Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Microbiol Res ; 285: 127750, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38761489

RESUMEN

The progress of viral infection involves numerous transcriptional regulatory events. The identification of the newly synthesized transcripts helps us to understand the replication mechanisms and pathogenesis of the virus. Here, we utilized a time-resolved technique called metabolic RNA labeling approach called thiol(SH)-linked alkylation for the metabolic sequencing of RNA (SLAM-seq) to differentially elucidate the levels of steady-state and newly synthesized RNAs of BHK21 cell line in response to human coronavirus OC43 (HCoV-OC43) infection. Our results showed that the Wnt/ß-catenin signaling pathway was significantly enriched with the newly synthesized transcripts of BHK21 cell line in response to HCoV-OC43 infection. Moreover, inhibition of the Wnt pathway promoted viral replication in the early stage of infection, but inhibited it in the later stage of infection. Furthermore, remdesivir inhibits the upregulation of the Wnt/ß-catenin signaling pathway induced by early infection with HCoV-OC43. Collectively, our study showed the diverse roles of Wnt/ß-catenin pathway at different stages of HCoV-OC43 infection, suggesting a potential target for the antiviral treatment. In addition, although infection with HCoV-OC43 induces cytopathic effects in BHK21 cells, inhibiting apoptosis does not affect the intracellular replication of the virus. Monitoring newly synthesized RNA based on such time-resolved approach is a highly promising method for studying the mechanism of viral infections.

2.
iScience ; 26(9): 107691, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37694154

RESUMEN

The liver has long been deemed a tolerogenic organ. We employed high-dimensional mass cytometry and immunohistochemistry to depict the temporal and spatial dynamics of immune cells in the spleen and liver in a murine model of spontaneous liver allograft acceptance. We depicted the immune landscape of spontaneous liver tolerance throughout the rejection and acceptance stages after liver transplantation and highlighted several points of importance. Of note, the CD4+/CD8+ T cell ratio remained low, even in the tolerance phase. Furthermore, a PhenoGraph clustering analysis revealed that exhausted CD8+ T cells were the most dominant metacluster in graft-infiltrating lymphocytes (GILs), which highly expressed the costimulatory molecule CD86. The temporal and spatial dynamics of immune cells revealed by high-dimensional analyses enable a fine-grained analysis of GIL subsets, contribute to new insights for the discovery of immunological mechanisms of liver tolerance, and provide potential ways to achieve clinical operational tolerance after liver transplantation.

3.
Front Immunol ; 14: 1158360, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37483608

RESUMEN

Background: Liver hepatocellular carcinoma (LIHC) is one of the malignant tumors with high incidence as well as high death, which is ranked as the sixth most common tumor and the third highest mortality worldwide. CD93, a transmembrane protein, has been widely reported to play an important role in different types of diseases, including many types of cancer by mainly functioning in extracellular matrix formation and vascular maturation. However, there are few researches focusing on the role and potential function of CD93 in LIHC. Methods: In this study, we comprehensively analyzed the relationship between CD93 and LIHC. We not only discovered transcriptional expression of CD93 in LIHC by using the TIMER, GEPIA and UALCAN database, but also performed WB and IHC to verify the protein expression of CD93 in LIHC. Meantime, Kaplan-Meier Plotter Database Analysis were used to assess the prognosis of CD93 in LIHC. After knowing close correlation between CD93 expression and LIHC, there were STRING, GeneMania and GO and KEGG enrichment analyses to find how CD93 functions in LIHC. We further applied CIBERSORT Algorithm to explore the correlation between CD93 and immune cells and evaluate prognostic value of CD93 based on them in LIHC patients. Results: The transcriptional and protein expression of CD93 were both obviously increased in LIHC by above methods. There was also a significant and close correlation between the expression of CD93 and the prognosis of LIHC patients by using Kaplan-Meier Analysis, which showed that LIHC patients with elevated expression of CD93 were associated with a predicted poor prognosis. We found that the functions of CD93 in different cancers are mainly related to Insulin like growth factor binding protein 7 Gene (IGFBP7)/CD93 pathway via STRING, GeneMania and functional enrichment analyses. Further, our data obtained from CIBERSORT Algorithm suggested CD93 was also associated with the immune response. There is a close positive correlation between CD93 expression and the infiltration levels of all six types of immune cells (B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells). Importantly, CD93 can affect the prognosis of patients with LIHC partially due to immune infiltration. Conclusion: Our results demonstrated CD93 may be a candidate predictor of clinical prognosis and immunotherapy response in LIHC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Algoritmos , Inmunoterapia
4.
Sci Adv ; 8(31): eabo4413, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35921418

RESUMEN

Type 1 regulatory T (Tr1) cells represent a subset of IL-10-producing CD4+Foxp3- T cells and play key roles in promoting transplant tolerance. However, no effective pharmacological approaches have been able to induce Tr1 cells in vivo. We herein report the combined use of a CD28 superagonist (D665) and anti-glucocorticoid-induced tumor necrosis factor receptor-related protein monoclonal antibody (G3c) to induce Tr1 cells in vivo. Large amounts of IL-10/interferon-γ-co-producing CD4+Foxp3- Tr1 cells were generated by D665-G3c sequential treatment in mice. Mechanistic studies suggested that D665-G3c induced Tr1 cells via transcription factors Prdm1 and Maf. G3c contributed to Tr1 cell generation via the activation of mitogen-activated protein kinase-signal transducer and activator of transcription 3 signaling. Tr1 cells suppressed dendritic cell maturation and T cell responses and mediated permanent allograft acceptance in fully major histocompatibility complex-mismatched mice in an IL-10-dependent manner. In vivo Tr1 cell induction is a promising strategy for achieving transplant tolerance.

5.
Int Immunopharmacol ; 110: 109028, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35803130

RESUMEN

Although double positive CD4+CD8+ T (DPT) cells has been reported to be involved in some diseases, their trajectory and function as associated with liver transplantation (LT) remain unclear. In the present study, we found that the number of DPT cells was increased in the blood and liver tissue of LT patients. Meanwhile, we compared the distribution of DPT cells in peripheral blood samples and in penetrating liver tissue between liver rejection versus non-rejection patients, as well as the proportion of DPT cells as a function of the extent of liver rejection. The number of DPT cells in the rejection group was significantly increased. An analysis of the spatial distance and correlations between DPT and Treg cells, revealed that these cells showed a high degree of contiguity. In a mouse liver transplant model, the number of DPT cells were significantly increased in liver tissue, and the number of CD8+ T cells gradually increased, while CD4+ T cells decreased as a function of time post-transplantation. Expression level of PD-1 in DPT cells also increased in a temporally-dependent manner post liver transplantation and the changes of PD-1+ DPT cells were related to the degree of liver transplant rejection. In DPT cells interacting with Treg, there was an increased expression of PD-1, which enhanced cellular exhaustion. In conclusion, the capacity for DPT cells to induce immune tolerance may represent a new and important protocol for use in targeting treatments for the prevention of liver transplant rejection.


Asunto(s)
Trasplante de Hígado , Animales , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Rechazo de Injerto , Ratones , Receptor de Muerte Celular Programada 1 , Linfocitos T Reguladores
6.
Clin Exp Immunol ; 208(3): 340-350, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35511600

RESUMEN

Ulcerative colitis (UC) is one of the two main forms of inflammatory bowel disease (IBD) and is an idiopathic, chronic inflammatory disease of the colonic mucosa with an unclear etiology. Interleukin (IL)-10 has been reported to play a crucial role in the maintenance of immune homeostasis in the intestinal environment. Type 1 regulatory T (Tr1) cells are a subset of CD4+Foxp3- T cells able to secrete high amounts of IL-10 with potent immunosuppressive properties. In this study, we found that the combination of anti-GITR antibody (G3c) and CD28 superagonist (D665) treatment stimulated the generation of a large amount of Tr1 cells. Furthermore, G3c/D665 treatment not only significantly relieved severe mucosal damage but also reduced the incidence of colonic shortening, weight loss, and hematochezia. Dextran sodium sulfate (DSS) upregulated the mRNA levels of IL-6, IL-1ß, IL-17, IL-12, tumor necrosis factor-alpha, C-C chemokine receptor type 5, and Bax in splenic lymphocytes (SPLs) and colon tissues, while G3c/D665 treatment conversely inhibited the increase in mRNA levels of these genes. In addition, G3c/D665 treatment altered the proportion of CD4+ and CD8+ T cells and increased CD4+CD25+Foxp3+ regulatory T cells in SPLs, mesenteric lymph nodes (MLNs), and lamina propria lymphocytes (LPLs). Thus, the combination of G3c and D665 treatment showed efficacy against DSS-induced UC in mice by inducing a large amount of Tr1 cell generation via the musculoaponeurotic fibrosarcoma pathways in vivo and relieving inflammatory responses both systematically and locally.


Asunto(s)
Colitis Ulcerosa , Colitis , Animales , Antígenos CD28/metabolismo , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colon , Sulfato de Dextran , Factores de Transcripción Forkhead/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Sulfatos , Linfocitos T Reguladores
7.
Clin Transl Med ; 12(5): e849, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35593197

RESUMEN

Sepsis remains the most lethal infectious disease and substantially impairs patient prognosis after liver transplantation (LT). Our previous study reported a role of the pannexin 1 (PANX1)-interleukin-33 (IL-33) axis in activating innate immunity to protect against methicillin-resistant Staphylococcus aureus infection; however, the role of PANX1 in regulating adaptive immunity in sepsis and the underlying mechanism are unclear. In this study, we examined the role of the PANX1-IL-33 axis in protecting against sepsis caused by a gram-negative bacterial infection in an independent LT cohort. Next, in animal studies, we assessed the immunological state of Panx1-/- mice with lipopolysaccharide (LPS)-induced endotoxemia and then focused on the cytokine storm and regulatory T cells (Tregs), which are crucial for the resolution of inflammation. To generate liver-specific Panx1-deficient mice and mimic clinical LT procedures, a mouse LT model was established. We demonstrated that hepatic PANX1 deficiency exacerbated LPS-induced endotoxemia and dysregulated the immune response in the mouse LT model. In hepatocytes, we confirmed that PANX1 positively regulated IL-33 synthesis after LPS administration. We showed that the adenosine triphosphate-P2X7 pathway regulated the hepatic PANX1-IL-33 axis during endotoxemia in vitro and in vivo. Recombinant IL-33 treatment rescued LPS-induced endotoxemia by increasing the numbers of liver-infiltrating ST2+ Tregs and attenuating the cytokine storm in hepatic PANX1-deficient mice. In conclusion, our findings revealed that the hepatic PANX1-IL-33 axis protects against endotoxemia and liver injury by targeting ST2+ Tregs and promoting the early resolution of hyperinflammation.


Asunto(s)
Endotoxemia , Staphylococcus aureus Resistente a Meticilina , Sepsis , Animales , Conexinas/genética , Conexinas/metabolismo , Síndrome de Liberación de Citoquinas , Modelos Animales de Enfermedad , Endotoxemia/inducido químicamente , Humanos , Inflamación/inducido químicamente , Inflamación/complicaciones , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Lipopolisacáridos/toxicidad , Hígado/metabolismo , Staphylococcus aureus Resistente a Meticilina/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Sepsis/complicaciones , Linfocitos T Reguladores/metabolismo
8.
MedComm (2020) ; 3(2): e137, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35474948

RESUMEN

CD4+CD25+ regulatory T cells (Tregs), a subpopulation of naturally CD4+ T cells that characteristically express transcription factor Forkhead box P3 (FOXP3), play a pivotal role in the maintenance of immune homeostasis and the prevention of autoimmunity. With the development of biological technology, the understanding of plasticity and stability of Tregs has been further developed. Recent studies have suggested that human Tregs are functionally and phenotypically diverse. The functions and mechanisms of different phenotypes of Tregs in different disease settings, such as tumor microenvironment, autoimmune diseases, and transplantation, have gradually become hot spots of immunology research that arouse extensive attention. Among the complex functions, CD4+CD25+FOXP3+ Tregs possess a potent immunosuppressive capacity and can produce various cytokines, such as IL-2, IL-10, and TGF-ß, to regulate immune homeostasis. They can alleviate the progression of diseases by resisting inflammatory immune responses, whereas promoting the poor prognosis of diseases by helping cells evade immune surveillance or suppressing effector T cells activity. Therefore, methods for targeting Tregs to regulate their functions in the immune microenvironment, such as depleting them to strengthen tumor immunity or expanding them to treat immunological diseases, need to be developed. Here, we discuss that different subpopulations of Tregs are essential for the development of immunotherapeutic strategies involving Tregs in human diseases.

9.
Liver Transpl ; 28(5): 782-793, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34529892

RESUMEN

Small-for-size syndrome (SFSS) is a common complication following partial liver transplantation and extended hepatectomy. SFSS is characterized by postoperative liver dysfunction caused by insufficient regenerative capacity and portal hyperperfusion and is more frequent in patients with preexisting liver disease. We explored the effect of the Mesenchymal-epithelial transition factor (MET)-agonistic antibody 71D6 on liver regeneration and functional recovery in a mouse model of SFSS. Male C57/BL6 mice were exposed to repeated carbon tetrachloride injections for 10 weeks and then randomized into 2 arms receiving 3 mg/kg 71D6 or a control immunoglobulin G (IgG). At 2 days after the randomization, the mice were subjected to 70% hepatectomy. Mouse survival was recorded up to 28 days after hepatectomy. Satellite animals were euthanized at different time points to analyze liver regeneration, fibrosis, and inflammation. Serum 71D6 administration significantly decreased mouse mortality consequent to insufficient regeneration of the cirrhotic liver. Analysis of liver specimens in satellite animals revealed that 71D6 promoted powerful activation of the extracellular signal-regulated kinase pathway and accelerated liver regeneration, characterized by increased liver-to-body weight, augmented mitotic index, and higher serum albumin levels. Moreover, 71D6 accelerated the resolution of hepatic fibrosis as measured by picrosirius red, desmin, and α-smooth muscle actin staining, and suppressed liver infiltration by macrophages as measured by CD68 and F4/80 staining. Analysis of gene expression by reverse-transcription polymerase chain reaction confirmed that 71D6 administration suppressed the expression of key profibrotic genes, including platelet-derived growth factor, tissue inhibitor of metalloproteinase 3, and transforming growth factor-ß1, and of key proinflammatory genes, including tumor necrosis factor-α, interleukin-1ß, chemokine (C-C motif) ligand 3, and chemokine (C-C motif) ligand 5. These results suggest that activating the MET pathway via an hepatocyte growth factor-mimetic antibody may be beneficial in patients with SFSS and possibly other types of acute and chronic liver disorders.


Asunto(s)
Regeneración Hepática , Trasplante de Hígado , Animales , Hepatectomía/efectos adversos , Humanos , Hígado/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones
10.
Front Immunol ; 12: 719574, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566976

RESUMEN

Background: Oridonin (Ori), the main bioactive ingredient of the natural anti-inflammatory herb Rabdosia rubescens, could be a covalent inhibitor of the NLRP3 inflammasome. Solid organ transplantation provides a life-saving optional therapy for patients with end-stage organ dysfunction. The long-term survival of solid organ transplantation remains restricted because of the possibility of rejection and the toxicity, infection, cardiovascular disease, and malignancy related to immunosuppressive (IS) drugs. However, the pathogenic mechanisms involved remain unclear. The ideal IS drugs to prevent allograft rejection have not been identified. Here, we investigated whether Ori could prolong the in vivo survival of completely mismatched cardiac allografts. Methods: The cardiac transplantation models were conducted among three groups of mice from C57BL/6NCrSlc (B6/N) or C3H/HeNSlc (C3H) to C3H: the syngeneic and the allogeneic group, whose recipients were treated with vehicle of Ori, and the Ori treatment group, in which the recipients were transplanted hearts from MHC-I mismatched donors and treated with different dosages of Ori from post-operative day (POD) 0 to 7. Then, we investigated the effect of Ori on bone marrow-derived dendritic cell (BMDC) and allogeneic mixed lymphocyte reaction in vitro. Results: Ori with 3, 10, and 15 mg/kg Ori could prolong the survival (MST = 22.8, 49.2, and 65.3 days, respectively). We found that infiltrating CD8+ T cells and macrophages were decreased, and regulatory T cells (Tregs) were expanded in allografts on POD7. The mRNA level of IL-1ß and IFN-γ of allografts was downregulated. Mechanistically, Ori-treated BMDCs suppressed T-cell proliferation and IFN-γ+CD4+ T-cell differentiation, along with the expansion of Tregs and IL-10+CD4+ T cells. Ori inhibited NOD, LRR-, and pyrin domain-containing protein 3 (NLRP3) expression; attenuated NF-κB and IκBα phosphorylation in LPS-activated BMDCs; downregulated NLRP3, Caspase-1, IL-1ß, IL-18, and IFN-γ; and upregulated IL-10 expression. Conclusions: Our findings highlight the potential of Ori as a novel and natural IS agent to improve transplant tolerance. Ori could exert IS activity through decreasing IL-1ß and IL-18 production and Th1 differentiation and proliferation and expanding Tregs via inhibiting the NF-κB/NLRP3 signaling pathway.


Asunto(s)
Diterpenos de Tipo Kaurano/farmacología , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Corazón , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal/efectos de los fármacos , Aloinjertos , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Rechazo de Injerto , Supervivencia de Injerto/inmunología , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Modelos Biológicos , Pronóstico , Resultado del Tratamiento
11.
World J Gastroenterol ; 27(22): 3022-3036, 2021 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-34168405

RESUMEN

In the early December 2019, a novel coronavirus named severe acute respiratory syndrome coronavirus 2 was first reported in Wuhan, China, followed by an outbreak that spread around the world. Numerous studies have shown that liver injury is common in patients with coronavirus disease 2019 (COVID-19), and may aggravate the severity of the disease. However, the exact cause and specific mechanism of COVID-associated liver injury needs to be elucidated further. In this review, we present an analysis of the clinical features, potential mechanisms, and treatment strategies for liver injury associated with COVID-19. We hope that this review would benefit clinicians in devising better strategies for management of such patients.


Asunto(s)
COVID-19 , Hepatopatías/virología , COVID-19/complicaciones , China/epidemiología , Humanos , SARS-CoV-2
12.
Cancer Sci ; 112(7): 2652-2663, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33934440

RESUMEN

Mitochondria are key cytoplasmic organelles. Their activation is critical for the generation of T cell proliferation and cytotoxicity. Exhausted tumor-infiltrating T cells show a decreased mitochondrial function and mass. 5-Aminolevulinic acid (5-ALA), a natural amino acid that is only produced in the mitochondria, has been shown to influence metabolic functions. We hypothesized that 5-ALA with sodium ferrous citrate (SFC) might provide metabolic support for tumor-infiltrating T cells. In a mouse melanoma model, we found that 5-ALA/SFC with a programmed cell death-ligand 1 (PD-L1) blocking Ab synergized tumor regression. After treatment with 5-ALA/SFC and anti-PD-L1 Ab, tumor infiltrating lymphocytes (TILs) were not only competent for the production of cytolytic particles and cytokines (granzyme B, interleukin-2, and γ-interferon) but also showed enhanced Ki-67 activity (a proliferation marker). The number of activated T cells (PD-1+ Tim-3- ) was also significantly increased. Furthermore, we found that 5-ALA/SFC activated the mitochondrial functions, including the oxygen consumption rate, ATP level, and complex V expression. The mRNA levels of Nrf-2, HO-1, Sirt-1, and PGC-1α and the protein levels of Sirt-1 were upregulated by treatment with 5-ALA/SFC. Taken together, our findings revealed that 5-ALA/SFC could be a key metabolic regulator in exhausted T cell metabolism and suggested that 5-ALA/SFC might synergize with anti-PD-1/PD-L1 therapy to boost the intratumoral efficacy of tumor-specific T cells. Our study not only revealed a new aspect of immune metabolism, but also paved the way to develop a strategy for combined anti-PD-1/PD-L1 cancer immunotherapy.


Asunto(s)
Ácido Aminolevulínico/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Ácido Cítrico/farmacología , Compuestos Ferrosos/farmacología , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Línea Celular Tumoral , Terapia Combinada , Femenino , Hemo-Oxigenasa 1/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Antígeno Ki-67/metabolismo , Recuento de Linfocitos , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Melanoma Experimental/terapia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Factor Nuclear 1 de Respiración/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Sirtuina 1/metabolismo
13.
BMC Cancer ; 21(1): 521, 2021 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-33964921

RESUMEN

BACKGROUND: Complement component(C7) gene has been shown to influence the prognosis in Hepatocellular carcinoma (HCC) patients. The association between C7 and HCC recurrence after orthotopic liver transplantation (OLT), however, is still unknown. The purpose of this study was to evaluate whether the donor and recipient C7 gene polymorphisms are related to HCC recurrence after OLT in the Han Chinese population. METHODS: A total of 73 consecutive patients with HCC who had undergone OLT, both donors and recipients, were involved in this research. A single nucleotide polymorphism of C7, rs9292795, was genotyped using Sequenom MassARRAY in the cohort. The expression of C7 and the association between C7 gene polymorphisms and HCC recurrence following OLT were analyzed by bioinformatics and statistical analysis, respectively. RESULTS: As shown in database, the expression of C7 was higher in HCC tissues than that in normal tissues, and represented a worse prognosis. We also found that recipient C7 rs9292795 polymorphism, rather than the donor, was significantly associated with HCC recurrence after OLT. Multivariate logistic regression analysis confirmed that TNM stage (P = 0.001), Milan criteria (P = 0.000) and recipient rs9292795 genotype (TT vs AA/AT, P = 0.008) were independent risk factors for HCC recurrence. Furthermore, the recipient carrying AA/AT showed higher recurrence-free survival (RFS) and overall survival (OS) than that carrying TT (P < 0.05). In Cox proportional hazards model, TNM stage, recipient rs9292795 genotype, and Milan criteria were identified as independent factors for RFS and OS (P < 0.05) as well as pre-OLT serum alpha fetoprotein (AFP) level was associated with OS (P < 0.05). CONCLUSIONS: Recipient C7 rs9292795 gene polymorphism is related to the recurrence of HCC after OLT, which may be a helpful prognostic marker for HCC patients who receive OLT.


Asunto(s)
Carcinoma Hepatocelular/genética , Complemento C7/genética , Neoplasias Hepáticas/genética , Trasplante de Hígado/efectos adversos , Recurrencia Local de Neoplasia/genética , Polimorfismo de Nucleótido Simple , Adulto , Anciano , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/cirugía , Femenino , Genotipo , Humanos , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/cirugía , Modelos Logísticos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/mortalidad , Modelos de Riesgos Proporcionales , Factores de Riesgo , Adulto Joven
14.
Adv Exp Med Biol ; 1278: 289-302, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33523454

RESUMEN

Organ transplantation is the optimal treatment for terminal and irreversible organ failure. Achieving transplantation tolerance has long been the ultimate goal in the field of transplantation. Regulatory T cell (Treg)-based therapy is a promising novel approach for inducing donor organ-specific tolerance. Tregs play critical roles in the maintenance of immune homeostasis and self-tolerance, by promoting transplantation tolerance through a variety of mechanisms on different target cells, including anti-inflammatory cytokine production, induction of apoptosis, disruption of metabolic pathways, and mutual interaction with dendritic cells. The continued success of Treg-based therapy in the clinical setting is critically dependent on preclinical studies that support its translational potential. However, although some initial clinical trials of adoptive Treg therapy have successively demonstrated safety and efficacy for immunosuppressant minimization and transplantation tolerance induction, most Treg-based hematopoietic stem cell and solid organ clinical trials are still in their infancy. These clinical trials have not only focused on safety and efficacy but also included optimization and standardization protocols of good manufacturing practice regarding cell isolation, expansion, dosing, timing, specificity, quality control, concomitant immunosuppressants, and post-administration monitoring. We herein report a brief introduction of Tregs, including their phenotypic and functional characterization, and focus on the clinical translation of Treg-based therapeutic applications in the setting of transplantation.


Asunto(s)
Trasplante de Órganos , Tolerancia al Trasplante , Tolerancia Inmunológica , Linfocitos T Reguladores
15.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G450-G463, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33439102

RESUMEN

Nonalcoholic steatohepatitis (NASH) could progress to hepatic fibrosis in the absence of effective control. The purpose of our experiment was to investigate the protective effect of drinking water with a high concentration of hydrogen, namely, hydrogen-rich water (HRW), on mice with nonalcoholic fatty liver disease to elucidate the mechanism underlying the therapeutic action of molecular hydrogen. The choline-supplemented, l-amino acid-defined (CSAA) or the choline-deficient, l-amino acid-defined (CDAA) diet for 20 wk was used to induce NASH and fibrosis in the mice model and simultaneously treated with the high-concentration 7-ppm HRW for different periods (4 wk, 8 wk, and 20 wk). Primary hepatocytes were stimulated by palmitate to mimic liver lipid metabolism during fatty liver formation. Primary hepatocytes were cultured in a closed vessel filled with 21% O2 + 5% CO2 + 3.8% H2 and N2 as the base gas to verify the response of primary hepatocytes in a high concentration of hydrogen gas in vitro. Mice in the CSAA + HRW group had lower serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and milder histological damage. The inflammatory cytokines were expressed at lower levels in the HRW group than in the CSAA group. Importantly, HRW reversed hepatocyte fatty acid oxidation and lipogenesis as well as hepatic inflammation and fibrosis in preexisting hepatic fibrosis specimens. Molecular hydrogen inhibits the lipopolysaccharide-induced production of inflammation cytokines through increasing heme oxygenase-1 (HO-1) expression. Furthermore, HRW improved hepatic steatosis in the CSAA + HRW group. Sirtuin 1 (Sirt1) induction by molecular hydrogen via the HO-1/adenosine monophosphate activated protein kinase (AMPK)/peroxisome proliferator-activated receptor α (PPARα)/peroxisome proliferator-activated receptor γ (PPAR-γ) pathway suppresses palmitate-mediated abnormal fat metabolism. Orally administered HRW suppressed steatosis induced by CSAA and attenuated fibrosis induced by CDAA, possibly by reducing oxidative stress and the inflammation response.NEW & NOTEWORTHY The mRNA expression of inflammatory cytokines in the HRW group was lower than in the CSAA group. HRW reversed hepatocyte apoptosis as well as hepatic inflammation and fibrosis in NASH specimens. Molecular hydrogen inhibits LPS-induced inflammation via an HO-1/interleukin 10 (IL-10)-independent pathway. HRW improved hepatic steatosis in the CSAA + HRW group. Sirt1 induction by molecular hydrogen via the HO-1/AMPK/PPARα/PPARγ pathway suppresses palmitate-mediated abnormal fat metabolism.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Hepatocitos/efectos de los fármacos , Hidrógeno/farmacología , Interleucina-10/metabolismo , Cirrosis Hepática Experimental/prevención & control , Hígado/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Sirtuina 1/metabolismo , Agua/farmacología , Animales , Hepatocitos/enzimología , Hepatocitos/patología , Hidrógeno/química , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Lipólisis/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Cirrosis Hepática Experimental/enzimología , Cirrosis Hepática Experimental/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/patología , Células RAW 264.7 , Transducción de Señal
16.
Drug Discov Ther ; 14(6): 304-312, 2021 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-33390570

RESUMEN

Acute graft-versus-host disease (aGvHD) remains lethal as a life-threatening complication after allogeneic hematopoietic stem cell transplantation (HSCT). Inflammatory responses play an important role in aGvHD. 5-Aminolevulinic acid combined with sodium ferrous citrate (5-ALA/SFC) has been widely reported to have a major effect on the anti-inflammatory response; however, these effects in aGvHD models have never been reported. In this study, a murine aGvHD model was developed by transferring spleen cells from donor B6/N (H-2kb) mice into recipient B6D2F1 (H-2kb/d) mice. In addition to evaluating manifestations in aGvHD mice, we analyzed the serum ALT/AST levels, liver pathological changes, infiltrating cells and mRNA expression of inflammation-related cytokines and chemokines. 5-ALA/SFC treatment significantly ameliorated liver injury due to aGvHD and decreased the population of liver-infiltrating T cells, resulting in a reduced expression of pro-inflammatory cytokines and chemokines. Furthermore, the mRNA expression proliferator-activated receptor-γcoactivator (PGC-1α) was enhanced, which might explain why 5-ALA/SFC treatment downregulates inflammatory signaling pathways. Our results indicated that 5-ALA/SFC can ameliorate liver injury induced by aGvHD through the activation of PGC-1α and modulation of the liver mRNA expression of inflammatory-related cytokines and chemokines. This may be a novel strategy for treating this disease.


Asunto(s)
Citocinas/genética , Compuestos Ferrosos/administración & dosificación , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Ácidos Levulínicos/administración & dosificación , Hígado/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Regulación hacia Arriba , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Ácido Cítrico , Modelos Animales de Enfermedad , Quimioterapia Combinada , Compuestos Ferrosos/química , Compuestos Ferrosos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/genética , Ácidos Levulínicos/farmacología , Hígado/inmunología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Citrato de Sodio/química , Linfocitos T/metabolismo , Resultado del Tratamiento , Ácido Aminolevulínico
17.
Front Immunol ; 11: 582658, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33162996

RESUMEN

Current organ transplantation therapy is life-saving but accompanied by well-recognized side effects due to post-transplantation systematic immunosuppressive treatment. Dendritic cells (DCs) are central instigators and regulators of transplantation immunity and are responsible for balancing allograft rejection and tolerance. They are derived from monocyte-macrophage DC progenitors originating in the bone marrow and are classified into different subsets based on their developmental, phenotypical, and functional criteria. Functionally, DCs instigate allograft immunity by presenting donor antigens to alloreactive T cells via direct, indirect, and semidirect recognition pathways and provide essential signaling for alloreactive T cell activation via costimulatory molecules and pro-inflammatory cytokines. Regulatory DCs (DCregs) are characterized by a relatively low expression of major histocompatibility complex, costimulatory molecules, and altered cytokine production and exert their regulatory function through T cell anergy, T cell deletion, and regulatory T cell induction. In rodent transplantation studies, DCreg-based therapy, by in situ targeting or infusion of ex vivo generated DCregs, exhibits promising potential as a natural, well-tolerated, organ-specific therapeutic strategy for promoting lasting organ-specific transplantation tolerance. Recent early-phase studies of DCregs have begun to examine the safety and efficacy of DCreg-induced allograft tolerance in living-donor renal or liver transplantations. The present review summarizes the basic characteristics, function, and translation of DCregs in transplantation tolerance induction.


Asunto(s)
Células Dendríticas/inmunología , Rechazo de Injerto/inmunología , Trasplante de Órganos , Linfocitos T/inmunología , Aloinjertos/inmunología , Animales , Diferenciación Celular , Supervivencia de Injerto , Humanos , Inmunomodulación , Activación de Linfocitos , Tolerancia al Trasplante
18.
Front Immunol ; 11: 1203, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32595648

RESUMEN

Liver transplantation is the ideal treatment approach for a variety of end-stage liver diseases. However, life-long, systemic immunosuppressive treatment after transplantation is required to prevent rejection and graft loss, which is associated with severe side effects, although liver allograft is considered more tolerogenic. Therefore, understanding the mechanism underlying the unique immunologically privileged liver organ is valuable for transplantation management and autoimmune disease treatment. The unique hepatic acinus anatomy and a complex cellular network constitute the immunosuppressive hepatic microenvironment, which are responsible for the tolerogenic properties of the liver. The hepatic microenvironment contains a variety of hepatic-resident immobile non-professional antigen-presenting cells, including hepatocytes, liver sinusoidal endothelial cells, Kupffer cells, and hepatic stellate cells, that are insufficient to optimally prime T cells locally and lead to the removal of alloreactive T cells due to the low expression of major histocompatibility complex (MHC) molecules, costimulatory molecules and proinflammatory cytokines but a rather high expression of coinhibitory molecules and anti-inflammatory cytokines. Hepatic dendritic cells (DCs) are generally immature and less immunogenic than splenic DCs and are also ineffective in priming naïve allogeneic T cells via the direct recognition pathway in recipient secondary lymphoid organs. Although natural killer cells and natural killer T cells are reportedly associated with liver tolerance, their roles in liver transplantation are multifaceted and need to be further clarified. Under these circumstances, T cells are prone to clonal deletion, clonal anergy and exhaustion, eventually leading to tolerance. Other proposed liver tolerance mechanisms, such as soluble donor MHC class I molecules, passenger leukocytes theory and a high-load antigen effect, have also been addressed. We herein comprehensively review the current evidence implicating the tolerogenic properties of diverse liver cells in liver transplantation tolerance.


Asunto(s)
Trasplante de Hígado , Hígado/inmunología , Tolerancia al Trasplante/inmunología , Animales , Humanos , Hígado/citología
19.
Transplantation ; 104(9): 1879-1889, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31895334

RESUMEN

BACKGROUND: Supercooling preservation techniques store a donor organ below 0°C without freezing. This has great advantages in inhibiting metabolism and preserving the organ in comparison to conventional preservation at 4°C. We developed a novel supercooling technique using a liquid cooling apparatus and novel preservation and perfusion solutions. The purpose of this study was to evaluate the preservation effect of our supercooling preservation technique in a mouse heart transplantation model. METHODS: Syngeneic heterotopic heart transplantation was performed in 3 groups of mice: (1) the nonpreservation group, in which the cardiac grafts were transplanted immediately after retrieval; (2) the conventional University of Wisconsin (UW) group, in which the cardiac grafts were stored in UW solution at 4°C for different periods of time; and (3) the supercooling group, in which the cardiac grafts were stored in a novel supercooling preservation solution at -8°C for different periods of time. The maximal preservation time was investigated. Twenty-four-hour sample data were collected and analyzed to compare supercooling preservation to conventional UW preservation. RESULTS: Our technique yielded a stable -8°C supercooling state. Cardiac graft revival was successfully achieved after supercooling preservation for 144 hours, and long-term survival was observed after supercooling preservation for 96 hours. Posttransplant outcomes, including myocardial ischemia-reperfusion injury, oxidative stress-related damage, and myocardial cell apoptosis, were improved in comparison to conventional 4°C UW preservation. CONCLUSIONS: Supercooling heart preservation at -8°C greatly prolonged the preservation time and improved the posttransplant outcomes in comparison to conventional 4°C UW preservation. Supercooling preservation is a promising technique for organ preservation.


Asunto(s)
Isquemia Fría , Trasplante de Corazón , Preservación de Órganos/métodos , Adenosina , Alopurinol , Animales , Glutatión , Insulina , Masculino , Ratones , Ratones Endogámicos C3H , Soluciones Preservantes de Órganos , Rafinosa , Factores de Tiempo
20.
J Reprod Immunol ; 137: 103075, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31918160

RESUMEN

Pregnancy is a kind of natural immune tolerance. Immune factors play an important role in recurrent spontaneous abortion and repeated implantation failure. Salvianolic acid B (SalB) has anti-tumor, anti-inflammatory, anti-oxidation and immunomodulatory functions. However, there are few reports on the relationship between SalB and maternal-fetal immune tolerance. In this study, CBA/J × DBA/2 J mice as a spontaneous abortion mouse model were given SalB. The results showed that the abortion rate was significantly decreased after SalB treatment. The populations of Nkp46 and cytotoxic CD8+ T cells in the placenta of female mice treated with SalB were significantly decreased. The qRT-PCR showed that SalB was able to significantly reduce the expression of pro-inflammatory factors and Toll-like Receptor in the placenta. In addition, SalB was able to increase the area of the labyrinth in the placenta. In conclusion, these findings suggest that SalB is beneficial for the immune-modulation at the maternal-fetal interface in a spontaneous abortion mouse model, resulting in a decrease in the abortion rate. This may encourage new ideas for the treatment of patients with repeated implantation failure.


Asunto(s)
Aborto Habitual/tratamiento farmacológico , Aborto Espontáneo/tratamiento farmacológico , Benzofuranos/administración & dosificación , Implantación del Embrión/efectos de los fármacos , Factores Inmunológicos/administración & dosificación , Aborto Habitual/inmunología , Aborto Habitual/patología , Aborto Espontáneo/inmunología , Aborto Espontáneo/patología , Animales , Vellosidades Coriónicas/efectos de los fármacos , Vellosidades Coriónicas/inmunología , Vellosidades Coriónicas/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Implantación del Embrión/inmunología , Femenino , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/genética , Mediadores de Inflamación/metabolismo , Masculino , Intercambio Materno-Fetal/efectos de los fármacos , Intercambio Materno-Fetal/inmunología , Ratones , Embarazo , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...