Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 13: 801945, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36032128

RESUMEN

Lymph nodes (LNs) are the critical sites of immunity, and the stromal cells of LNs are crucial to their function. Our understanding of the stromal compartment of the LN has deepened recently with the characterization of nontraditional stromal cells. CD41 (integrin αIIb) is known to be expressed by platelets and hematolymphoid cells. We identified two distinct populations of CD41+Lyve1+ and CD41+Lyve1- cells in the LNs. CD41+Lyve1- cells appear in the LN mostly at the later stages of the lives of mice. We identified CD41+ cells in human LNs as well. We demonstrated that murine CD41+ cells express mesodermal markers, such as Sca-1, CD105 and CD29, but lack platelet markers. We did not observe the presence of platelets around the HEVs or within proximity to fibroblastic reticular cells of the LN. Examination of thoracic duct lymph fluid showed the presence of CD41+Lyve1- cells, suggesting that these cells recirculate throughout the body. FTY720 reduced their trafficking to lymph fluid, suggesting that their egress is controlled by the S1P1 pathway. CD41+Lyve1- cells of the LNs were sensitive to radiation, suggestive of their replicative nature. Single cell RNA sequencing data showed that the CD41+ cell population in naïve mouse LNs expressed largely stromal cell markers. Further studies are required to examine more deeply the role of CD41+ cells in the function of LNs.


Asunto(s)
Ganglios Linfáticos , Células del Estroma , Animales , Fibroblastos , Humanos , Ratones
2.
Nat Commun ; 13(1): 2810, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35589724

RESUMEN

Cellular therapies offer a promising therapeutic strategy for the highly malignant brain tumor, glioblastoma (GBM). However, their clinical translation is limited by the lack of effective target identification and stringent testing in pre-clinical models that replicate standard treatment in GBM patients. In this study, we show the detection of cell surface death receptor (DR) target on CD146-enriched circulating tumor cells (CTC) captured from the blood of mice bearing GBM and patients diagnosed with GBM. Next, we developed allogeneic "off-the-shelf" clinical-grade bifunctional mesenchymal stem cells (MSCBif) expressing DR-targeted ligand and a safety kill switch. We show that biodegradable hydrogel encapsulated MSCBif (EnMSCBif) has a profound therapeutic efficacy in mice bearing patient-derived invasive, primary and recurrent GBM tumors following surgical resection. Activation of the kill switch enhances the efficacy of MSCBif and results in their elimination post-tumor treatment which can be tracked by positron emission tomography (PET) imaging. This study establishes a foundation towards a clinical trial of EnMSCBif in primary and recurrent GBM patients.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Trasplante de Células Madre Hematopoyéticas , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Glioblastoma/terapia , Humanos , Ratones , Recurrencia Local de Neoplasia/terapia
3.
JCI Insight ; 6(13)2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34236047

RESUMEN

Heart transplantation is the optimal therapy for patients with end-stage heart disease, but its long-term outcome remains inadequate. Recent studies have highlighted the importance of the melanocortin receptors (MCRs) in inflammation, but how MCRs regulate the balance between alloreactive T cells and Tregs, and whether they impact chronic heart transplant rejection, is unknown. Here, we found that Tregs express MC2R, and MC2R expression was highest among all MCRs by Tregs. Our data indicate that adrenocorticotropic hormone (ACTH), the sole ligand for MC2R, promoted the formation of Tregs by increasing the expression of IL-2Rα (CD25) in CD4+ T cells and activation of STAT5 in CD4+CD25+ T cells. ACTH treatment also improved the survival of heart allografts and increased the formation of Tregs in CD28KO mice. ACTH treatment synergized with the tolerogenic effect of CTLA-4-Ig, resulting in long-term survival of heart allografts and an increase in intragraft Tregs. ACTH administration also demonstrated higher prolongation of heart allograft survival in transgenic mouse recipients with both complete KO and conditional KO of PI3Kγ in T cells. Finally, ACTH treatment reduced chronic rejection markedly. These data demonstrate that ACTH treatment improved heart transplant outcomes, and this effect correlated with an increase in Tregs.


Asunto(s)
Hormona Adrenocorticotrópica , Rechazo de Injerto , Trasplante de Corazón/efectos adversos , Proteínas de la Membrana/inmunología , Linfocitos T Reguladores/inmunología , Hormona Adrenocorticotrópica/inmunología , Hormona Adrenocorticotrópica/farmacología , Animales , Antígenos CD4/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Rechazo de Injerto/tratamiento farmacológico , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Hormonas/farmacología , Inflamación/inmunología , Inflamación/patología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Ratones , Factor de Transcripción STAT5/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Tolerancia al Trasplante/efectos de los fármacos , Tolerancia al Trasplante/inmunología
4.
Front Immunol ; 12: 730438, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35111151

RESUMEN

Lymph node (LN)-resident stromal cells play an essential role in the proper functioning of LNs. The stromal compartment of the LN undergoes significant compensatory changes to produce a milieu amenable for regulation of the immune response. We have identified a distinct population of leptin receptor-expressing (LepR+) stromal cells, located in the vicinity of the high endothelial venules (HEVs) and lymphatics. These LepR+ stromal cells expressed markers for fibroblastic reticular cells (FRCs), but they lacked markers for follicular dendritic cells (FDCs) and marginal reticular cells (MRCs). Leptin signaling deficiency led to heightened inflammatory responses within the LNs of db/db mice, leakiness of HEVs, and lymphatic fragmentation. Leptin signaling through the JAK/STAT pathway supported LN stromal cell survival and promoted the anti-inflammatory properties of these cells. Conditional knockout of the LepR+ stromal cells in LNs resulted in HEV and extracellular matrix (ECM) abnormalities. Treatment of ob/ob mice with an agonist leptin fusion protein restored the microarchitecture of LNs, reduced intra-LN inflammatory responses, and corrected metabolic abnormalities. Future studies are needed to study the importance of LN stomal cell dysfunction to the pathogenesis of inflammatory responses in type 2 diabetes (T2D) in humans.


Asunto(s)
Ganglios Linfáticos/metabolismo , Receptores de Leptina/metabolismo , Células del Estroma/metabolismo , Animales , Línea Celular , Células Dendríticas Foliculares/metabolismo , Endotelio/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Inmunidad/fisiología , Inflamación/metabolismo , Vasos Linfáticos/metabolismo , Ratones , Transducción de Señal/fisiología , Vénulas/metabolismo
5.
Cell ; 183(5): 1219-1233.e18, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33242418

RESUMEN

Cancer therapies kill tumors either directly or indirectly by evoking immune responses and have been combined with varying levels of success. Here, we describe a paradigm to control cancer growth that is based on both direct tumor killing and the triggering of protective immunity. Genetic ablation of serine protease inhibitor SerpinB9 (Sb9) results in the death of tumor cells in a granzyme B (GrB)-dependent manner. Sb9-deficient mice exhibited protective T cell-based host immunity to tumors in association with a decline in GrB-expressing immunosuppressive cells within the tumor microenvironment (TME). Maximal protection against tumor development was observed when the tumor and host were deficient in Sb9. The therapeutic utility of Sb9 inhibition was demonstrated by the control of tumor growth, resulting in increased survival times in mice. Our studies describe a molecular target that permits a combination of tumor ablation, interference within the TME, and immunotherapy in one potential modality.


Asunto(s)
Citotoxicidad Inmunológica , Inmunoterapia , Proteínas de la Membrana/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Serpinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Progresión de la Enfermedad , Femenino , Eliminación de Gen , Granzimas/metabolismo , Inmunidad/efectos de los fármacos , Melanoma/patología , Ratones Endogámicos C57BL , Neoplasias/prevención & control , Bibliotecas de Moléculas Pequeñas/farmacología , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Microambiente Tumoral/efectos de los fármacos
6.
Transplant Direct ; 6(8): e589, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32766437

RESUMEN

BACKGROUND: Organ transplantation is a standard therapeutic strategy for irreversible organ damage, but the utility of nerve transplantation remains generally unexplored, despite its potential benefit to a large patient population. Here, we aimed to establish a feasible preclinical mouse model for understanding the cellular mechanisms behind the rejection of peripheral and optic nerves. METHODS: We performed syngenic and allogenic transplantation of optic and sciatic nerves in mice by inserting the nerve grafts inside the kidney capsule, and we assessed the allografts for signs of rejection through 14 d following transplantation. Then, we assessed the efficacy of CTLA4 Ig, Rapamycin, and anti-CD3 antibody in suppressing immune cell infiltration of the nerve allografts. RESULTS: By 3 d posttransplantation, both sciatic and optic nerves transplanted from BALB/c mice into C57BL/6J recipients contained immune cell infiltrates, which included more CD11b+ macrophages than CD3+ T cells or B220+ B cells. Ex vivo immunogenicity assays demonstrated that sciatic nerves demonstrated higher alloreactivity in comparison with optic nerves. Interestingly, optic nerves contained higher populations of anti-inflammatory PD-L1+ cells than sciatic nerves. Treatment with anti-CD3 antibody reduced immune cell infiltrates in the optic nerve allograft, but exerted no significant effect in the sciatic nerve allograft. CONCLUSIONS: These findings establish the feasibility of a preclinical allogenic nerve transplantation model and provide the basis for future testing of directed, high-intensity immunosuppression in these mice.

7.
J Clin Invest ; 130(8): 4182-4194, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32597832

RESUMEN

Although the immune response within draining lymph nodes (DLNs) has been studied for decades, how their stromal compartment contributes to this process remains to be fully explored. Here, we show that donor mast cells were prominent activators of collagen I deposition by fibroblastic reticular cells (FRCs) in DLNs shortly following transplantation. Serial analysis of the DLN indicated that the LN stroma did not return to its baseline microarchitecture following organ rejection and that the DLN contained significant fibrosis following repetitive organ transplants. Using several FRC conditional-knockout mice, we show that induction of senescence in the FRCs of the DLN resulted in massive production of collagen I and a proinflammatory milieu within the DLN. Stimulation of herpes virus entry mediator (HVEM) on FRCs by its ligand LIGHT contributed chiefly to the induction of senescence in FRCs and overproduction of collagen I. Systemic administration of ex vivo-expanded FRCs to mice decreased DLN fibrosis and strengthened the effect of anti-CD40L in prolonging heart allograft survival. These data demonstrate that the transformation of FRCs into proinflammatory myofibroblasts is critically important for the maintenance of a proinflammatory milieu within a fibrotic DLN.


Asunto(s)
Fibroblastos/metabolismo , Trasplante de Corazón , Ganglios Linfáticos/metabolismo , Animales , Fibroblastos/patología , Fibrosis , Ganglios Linfáticos/patología , Ratones , Ratones Noqueados
8.
ACS Nano ; 13(11): 12393-12407, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31518498

RESUMEN

Inflammation occurring within the transplanted organ from the time of harvest is an important stimulus of early alloimmune reactivity and promotes chronic allograft rejection. Chronic immune-mediated injury remains the primary obstacle to the long-term success of organ transplantation. However, organ transplantation represents a rare clinical setting in which the organ is accessible ex vivo, providing an opportunity to use nanotechnology to deliver therapeutics directly to the graft. This approach facilitates the directed delivery of immunosuppressive agents (ISA) to target local pathogenic immune responses prior to the transplantation. Here, we have developed a system of direct delivery and sustained release of mycophenolate mofetil (MMF) to treat the donor organ prior to transplantation. Perfusion of a donor mouse heart with MMF-loaded PEG-PLGA nanoparticles (MMF-NPs) prior to transplantation abrogated cardiac transplant vasculopathy by suppressing intragraft pro-inflammatory cytokines and chemokines. Our findings demonstrate that ex vivo delivery of an ISA to donor organs using a nanocarrier can serve as a clinically feasible approach to reduce transplant immunity.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanomedicina/métodos , Trasplantes , Animales , Femenino , Rechazo de Injerto/fisiopatología , Rechazo de Injerto/prevención & control , Trasplante de Corazón , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/fisiopatología , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ácido Micofenólico/administración & dosificación , Ácido Micofenólico/farmacocinética , Ácido Micofenólico/farmacología , Nanopartículas/química , Cuidados Preoperatorios , Trasplantes/irrigación sanguínea , Trasplantes/metabolismo , Trasplantes/fisiopatología
9.
Sci Rep ; 9(1): 9778, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31278331

RESUMEN

The pancreatic lymph node is critical to the pathogenesis of autoimmune diabetes, as it constitutes the initial site for the priming of autoreactive T cells. In this study, we compared the histopathology of the head pancreatic lymph node (HPLN) to the tail pancreatic lymph node (TPLN) in NOD mice. HPLNs and TPLNs were harvested from 4 week-, 8 week-, and 12 week-old NOD mice, and their microvasculature, extracellular matrix, and immune cell subsets were characterized. The percentages of B cells and antigen-presenting cells (APCs) were much higher in the HPLN, as compared to the TPLN. Notably, the HPLNs of 12 week-old mice were characterized by greater expansion of high endothelial venules (HEVs) and lymphatic vessels in comparison to the TPLNs. Finally, we observed a higher density of extracellular matrix (ECM) fibers surrounding the lymphatic vasculature in the HPLNs than in the TPLNs. These data for the first time demonstrate that the HPLN possesses a different immune microanatomy and organization from the TPLN. These novel observations unveil a major phenotypic difference between two types of LNs from the same organ and may highlight an independent fundamental role played by each PLN during the establishment of T1D.


Asunto(s)
Inmunidad , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Páncreas/inmunología , Páncreas/metabolismo , Animales , Biomarcadores , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Inmunofenotipificación , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos NOD
10.
Sci Rep ; 9(1): 6535, 2019 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-31024011

RESUMEN

A primary goal in the management of burn wounds is early wound closure. The use of skin allografts represents a lifesaving strategy for severe burn patients, but their ultimate rejection limits their potential efficacy and utility. IL-6 is a major pleiotropic cytokine which critically links innate and adaptive immune responses. Here, we devised anti-IL-6 receptor eluting gelatin methacryloyl (GelMA) biomaterials (GelMA/anti-IL-6), which were implanted at the interface between the wound beds and skin allografts. Our visible light crosslinked GelMA/anti-IL-6 immunomodulatory biomaterial (IMB) demonstrated a stable kinetic release profile of anti-IL-6. In addition, the incorporation of anti-IL-6 within the GelMA hydrogel had no effect on the mechanical properties of the hydrogels. Using a highly stringent skin transplant model, the GelMA/anti-IL-6 IMB almost doubled the survival of skin allografts. The use of GelMA/anti-IL-6 IMB was far superior to systemic anti-IL-6 receptor treatment in prolonging skin allograft survival. As compared to the untreated control group, skin from the GelMA/anti-IL-6 IMB group contained significantly fewer alloreactive T cells and macrophages. Interestingly, the environmental milieu of the draining lymph nodes (DLNs) of the mice implanted with the GelMA/anti-IL-6 IMB was also considerably less pro-inflammatory. The percentage of CD4+ IFNγ+ cells was much lower in the DLNs of the GelMA/anti-IL-6 IMB group in comparison to the GelMA group. These data highlight the importance of localized immune delivery in prolonging skin allograft survival and its potential utility in treating patients with severe burns.


Asunto(s)
Aloinjertos/efectos de los fármacos , Materiales Biocompatibles/farmacología , Supervivencia de Injerto/efectos de los fármacos , Factores Inmunológicos/farmacología , Interleucina-6/inmunología , Trasplante de Piel , Animales , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Femenino , Fibrosis , Gelatina/química , Supervivencia de Injerto/inmunología , Inflamación/patología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Metacrilatos/química , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Porcinos , Linfocitos T/efectos de los fármacos , Adhesivos Tisulares/farmacología
11.
EBioMedicine ; 38: 79-88, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30497977

RESUMEN

BACKGROUND: Nanomedicine offers an excellent opportunity to tackle treatment-refractory malignancies by enhancing the delivery of therapeutics to the tumor site. High endothelial venules (HEVs) are found primarily in lymph nodes or formed de novo in peripheral tissues during inflammatory responses. They express peripheral node addressin (PNAd), which is recognized by the monoclonal antibody MECA79. METHODS: Here, we demonstrated that HEVs form de novo in human pancreatic ductal adenocarcinoma (PDAC). We engineered MECA79 coated nanoparticles (MECA79-NPs) that recognize these ectopic HEVs in PDAC. FINDINGS: The trafficking of MECA79-NPs following intravenous delivery to human PDAC implanted in a humanized mouse model was more robust than non-conjugated NPs. Treatment with MECA79-Taxol-NPs augmented the delivery of Paclitaxel (Taxol) to the tumor site and significantly reduced the tumor size. This effect was associated with a higher apoptosis rate of PDAC cells and reduced vascularization within the tumor. INTERPRETATION: Targeting the HEVs of PDAC using MECA79-NPs could lay the ground for the localized delivery of a wide variety of drugs including chemotherapeutic agents. FUND: National Institutes of Health (NIH) grants: T32-EB016652 (B·B.), NIH Cancer Core Grant CA034196 (L.D.S.), National Institute of Allergy and Infectious Diseases grants R01-AI126596 and R01-HL141815 (R.A.).


Asunto(s)
Carcinoma Ductal Pancreático/patología , Endotelio Linfático/patología , Ganglios Linfáticos/patología , Neovascularización Patológica , Neoplasias Pancreáticas/patología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Biomarcadores , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Endotelio Linfático/efectos de los fármacos , Endotelio Linfático/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Terapia Molecular Dirigida , Nanopartículas/química , Nanopartículas/ultraestructura , Neovascularización Patológica/tratamiento farmacológico , Paclitaxel/administración & dosificación , Paclitaxel/química , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Nanomedicina Teranóstica , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
12.
J Clin Invest ; 128(11): 4770-4786, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30277476

RESUMEN

The targeted delivery of therapeutic drugs to lymph nodes (LNs) provides an unprecedented opportunity to improve the outcomes of transplantation and immune-mediated diseases. The high endothelial venule is a specialized segment of LN vasculature that uniquely expresses peripheral node addressin (PNAd) molecules. PNAd is recognized by MECA79 mAb. We previously generated a MECA79 mAb-coated microparticle (MP) that carries tacrolimus. Although this MP trafficked to LNs, it demonstrated limited therapeutic efficacy in our transplant model. Here, we have synthesized a nanoparticle (NP) as a carrier of anti-CD3, and optimized the conjugation strategy to coat the NP surface with MECA79 mAb (MECA79-anti-CD3-NP) to enhance LN accumulation. As compared with nonconjugated NPs, a significantly higher quantity of MECA79-NPs accumulated in the draining lymph node (DLN). Many MECA79-NPs underwent internalization by T cells and dendritic cells within the LNs. Short-term treatment of murine cardiac allograft recipients with MECA79-anti-CD3-NP resulted in significantly prolonged allograft survival in comparison with the control groups. Prolonged graft survival following treatment with MECA79-anti-CD3-NP was characterized by a significant increase in intragraft and DLN Treg populations. Treg depletion abrogated the prolongation of heart allograft survival. We believe this targeted approach of drug delivery could redefine the methods of administering immune therapeutics in transplantation.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Portadores de Fármacos/farmacología , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Corazón , Ganglios Linfáticos/inmunología , Nanopartículas/uso terapéutico , Tacrolimus/farmacología , Aloinjertos , Animales , Anticuerpos Monoclonales de Origen Murino/química , Células Dendríticas/inmunología , Células Dendríticas/patología , Portadores de Fármacos/química , Supervivencia de Injerto/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Nanopartículas/química , Linfocitos T/inmunología , Tacrolimus/química
13.
JCI Insight ; 3(13)2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29997302

RESUMEN

The contribution of the kidney-draining lymph node (KLN) to the pathogenesis of ischemia-reperfusion injury (IRI) of the kidney and its subsequent recovery has not been explored in depth. In addition, the mechanism by which repetitive IRI contributes to renal fibrosis remains poorly understood. Herein, we have found that IRI of the kidney is associated with expansion of high endothelial venules (HEVs) and activation of fibroblastic reticular cells (FRCs) in the KLN, as demonstrated by significant expansion in the extracellular matrix. The lymphotoxin α signaling pathway mediates activation of FRCs, and chronic treatment with lymphotoxin ß receptor-immunoglobulin fusion protein (LTßr-Ig) resulted in marked alteration of the KLN as well as augmentation of renal fibrosis. Depletion of FRCs reduced T cell activation in the KLN and ameliorated renal injury in acute IRI. Repetitive renal IRI was associated with senescence of FRCs, fibrosis of the KLN, and renal scarring, which were ameliorated by FRC administration. Therefore, our study emphasizes the critical role of FRCs in both the initiation and repair phases of injury following IRI of the kidney.


Asunto(s)
Lesión Renal Aguda/complicaciones , Fibrosis/complicaciones , Riñón/patología , Ganglios Linfáticos/patología , Daño por Reperfusión/complicaciones , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/patología , Animales , Modelos Animales de Enfermedad , Fibrosis/inmunología , Proteínas de Homeodominio/genética , Inmunoglobulinas , Receptor beta de Linfotoxina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión/inmunología , Transducción de Señal
14.
Sci Rep ; 8(1): 2461, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29410442

RESUMEN

Ischemia reperfusion injuries (IRI) are unavoidable in solid organ transplantation. IRI augments alloimmunity but the mechanisms involved are poorly understood. Herein, we examined the effect of IRI on antigen specific alloimmunity. We demonstrate that ischemia promotes alloimmune activation, leading to more severe histological features of rejection, and increased CD4+ and CD8+ T cell graft infiltration, with a predominantly CD8+ IFNγ+ infiltrate. This process is dependent on the presence of alloreactive CD4+ T cells, where depletion prevented infiltration of ischemic grafts by CD8+ IFNγ+ T cells. IL-6 is a known driver of ischemia-induced rejection. Herein, depletion of donor antigen-presenting cells reduced ischemia-induced CD8+ IFNγ+ allograft infiltration, and improved allograft outcomes. Following prolonged ischemia, accelerated rejection was observed despite treatment with CTLA4Ig, indicating that T cell costimulatory blockade failed to overcome the immune activating effect of IRI. However, despite severe ischemic injury, treatment with anti-IL-6 and CTLA4Ig blocked IRI-induced alloimmune injury and markedly improved allograft survival. We describe a novel pathway where IRI activates innate immunity, leading to upregulation of antigen specific alloimmunity, resulting in chronic allograft injury. Based on these findings, we describe a clinically relevant treatment strategy to overcome the deleterious effect of IRI, and provide superior long-term allograft outcomes.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Rechazo de Injerto/inmunología , Trasplante de Corazón , Interleucina-6/inmunología , Daño por Reperfusión Miocárdica/inmunología , Abatacept/farmacología , Animales , Anticuerpos Neutralizantes/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/patología , Movimiento Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/patología , Regulación de la Expresión Génica , Rechazo de Injerto/genética , Rechazo de Injerto/fisiopatología , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Inmunosupresores/farmacología , Interferón gamma/antagonistas & inhibidores , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Transducción de Señal , Trasplante Heterotópico , Trasplante Homólogo
15.
Nat Commun ; 8(1): 951, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29038423

RESUMEN

Phosphatidylinositol-3-kinases (PI3K) γ and δ are preferentially enriched in leukocytes, and defects in these signaling pathways have been shown to impair T cell activation. The effects of PI3Kγ and PI3Kδ on alloimmunity remain underexplored. Here, we show that both PI3Kγ -/- and PI3Kδ D910A/D910A mice receiving heart allografts have suppression of alloreactive T effector cells and delayed acute rejection. However, PI3Kδ mutation also dampens regulatory T cells (Treg). After treatment with low dose CTLA4-Ig, PI3Kγ -/- , but not PI3Κδ D910A/D910A , recipients exhibit indefinite prolongation of heart allograft survival. PI3Kδ D910A/D910A Tregs have increased apoptosis and impaired survival. Selective inhibition of PI3Kγ and PI3Kδ (using PI3Kδ and dual PI3Kγδ chemical inhibitors) shows that PI3Kγ inhibition compensates for the negative effect of PI3Kδ inhibition on long-term allograft survival. These data serve as a basis for future PI3K-based immune therapies for transplantation.Phosphatidylinositol-3-kinases (PI3K) γ and δ are key regulators of T cell signaling. Here the author show, using mouse heart allograft transplantation models, that PI3Kγ or PI3Kδ deficiency prolongs graft survival, but selective inhibition of PI3Kγ or PI3Kδ reveals alternative transplant survival outcomes post CTLA4-Ig treatment.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/inmunología , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Corazón , Activación de Linfocitos/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Trasplante de Piel , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante/inmunología , Abatacept/farmacología , Animales , Fosfatidilinositol 3-Quinasa Clase I , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Técnicas de Silenciamiento del Gen , Supervivencia de Injerto/efectos de los fármacos , Inmunosupresores/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Noqueados , Modelos Animales , Mutación , Fosfatidilinositol 3-Quinasas/genética , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Tolerancia al Trasplante/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA