the HLA-G gene and its receptors in transgenic mice, and continuing with applications of HLA-G and its receptors in prevention of allograft rejection, transplantation tolerance, and controlling the development of infection. Humanized mouse models have been discussed for developing in vivo studies of HLA-G in physiological and pathological conditions. Collectively, animal models provide an opportunity to evaluate the importance of the interaction between HLA-G and its receptors in terms of its ability to regulate immune responses during maternal-fetal tolerance, survival of allografts, tumor-escape mechanisms, and development of infections when both HLA-G and its receptors are expressed. In addition, in vivo studies on HLA-G also offer novel approaches to achieve a reproducible transplantation tolerance and to develop personalized medicine to prevent allograft rejection.


Asunto(s)
Rechazo de Injerto/inmunología , Antígenos HLA-G/metabolismo , Inmunoterapia , Infecciones/inmunología , Ratones , Modelos Animales , Neoplasias/inmunología , Trasplante de Órganos , Embarazo/inmunología , Animales , Femenino , Rechazo de Injerto/prevención & control , Antígenos HLA-G/genética , Humanos , Tolerancia Inmunológica , Infecciones/terapia , Ratones SCID , Ratones Transgénicos , Neoplasias/terapia , Medicina de Precisión , Escape del Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA