Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 36
Filtrer
1.
Cancer Immunol Res ; 12(5): 631-643, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38407902

RÉSUMÉ

Chimeric antigen receptor (CAR) T cells are emerging as an effective antitumoral therapy. However, their therapeutic effects on solid tumors are limited because of their short survival time and the immunosuppressive tumor microenvironment. Memory T cells respond more vigorously and persist longer than their naïve/effector counterparts. Therefore, promoting CAR T-cell development into memory T cells could further enhance their antitumoral effects. HI-TOPK-032 is a T-LAK cell-originated protein kinase (TOPK)-specific inhibitor that moderately represses some types of tumors. However, it is unknown whether HI-TOPK-032 works on hepatocellular carcinoma (HCC) and whether it impacts antitumoral immunity. Using both subcutaneous and orthotopic xenograft tumor models of two human HCC cell lines, Huh-7 and HepG2, we found that HI-TOPK-032 significantly improved proliferation/persistence of CD8+ CAR T cells, as evidenced by an increase in CAR T-cell counts or frequency of Ki-67+CD8+ cells and a decrease in PD-1+LAG-3+TIM-3+CD8+ CAR T cells in vivo. Although HI-TOPK-032 did not significantly suppress HCC growth, it enhanced the capacity of CAR T cells to inhibit tumor growth. Moreover, HI-TOPK-032 augmented central memory CD8+ T cell (TCM) frequency while increasing eomesodermin expression in CD8+ CAR T cells in tumor-bearing mice. Moreover, it augmented CD8+ CAR TCM cells in vitro and reduced their expression of immune checkpoint molecules. Finally, HI-TOPK-032 inhibited mTOR activation in CAR T cells in vitro and in tumors, whereas overactivation of mTOR reversed the effects of HI-TOPK-032 on CD8+ TCM cells and tumor growth. Thus, our studies have revealed mechanisms underlying the antitumoral effects of HI-TOPK-032 while advancing CAR T-cell immunotherapy.


Sujet(s)
Carcinome hépatocellulaire , Immunothérapie adoptive , Indolizine , Tumeurs du foie , Cellules T mémoire , Quinoxalines , Animaux , Humains , Souris , Carcinome hépatocellulaire/thérapie , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/anatomopathologie , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Immunothérapie adoptive/méthodes , Tumeurs du foie/immunologie , Tumeurs du foie/thérapie , Tumeurs du foie/anatomopathologie , Cellules T mémoire/effets des médicaments et des substances chimiques , Cellules T mémoire/immunologie , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Récepteurs chimériques pour l'antigène/immunologie , Microenvironnement tumoral/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe , Indolizine/pharmacologie , Indolizine/usage thérapeutique , Quinoxalines/pharmacologie , Quinoxalines/usage thérapeutique
2.
Biochem Pharmacol ; 219: 115979, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-38081367

RÉSUMÉ

Methotrexate (MTX) is an immunosuppressant used to treat autoimmune diseases, including psoriasis. However, like other immunosuppressants, MTX alone does not prevent their recurrence. Electrostimulation (ES) has been utilized to treat some inflammatory disorders without any major side-effect. But it remains unknown if ES alone, or together with MTX, ameliorates autoimmune disease relapse: a sticky medical problem. In particular, the mechanisms underlying ES action remain unclear. The objective of this study was to determine an impact of ES and/or MTX on psoriasis relapse and their potential cooperation. We found that regional ES, but not MTX, ameliorated psoriasiform skin inflammation recurrence. Interestingly, treatment with both MTX and ES further prevented psoriasis recurrence compared to ES alone. Moreover, ES downregulated potassium channel Kv1.3 on T-cells and reduced CD4+/CD8+ effector memory (TEM) and CD8+ skin-resident memory T (TRM) cells, while ES plus MTX further decreased CD8+ TEM/TRM cells compared to ES alone. However, ES failed to further attenuate psoriasis recurrence or suppress T cell memory in Kv1.3-deficient mice, whereas lack of Kv1.3 itself ameliorated psoriasis relapse by shrinking T cell memory pool. Importantly, ES moderately inhibited T-cell proliferation in vitro. ES also reduced human CD8+ TRM cells and attenuated human skin lesions in humanized mice grafted with lesional skin from patients with recurrent psoriasis, with an enhanced efficacy in mice treated with both ES and MTX. Thus, ES and MTX cooperated to prevent psoriasis relapse by reducing T-cell memory via targeting potassium channel Kv1.3. Our studies may be implicated for treating human psoriasis.


Sujet(s)
Électrothérapie , Psoriasis , Humains , Animaux , Souris , Méthotrexate/pharmacologie , Méthotrexate/usage thérapeutique , Cellules T mémoire , Psoriasis/traitement médicamenteux , Peau , Maladie chronique , Inflammation/anatomopathologie , Canaux potassiques
3.
Front Pharmacol ; 14: 1285799, 2023.
Article de Anglais | MEDLINE | ID: mdl-38027010

RÉSUMÉ

Background: Ferroptosis is an emerging type of regulated cell death and associated with antitumoral therapy, while some microRNAs have been shown to regulate the tumorigenesis and cancer progression. Meanwhile, polyphyllin I (PPI) has exhibited antitumoral effects by promoting cancer cell apoptosis and ferroptosis. However, it is unclear whether PPI induces cancer cell ferroptosis by regulating microRNAs. Methods: We used two gastric cancer cell lines (AGS and MKN-45) to set up a tumor model of the nude mice, which were then treated daily with PPI to measure the cancer growth in vitro and in vivo. Ferroptosis was measured using immunofluorescence staining and flow cytometric analysis according to levels of intracellular ROS, lipid ROS and ferrous ions. Moreover, NRF2 expression was measured by Western blotting. In some experiments, the mimics or inhibitors of miR-124-3p were used to further confirm its involvement in PPI-induced cancer cell ferroptosis. Results: Here we found that miR-124-3p mediated cancer ferroptosis and tumor repression induced by PPI since PPI increased miR-124-3p expression in gastric cancer cells and promoted their ferroptosis, whereas inhibition of miR-124-3p mostly abolished the effects of PPI on tumor growth, ferroptosis and NRF2 expression. Moreover, miR-124-3p mimics promoted cancer cell ferroptosis by downregulating NRF2 through directly targeting 3'-UTR region of NRF2, confirming a role for miR-124-3p in regulating PPI-induced ferroptosis. Conclusion: PPI exerts its antitumoral effects on the gastric cancer by promoting cell ferroptosis via regulating miR-124-3p. Our findings have clinical implications for cancer chemotherapy.

5.
Front Pharmacol ; 14: 1145407, 2023.
Article de Anglais | MEDLINE | ID: mdl-37081971

RÉSUMÉ

Background: Ferroptosis is a new form of regulated cell death characterized by the accumulation of iron-dependent lipid peroxides and membrane damages. Recent studies have identified an important role for cancer cell ferroptosis in antitumor therapy. On the other hand, polyphyllin I (PPI) has been reported to exert antitumor effects on some types of cancers. However, it remains unknown whether or not PPI regulates cancer cell ferroptosis. Methods: Two types of human gastric cancer cells (AGS and MKN-45) were used to establish tumor xenograft models in nude mice that were treated with polyphyllin I (PPI) to observe tumor growth, while cells also were cultured for in vitro studies. Ferroptosis, based on the intracellular ROS/lipid ROS production and accumulation of ferrous ions, was detected using a fluorescence microscope and flow cytometer, while the expression of NRF2/FTH1 was measured using Western blotting assays. Results: Here we found that PPI inhibited the gastric cancer growth in vivo and in vitro while increasing the intracellular reactive oxygen species (ROS)/lipid peroxides and ferrous ions in the gastric cancer cells. PPI also decreased the levels of nuclear factor erythroid 2-related factor 2 (NRF2) and ferritin heavy chain 1 (FTH1) in gastric cancer cells in vitro. Moreover, liproxstain-1, an inhibitor of cell ferroptosis, mostly reversed the cell ferroptosis and tumor growth arrest induced by PPI. Finally, the effects of PPI on cancer cell ferroptosis were diminished by the overexpression of NRF2. Conclusion: For the first time, our results have demonstrated that PPI exerts its antitumor activity on the gastric cancer by, at least partially, inducing cancer cell ferroptosis via regulating NRF2/FTH1 pathway. These findings may be implicated for clinical replacement therapy of the gastric cancer.

7.
Front Immunol ; 12: 683249, 2021.
Article de Anglais | MEDLINE | ID: mdl-34290705

RÉSUMÉ

Macrophages, a major subset of innate immune cells, are main infiltrating cells in the kidney in lupus nephritis. Macrophages with different phenotypes exert diverse or even opposite effects on the development of lupus nephritis. Substantial evidence has shown that macrophage M2 polarization is beneficial to individuals with chronic kidney disease. Further, it has been reported that PD-1 ligands (PD-Ls) contribute to M2 polarization of macrophages and their immunosuppressive effects. Total glucosides of paeony (TGP), originally extracted from Radix Paeoniae Alba, has been approved in China to treat some autoimmune diseases. Here, we investigated the potentially therapeutic effects of TGP on lupus nephritis in a pristane-induced murine model and explored the molecular mechanisms regulating macrophage phenotypes. We found that TGP treatment significantly improved renal function by decreasing the urinary protein and serum creatinine, reducing serum anti-ds-DNA level and ameliorating renal immunopathology. TGP increased the frequency of splenic and peritoneal F4/80+CD11b+CD206+ M2-like macrophages with no any significant effect on F4/80+CD11b+CD86+ M1-like macrophages. Immunofluorescence double-stainings of the renal tissue showed that TGP treatment increased the frequency of F4/80+Arg1+ subset while decreasing the percentage of F4/80+iNOS+ subset. Importantly, TGP treatment increased the percentage of both F4/80+CD11b+PD-L1+ and F4/80+CD11b+PD-L2+ subsets in spleen and peritoneal lavage fluid as well as the kidney. Furthermore, TGP augmented the expressions of CD206, PD-L2 and phosphorylated STAT6 in IL-4-treated Raw264.7 macrophages in vitro while its effects on PD-L2 were abolished by pretreatment of the cells with an inhibitor of STAT6, AS1517499. However, TGP treatment did not affect the expressions of STAT1 and PD-L1 in Raw264.7 macrophages treated with LPS/IFN-γ in vitro, indicating a possibly indirect effect of TGP on PD-L1 expression on macrophages in vivo. Thus, for the first time, we demonstrated that TGP may be a potent drug to treat lupus nephritis by inducing F4/80+CD11b+CD206+ and F4/80+CD11b+PD-L2+ macrophages through IL-4/STAT6/PD-L2 signaling pathway.


Sujet(s)
Antigène CD274/métabolisme , Glucosides/pharmacologie , Glomérulonéphrite lupique/étiologie , Glomérulonéphrite lupique/métabolisme , Paeonia/composition chimique , Transduction du signal/effets des médicaments et des substances chimiques , Terpènes/effets indésirables , Animaux , Marqueurs biologiques , Lignée cellulaire , Prédisposition aux maladies , Femelle , Glucosides/composition chimique , Humains , Interleukine-4/métabolisme , Glomérulonéphrite lupique/diagnostic , Activation des macrophages , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/métabolisme , Souris , Ligand-2 de la protéine-1 de mort cellulaire programmée/métabolisme , Facteur de transcription STAT-6/métabolisme
8.
Front Immunol ; 12: 646831, 2021.
Article de Anglais | MEDLINE | ID: mdl-33643325

RÉSUMÉ

Emerging evidence has linked the gut microbiota dysbiosis to transplant rejection while memory T-cells pose a threat to long-term transplant survival. However, it's unclear if the gut microbiome alters the formation and function of alloreactive memory T-cells. Here we studied the effects of berberine, a narrow-spectrum antibiotic that is barely absorbed when orally administered, on the gut microbiota, memory T-cells, and allograft survival. In this study, C57BL/6 mice transplanted with islets or a heart from BALB/c mice were treated orally with berberine. Allograft survival was observed, while spleen, and lymph node T-cells from recipient mice were analyzed using a flow cytometer. High-throughput sequencing and qPCR were performed to analyze the gut microbiota. CD8+ T-cells from recipients were cultured with the bacteria to determine potential T-cell memory cross-reactivity to a specific pathogen. We found that berberine suppressed islet allograft rejection, reduced effector CD8+CD44highCD62Llow and central memory CD8+CD44highCD62Lhigh T-cells (TCM), altered the gut microbiota composition and specifically lowered Bacillus cereus abundance. Further, berberine promoted long-term islet allograft survival induced by conventional costimulatory blockade and induced cardiac allograft tolerance as well. Re-colonization of B. cereus upregulated CD8+ TCM cells and reversed long-term islet allograft survival induced by berberine plus the conventional costimulatory blockade. Finally, alloantigen-experienced memory CD8+ T-cells from transplanted recipients rapidly responded to B. cereus in vitro. Thus, berberine prolonged allograft survival by repressing CD8+ TCM through regulating the gut microbiota. We have provided the first evidence that donor-specific memory T-cell generation is linked to a specific microbe and uncovered a novel mechanism underlying the therapeutic effects of berberine. This study may be implicated for suppressing human transplant rejection since berberine is already used in clinic to treat intestinal infections.


Sujet(s)
Berbérine/pharmacologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Survie du greffon/effets des médicaments et des substances chimiques , Tolérance immunitaire/effets des médicaments et des substances chimiques , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/immunologie , Bactéries/classification , Bactéries/génétique , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Microbiome gastro-intestinal/génétique , Microbiome gastro-intestinal/immunologie , Survie du greffon/immunologie , Transplantation cardiaque/méthodes , Tolérance immunitaire/immunologie , Interféron gamma/immunologie , Interféron gamma/métabolisme , Transplantation d'ilots de Langerhans/méthodes , Mâle , Souris de lignée BALB C , Souris de lignée C3H , Souris de lignée C57BL , ARN ribosomique 16S/génétique , Transplantation homologue
9.
Biochem Pharmacol ; 185: 114434, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33513343

RÉSUMÉ

Treg cells are essential for re-establishing self-tolerance in lupus. However, given that direct Treg therapies may be inadequate to control autoimmunity and inflammation, a strategy of inducing or expanding endogenous Treg cells in vivo may be a good option. Macrophages are main tissue-infiltrating cells and play a role in promoting Treg differentiation while paeoniflorin (PF), a monoterpene glycoside, exhibits anti-inflammatory and immunoregulatory effects. Here, we studied the effects of PF on CD4+FoxP3+ Treg frequency and the potential mechanisms involving M2 macrophages. We demonstrated that PF ameliorated lupus nephritis in lupus-prone B6/gld mice by reducing urinary protein, serum creatinine and anti-dsDNA levels, diminishing renal cellular infiltration, improving renal immunopathology and downregulating renal gene and protein expressions of key cytokines, including IFN-γ, IL-6, IL-12 and IL-23. PF also lowered the percentage of CD44highCD62Llow effector T cells while augmenting CD4+FoxP3+ Treg frequency in B6/gld mice. Importantly, PF increased TNFR2 expression on CD4+FoxP3+ Tregs, but not CD4+FoxP3- T cells, in vivo and in vitro. Furthermore, we found that CD206+ subset of F4/80+CD11b+ macrophages expressed a higher level of mTNF-α than their CD206- counterparts while PF increased mTNF-α expression on CD206+ macrophages in vitro and in vivo. In vitro studies showed that mTNF-α+ M2 macrophages were more potent in inducing Treg differentiation and proliferation than their mTNF-α- counterparts, whereas the effects of mTNF-α+ M2 macrophages were largely reversed by separation of M2 macrophages using a transwell or TNFR2-blocking Ab in the culture. Finally, PF also promoted in vitro Treg generation induced by M2 macrophages. Thus, we demonstrated that mTNFα-TNFR2 interaction is a new mechanism responsible for Treg differentiation mediated by M2 macrophages. We provided the first evidence that PF may be used to treat lupus nephritis.


Sujet(s)
Facteurs de transcription Forkhead/métabolisme , Glucosides/usage thérapeutique , Glomérulonéphrite lupique/métabolisme , Monoterpènes/usage thérapeutique , Récepteur au facteur de nécrose tumorale de type II/métabolisme , Lymphocytes T régulateurs/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Animaux , Anti-inflammatoires non stéroïdiens/pharmacologie , Anti-inflammatoires non stéroïdiens/usage thérapeutique , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/métabolisme , Femelle , Glucosides/pharmacologie , Glomérulonéphrite lupique/traitement médicamenteux , Souris , Souris de lignée C57BL , Souris transgéniques , Monoterpènes/pharmacologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques
10.
Oncoimmunology ; 9(1): 1829346, 2020 10 24.
Article de Anglais | MEDLINE | ID: mdl-33150044

RÉSUMÉ

CD4+Foxp3+ regulatory T cells (Tregs) in the tumor microenvironment restrain antitumor immunity, resulting in tumor aggression and poor survival in hepatocellular carcinoma (HCC). CD8+CD122+ Tregs have been previously shown to be more potent in immunosuppression than are CD4+Foxp3+ Tregs. Previous studies have demonstrated that resveratrol exerts its anti-cancer effects by downregulating CD4+Foxp3+ and M2-like macrophages, two key immunoregulatory cells that maintain the immunosuppressive tumor microenvironment. In this study, we found that resveratrol inhibited the tumor growth in a subcutaneous Hepa1-6 HCC model and decreased the frequency of CD8+CD122+ Tregs in the tumor as well as lymph nodes and spleen of the tumor-bearing mice. It also increased the percentage of IFN-γ-expressing CD8+ T cells in the tumor and peripheral lymphoid organs. The antitumor effects of resveratrol were partially reversed by the adoptive transfer of exogenous CD8+CD122+ Tregs into the tumor-bearing mice. Meanwhile, resveratrol treatment downregulated immunosuppressive cytokines, including TGF-ß1 and interleukin-10, in the tumor while elevating antitumor cytokines, TNF-α and IFN-γ. It also inhibited the activation of STAT3 signaling in the tumor. As expected, resveratrol reduced the percentage of M2-like macrophages in the mice. Importantly, resveratrol suppressed orthotopic H22 tumor growth and decreased the frequency of CD8+CD122+ Tregs and M2-like macrophages in the tumor-bearing mice. Furthermore, our studies showed that resveratrol, at non-cytotoxic concentrations, inhibited CD8+CD122+ Treg differentiation from CD8+CD122- T cells in vitro. Thus, our studies unveiled a new immune mechanism underlying the immunosuppressive tumor microenvironment and demonstrated that resveratrol could help reverse it by diminishing CD8+CD122+ Tregs.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Animaux , Lymphocytes T CD8+ , Carcinome hépatocellulaire/traitement médicamenteux , Tumeurs du foie/traitement médicamenteux , Souris , Resvératrol/pharmacologie , Lymphocytes T régulateurs , Microenvironnement tumoral
11.
Theranostics ; 10(23): 10466-10482, 2020.
Article de Anglais | MEDLINE | ID: mdl-32929360

RÉSUMÉ

Conventional immunosuppressants cause side effects and do not prevent the recurrence of autoimmune diseases. Moreover, they may not inhibit autoimmunity mediated by pathogenic memory T-cells. Dihydroartemisinin (DHA) has been shown to regulate autoimmunity. However, it remains unknown whether DHA impacts psoriasis and its recurrence. The objective of this study was to determine therapeutic effects of DHA on psoriasis and its relapse as well as its underlying mechanisms. Methods: We established animal models of imiquimod (IMQ)-induced psoriasis-like wild-type mice and humanized NSG mice receiving lesional human skin from patients with psoriasis. Many immunoassays, including immunohistochemistry, flow cytometry, quantitative RT-PCR and Western blotting, were performed. Results: We found that DHA not only ameliorated acute skin lesion of psoriatic mice, but also alleviated its recurrence by diminishing CD8+ central memory T (TCM) and CD8+ resident memory T (TRM) cells. It attenuated epidermal pathology and T-cell infiltration in the skin of IMQ-induced psoriatic mice while suppressing expression of IL-15, IL-17 and other proinflammatory cytokines in the skin. Surprisingly, DHA reduced the frequency and number of CD8+, but not CD4+, subset of CD44highCD62Lhigh TCM in psoriatic mice, whereas methotrexate (MTX) lowered CD4+, but not CD8+, TCM frequency and number. Indeed, DHA, but not MTX, downregulated eomesodermin (EOMES) and BCL-6 expression in CD8+ T-cells. Furthermore, DHA, but not MTX, reduced the presence of CD8+CLA+, CD8+CD69+ or CD8+CD103+ TRM cells in mouse skin. Interestingly, treatment with DHA, but not MTX, during the first onset of psoriasis largely prevented psoriasis relapse induced by low doses of IMQ two weeks later. Administration of recombinant IL-15 or CD8+, but not CD4+, TCM cells resulted in complete recurrence of psoriasis in mice previously treated with DHA. Finally, we demonstrated that DHA alleviated psoriatic human skin lesions in humanized NSG mice grafted with lesional skin from psoriatic patients while reducing human CD8+ TCM and CD103+ TRM cells in humanized mice. Conclusion: We have provided the first evidence that DHA is advantageous over MTX in preventing psoriasis relapse by reducing memory CD8+ T-cells.


Sujet(s)
Artémisinines/pharmacologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Psoriasis/traitement médicamenteux , Animaux , Artémisinines/usage thérapeutique , Lymphocytes T CD8+/immunologie , Modèles animaux de maladie humaine , Évaluation préclinique de médicament , Humains , Imiquimod/administration et posologie , Imiquimod/immunologie , Mémoire immunologique/effets des médicaments et des substances chimiques , Interleukine-15/métabolisme , Interleukine-17/métabolisme , Mâle , Méthotrexate/pharmacologie , Méthotrexate/usage thérapeutique , Souris , Psoriasis/immunologie , Psoriasis/anatomopathologie , Récidive , Prévention secondaire/méthodes , Peau/effets des médicaments et des substances chimiques , Peau/immunologie , Peau/anatomopathologie , Transplantation de peau , Chimère obtenue par transplantation
12.
Biomed Pharmacother ; 121: 109570, 2020 Jan.
Article de Anglais | MEDLINE | ID: mdl-31710893

RÉSUMÉ

Traditional Chinese Medicine (TCM) has been traditionally used to treat patients with cancers in China. It not only alleviates the symptoms of tumor patients and improves their quality of life, but also controls the size of tumors and prolongs the survival of tumor patients. While some herbs of TCM may exert therapeutic effects by directly targeting cancer cells or reducing side effects caused by antitumor drugs, others can control tumor growth and metastasis via enhancing antitumor immunity. In particular, TCM can exert antitumor effects by upregulating immune responses even in immunosuppressive tumor microenvironment. For instance, it reduces the number of M2-type macrophages and Treg cells in the tumor tissue. Although extensive reviews on directly killing cancer cells by TCM have been conducted, a review of anticancer activity of TCM solely based on its immunity-enhancing capacity is unusual. This review will summarize research progress of antitumor TCM that regulates the immune system, including both innate immunity, such as macrophages, dendritic cells, natural killer cells and MDSCs, and adaptive immunity, including CD4+/CD8+ T lymphocytes, regulatory T cells (Tregs) and B cells. As cancer immunotherapy has recently achieved certain success, it is expected that the clinical applications of immunity-enhancing TCM or traditional medicine for treating various cancer patients will be expanded. Further studies on the mechanisms by which TCM regulates immunity will provide new insights into how TCM controls tumor growth and metastasis, and may help improve its therapeutic effects on various cancers in clinic.


Sujet(s)
Médicaments issus de plantes chinoises/administration et posologie , Médecine traditionnelle chinoise/méthodes , Tumeurs/traitement médicamenteux , Animaux , Antinéoplasiques d'origine végétale/administration et posologie , Antinéoplasiques d'origine végétale/pharmacologie , Médicaments issus de plantes chinoises/pharmacologie , Humains , Immunothérapie/méthodes , Tumeurs/immunologie , Qualité de vie , Microenvironnement tumoral/immunologie
13.
J Immunol ; 203(12): 3436-3446, 2019 12 15.
Article de Anglais | MEDLINE | ID: mdl-31732527

RÉSUMÉ

An allograft is rejected in the absence of any immunosuppressive treatment because of vigorous alloimmunity and thus requires extensive immunosuppression for its survival. Although there are many conventional immunosuppressants for clinical use, it is necessary to seek alternatives to existing drugs, especially in case of transplant patients with complicated conditions. Luteolin, a natural ingredient, exists in many plants. It exhibits multiple biological and pharmacological effects, including anti-inflammatory properties. In particular, luteolin has been shown to upregulate CD4+CD25+ regulatory T cells (Tregs) in the context of airway inflammation. However, it remains unknown whether luteolin regulates alloimmune responses. In this study, we demonstrated that luteolin significantly prolonged murine skin allograft survival, ameliorated cellular infiltration, and downregulated proinflammatory cytokine gene expression in skin allografts. Furthermore, luteolin increased the percentage of CD4+Foxp3+ Tregs while reducing frequency of mature dendritic cells and CD44highCD62Llow effector CD4+/CD8+ T cells posttransplantation. It also suppressed the proliferation of T cells and their production of cytokines IFN-γ and IL-17A in vitro while increasing IL-10 level in the supernatant. Moreover, luteolin promoted CD4+Foxp3+ Treg generation from CD4+CD25- T cells in vitro. Depleting Tregs largely, although not totally, reversed luteolin-mediated extension of allograft survival. More importantly, luteolin inhibited AKT/mTOR signaling in T cells. Thus, for the first time, to our knowledge, we found that luteolin is an emerging immunosuppressant as an mTOR inhibitor in allotransplantation. This finding could be important for the suppression of human allograft rejection, although it remains to be determined whether luteolin has an advantage over other conventional immunosuppressants in suppression of allograft rejection.


Sujet(s)
Allogreffes/effets des médicaments et des substances chimiques , Allogreffes/immunologie , Rejet du greffon/immunologie , Immunosuppression thérapeutique , Immunosuppresseurs/pharmacologie , Lutéoline/pharmacologie , Animaux , Cytokines/génétique , Cytokines/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Rejet du greffon/traitement médicamenteux , Rejet du greffon/prévention et contrôle , Survie du greffon/effets des médicaments et des substances chimiques , Survie du greffon/immunologie , Immunophénotypage , Inflammation/étiologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Médiateurs de l'inflammation/métabolisme , Numération des lymphocytes , Mâle , Souris , Transplantation de peau , Sous-populations de lymphocytes T/effets des médicaments et des substances chimiques , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme
14.
Front Immunol ; 10: 652, 2019.
Article de Anglais | MEDLINE | ID: mdl-30988670

RÉSUMÉ

A transplanted organ is usually rejected without any major immunosuppressive treatment because of vigorous alloimmune responsiveness. However, continuous global immunosuppression may cause severe side effects, including nephrotoxicity, tumors, and infections. Therefore, it is necessary to seek novel immunosuppressive agents, especially natural ingredients that may provide sufficient efficacy in immunosuppression with minimal side effects. Shikonin is a bioactive naphthoquinone pigment, an ingredient originally extracted from the root of Lithospermum erythrorhizon. Previous studies have shown that shikonin regulates immunity and exerts anti-inflammatory effects. In particular, it can ameliorate arthritis in animal models. However, it is unclear whether shikonin inhibits alloimmunity or allograft rejection. In this study and for the first time, we demonstrated that shikonin significantly prolonged the survival of skin allografts in wild-type mice. Shikonin increased the frequencies of CD4+Foxp3+ regulatory T cells (Tregs) post-transplantation and induced CD4+Foxp3+ Tregs in vitro as well. Importantly, depleting the Tregs abrogated the extension of skin allograft survival induced by shikonin. It also decreased the frequencies of CD8+CD44highCD62Llow effector T cells and CD11c+CD80+/CD11c+CD86+ mature DCs after transplantation. Moreover, we found that shikonin inhibited the proliferation of T cells in vitro and suppressed their mTOR signaling. It also reduced the gene expression of pro-inflammatory cytokines, including IFNγ, IL-6, TNFα, and IL-17A, while increasing the gene expression of anti-inflammatory mediators IL-10, TGF-ß1, and indoleamine-2, 3-dioxygenase (IDO) in skin allografts. Further, shikonin downregulated IDO protein expression in skin allografts and DCs in vitro. Taken together, shikonin inhibits allograft rejection via upregulating CD4+Foxp3+ Tregs. Thus, shikonin is a novel immunosuppressant that could be potentially used in clinical transplantation.


Sujet(s)
Survie du greffon/effets des médicaments et des substances chimiques , Immunosuppresseurs/pharmacologie , Naphtoquinones/pharmacologie , Transplantation de peau , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Allogreffes , Animaux , Cytokines/génétique , Cytokines/immunologie , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Facteurs de transcription Forkhead/immunologie , Indoleamine-pyrrole 2,3,-dioxygenase/immunologie , Souris de lignée BALB C , Souris de lignée C57BL , Peau/effets des médicaments et des substances chimiques , Peau/immunologie , Lymphocytes T régulateurs/immunologie
15.
Front Immunol ; 10: 306, 2019.
Article de Anglais | MEDLINE | ID: mdl-30863408

RÉSUMÉ

A transplanted organ is always rejected in the absence of any immunosuppressive treatment due to vigorous alloimmunity. However, continuously global immunosuppression with a conventional immunosuppressant may result in severe side effects, including nephrotoxicity, tumors and infections. Tregs have been widely used to inhibit allograft rejection, especially in animal models. However, it's well accepted that administration of Tregs alone is not satisfactory in immune-competent wild-type animals. Therefore, it's imperative to promote Treg therapies under the cover of other approaches, including costimulatory blockade. In the present study, we demonstrated that administration of in vitro-expanded CD8+CD122+PD-1+ Tregs synergized with costimulatory blockade of CD40/CD154, but not B7/CD28, to prolong skin allograft survival in wild-type mice and to reduce cellular infiltration in skin allografts as well. Treg treatment and blockade of CD40/CD154, but not B7/CD28, also exhibited an additive effect on suppression of T cell proliferation in vitro and pro-inflammatory cytokine expression in skin allografts. Importantly, blocking B7/CD28, but not CD40/CD154, costimulation decreased the number of transferred CD8+CD122+PD-1+ Tregs and their expression of IL-10 in recipient mice. Furthermore, it's B7/CD28, but not CD40/CD154, costimulatory blockade that dramatically reduced IL-10 production by CD8+CD122+PD-1+ Tregs in vitro, suggesting that B7/CD28, but not CD40/CD154, costimulation is critical for their production of IL-10. Indeed, infusion of IL-10-deficient CD8+CD122+PD-1+ Tregs failed to synergize with anti-CD154 Ab treatment to further prolong allograft survival. Our data may explain why blocking B7/CD28 costimulatory pathway does not boost IL-10-dependent Treg suppression of alloimmunity. Thus, these findings could be implicated in clinical organ transplantation.


Sujet(s)
Allogreffes/immunologie , Antigènes CD40/immunologie , Ligand de CD40/immunologie , Antigènes CD8/immunologie , Sous-unité bêta du récepteur à l'interleukine-2/immunologie , Récepteur-1 de mort cellulaire programmée/immunologie , Lymphocytes T régulateurs/immunologie , Allogreffes/métabolisme , Animaux , Antigènes B7/immunologie , Antigènes B7/métabolisme , Antigène CD28/immunologie , Antigène CD28/métabolisme , Antigènes CD40/métabolisme , Ligand de CD40/métabolisme , Antigènes CD8/métabolisme , Survie du greffon/immunologie , Sous-unité bêta du récepteur à l'interleukine-2/métabolisme , Mâle , Souris de lignée BALB C , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée/métabolisme , Transplantation de peau/méthodes , Lymphocytes T régulateurs/métabolisme , Immunologie en transplantation/immunologie , Transplantation homologue
16.
Front Immunol ; 9: 2359, 2018.
Article de Anglais | MEDLINE | ID: mdl-30369931

RÉSUMÉ

Cellular therapies with polyclonal regulatory T-cells (Tregs) in transplantation and autoimmune diseases have been carried out in both animal models and clinical trials. However, The use of large numbers of polyclonal Tregs with unknown antigen specificities has led to unwanted effects, such as systemic immunosuppression, which can be avoided via utilization of antigen-specific Tregs. Antigen-specific Tregs are also more potent in suppression than polyclonal ones. Although antigen-specific Tregs can be induced in vitro, these iTregs are usually contaminated with effector T cells during in vitro expansion. Fortunately, Tregs can be efficiently engineered with a predetermined antigen-specificity via transfection of viral vectors encoding specific T cell receptors (TCRs) or chimeric antigen receptors (CARs). Compared to Tregs engineered with TCRs (TCR-Tregs), CAR-modified Tregs (CAR-Tregs) engineered in a non-MHC restricted manner have the advantage of widespread applications, especially in transplantation and autoimmunity. CAR-Tregs also are less dependent on IL-2 than are TCR-Tregs. CAR-Tregs are promising given that they maintain stable phenotypes and functions, preferentially migrate to target sites, and exert more potent and specific immunosuppression than do polyclonal Tregs. However, there are some major hurdles that must be overcome before CAR-Tregs can be used in clinic. It is known that treatments with anti-tumor CAR-T cells cause side effects due to cytokine "storm" and neuronal cytotoxicity. It is unclear whether CAR-Tregs would also induce these adverse reactions. Moreover, antibodies specific for self- or allo-antigens must be characterized to construct antigen-specific CAR-Tregs. Selection of antigens targeted by CARs and development of specific antibodies are difficult in some disease models. Finally, CAR-Treg exhaustion may limit their efficacy in immunosuppression. Recently, innovative CAR-Treg therapies in animal models of transplantation and autoimmune diseases have been reported. In this mini-review, we have summarized recent progress of CAR-Tregs and discussed their potential applications for induction of immunological tolerance.


Sujet(s)
Tolérance immunitaire , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs chimériques pour l'antigène/métabolisme , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Animaux , Auto-immunité , Humains , Immunothérapie adoptive , Récepteurs aux antigènes des cellules T/génétique , Récepteurs chimériques pour l'antigène/génétique , Immunologie en transplantation
17.
Cell Physiol Biochem ; 50(4): 1560-1573, 2018.
Article de Anglais | MEDLINE | ID: mdl-30359968

RÉSUMÉ

BACKGROUND/AIMS: Lupus nephritis (LN) is an autoimmune glomerulonephritis that frequently develops secondary to systemic lupus erythematosus. Patients with LN require extensive treatments with global immunosuppressive agents. However, long-term treatment with conventional immunosuppressants may cause various side effects. Therefore, it's important to seek alternative drugs for treating LN. Here we aimed to investigate the immunoregulatory effects of mangiferin (MG), an ingredient that was originally extracted from natural herbs, including Mangifera Indica Linn. and Rhizoma Anemarrhenae. METHODS: FasL-deficient B6/ gld mice were used as a spontaneous LN model. The serum anti-dsDNA Ab and creatinine levels were analyzed via ELISA. Renal histology and immunopathology were determined using H&E and PAS staining, immunofluorescence (IgG and C3), and IHC staining (CD3 and a-SMA). Cytokine gene expression was measured by RT-PCR assays while effector T cells and Tregs were enumerated by flow analysis. Finally, the proliferation and apoptosis of T cells were measured by CFSE staining and flow analysis while their mTOR signaling was detected through Western blotting. RESULTS: We found that administration of MG ameliorated LN in lupus-prone B6/gld mice by reducing the urinary protein and serum creatinine levels, diminishing T cell infiltration in kidneys and improving renal immunopathology. MG also significantly lowered the percentages of CD44highCD62Llow effector T cells in B6/gld mice. Importantly, treatments with MG augmented CD4+FoxP3+ Treg frequencies in spleens, lymph nodes and kidneys of B6/gld mice. It also induced CD4+FoxP3+ Tregs from CD3+ T cells in vitro and promoted Treg proliferation. Furthermore, it inhibited CD3+ T cell proliferation in vitro and suppressed their phosphorylation of mTOR and its downstream P70S6K. However, MG did not promote T cell apoptosis, implying that it is not cytotoxic. Depletion of CD4+CD25+FoxP3+ Tregs in B6/gld mice abrogated its therapeutic effects on LN. CONCLUSION: MG exerts a novel therapeutic effect on murine LN via upregulating CD4+FoxP3+ Tregs, downregulating mTOR/p70S6K pathway and improving renal immunopathology. It may be useful for treating LN in clinic.


Sujet(s)
Glomérulonéphrite lupique/traitement médicamenteux , Lymphocytes T régulateurs/immunologie , Xanthones/usage thérapeutique , Animaux , Antigènes CD4/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Créatinine/sang , Cytokines/métabolisme , Modèles animaux de maladie humaine , Facteurs de transcription Forkhead/métabolisme , Rein/immunologie , Rein/métabolisme , Rein/anatomopathologie , Glomérulonéphrite lupique/métabolisme , Glomérulonéphrite lupique/anatomopathologie , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Souris knockout , Phosphorylation/effets des médicaments et des substances chimiques , Ribosomal Protein S6 Kinases, 70-kDa/métabolisme , Transduction du signal , Lymphocytes T régulateurs/cytologie , Lymphocytes T régulateurs/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Xanthones/pharmacologie
18.
Front Immunol ; 9: 2092, 2018.
Article de Anglais | MEDLINE | ID: mdl-30258447

RÉSUMÉ

Psoriasis is an autoimmune and inflammatory skin disease affecting around 2-3% of the world's population. Patients with psoriasis need extensive treatments with global immunosuppressive agents that may cause severe side effects. Esculetin, a type of coumarins, is an active ingredient extracted mainly from the bark of Fraxinus rhynchophylla, which has been used to treat inflammatory and autoimmune diseases in China. However, the antipsoriatic effects of esculetin have not been reported. In this study, we aimed to investigate the effects of esculetin on psoriatic skin inflammation in a mouse model and explored the potential molecular mechanisms underlying its action. We found that esculetin ameliorated the skin lesion and reduced PASI scores as well as weight loss in imiquimod-induced psoriasis-like mice, accompanied with weakened proliferation and differentiation of keratinocytes and T cell infiltration in esculetin-treated psoriatic mice. In addition, esculetin reduced the frequency of CD8+CD44highCD62Llow effector T cells in psoriatic mice. In contrast, it increased the frequency of CD4+Foxp3+ Tregs in both lymph nodes and spleens of the psoriatic mice while promoting the differentiation of CD4+CD25- T cells into CD4+Foxp3+ Tregs in vitro. Interestingly, depleting CD4+Foxp3+ Tregs largely reversed esculetin-mediated reduction in PASI scores, indicating that esculetin attenuates murine psoriasis mainly by inducing CD4+Foxp3+ Tregs. Furthermore, the mRNA levels of proinflammatory cytokines in the psoriatic mouse skin, including IL-6, IL-17A, IL-22, IL-23, TNF-α, and IFN-γ, were dramatically decreased by the treatment with esculetin. Finally, we found that esculetin inhibited the phosphorylation of IKKα and P65 in the psoriatic skin, suggesting that it inhibits the activation of NF-κB signaling. Thus, we have demonstrated that esculetin attenuates psoriasis-like skin lesion in mice and may be a potential therapeutic candidate for the treatment of psoriasis in clinic.


Sujet(s)
Cytokines/immunologie , Psoriasis , Ombelliférones/pharmacologie , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/anatomopathologie , Imiquimod/effets indésirables , Imiquimod/pharmacologie , Souris , Souris de lignée BALB C , Psoriasis/induit chimiquement , Psoriasis/traitement médicamenteux , Psoriasis/immunologie , Psoriasis/anatomopathologie , Lymphocytes T régulateurs
19.
Int J Health Plann Manage ; 32(3): 307-316, 2017 Jul.
Article de Anglais | MEDLINE | ID: mdl-28707707

RÉSUMÉ

Currently, China has been experiencing rapid growth of medical costs, serious waste of medical resources, increasing disease burden for residents, and a medical insurance fund deficit. Therefore, an urgent problem that needs to be solved is to choose a rational payment for the insurance system. To empirically evaluate the long-term effects of capitation reform in a New Rural Cooperative Medical Scheme in Pudong New Area, we collected and analysed data regarding financing, fund operation, medical service cost, and medical care-seeking behaviour from 2011 to 2015, a duration that includes data before and after reform. The data for financing and behaviours were compared year by year, and the monthly data for inpatient and outpatient costs were evaluated in a retrospective time series study. The capitation reform in Pudong New Area showed strong evidence of the power of medical cost control in the long run, while it was weak in reversing the number of patients flowing into secondary and tertiary hospitals. To make the payment of capitation play a bigger role in cost control in China, a tighter alignment of capitation with the general practitioner system and achieving dual referral is critical for future studies.


Sujet(s)
Rémunération par capitation/organisation et administration , Réforme des soins de santé/organisation et administration , Services de santé ruraux/organisation et administration , Soins ambulatoires/économie , Chine , Maîtrise des coûts/économie , Maîtrise des coûts/organisation et administration , Coûts des soins de santé , Réforme des soins de santé/économie , Financement des soins de santé , Hospitalisation/économie , Humains , Études longitudinales , Acceptation des soins par les patients/statistiques et données numériques , Services de santé ruraux/économie
20.
Adv Clin Exp Med ; 26(9): 1431-1435, 2017 Dec.
Article de Anglais | MEDLINE | ID: mdl-29442466

RÉSUMÉ

BACKGROUND: Endometriosis (EM) interferes with the reproductive process and affects the success rate of in vitro fertilization (IVF). Inflammatory cytokines are suggested to play a role in infertility in patients with EM. OBJECTIVES: In this study, we aimed to investigate the relationship between resistin and interleukin 23 (IL-23) levels in follicular fluid (FF) and serum together with the severity of endometriosis and in vitro fertilization/ embryo transfer (IVF-ET) outcome. MATERIAL AND METHODS: Samples from 116 infertile women were studied using enzyme-linked immunosorbent assay (ELISA). The study group consisted of 76 infertile patients diagnosed with EM (40 with stages I-II and 36 with stages III-IV) undergoing IVF-ET. The control group included 40 women with tubal factor infertility. FF and serum samples were collected on the day of follicle aspiration and hCG administration, respectively. RESULTS: The serum and FF resistin levels were significantly higher in the EM group than in the control group (p-value <0.05). The FF resistin and IL-23 levels were significantly higher in EM stages III-IV than in stages I-II (p-value <0.05), and the serum resistin and IL-23 levels were also significantly (p-value <0.01) higher in stages III-IV than in stages I-II. The E2 level on the day of hCG administration and the implantation rate were both significantly lower in the EM group than in the control group. However, there were no differences in the Gn duration and dose, and the cleavage, implantation and clinical pregnancy rates between the 2 groups. CONCLUSIONS: Our results suggest that patients with EM exhibit increased resistin level in FF and serum. Advanced EM may contribute to infertility via decreased embryo implantation rates because of inflammation and immune rejection. No influence was observed on pregnancy outcomes after IVF-ET.


Sujet(s)
Transfert d'embryon , Endométriose/métabolisme , Fécondation in vitro , Liquide folliculaire/composition chimique , Infertilité féminine/métabolisme , Interleukine-23/analyse , Résistine/analyse , Adulte , Femelle , Humains , Interleukine-23/sang , Résistine/sang , Études rétrospectives
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...