Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
1.
J Am Heart Assoc ; 13(4): e030054, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38348774

ABSTRACT

BACKGROUND: This study investigated whether gCTRP9 (globular C1q/tumor necrosis factor-related protein-9) could restore high-glucose (HG)-suppressed endothelial progenitor cell (EPC) functions by activating the endothelial nitric oxide synthase (eNOS). METHODS AND RESULTS: EPCs were treated with HG (25 mmol/L) and gCTRP9. Migration, adhesion, and tube formation assays were performed. Adiponectin receptor 1, adiponectin receptor 2, and N-cadherin expression and AMP-activated protein kinase, protein kinase B, and eNOS phosphorylation were measured by Western blotting. eNOS activity was determined using nitrite production measurement. In vivo reendothelialization and EPC homing assays were performed using Evans blue and immunofluorescence in mice. Treatment with gCTRP9 at physiological levels enhanced migration, adhesion, and tube formation of EPCs. gCTRP9 upregulated the phosphorylation of AMP-activated protein kinase, protein kinase B, and eNOS and increased nitrite production in a concentration-dependent manner. Exposure of EPCs to HG-attenuated EPC functions induced cellular senescence and decreased eNOS activity and nitric oxide synthesis; the effects of HG were reversed by gCTRP9. Protein kinase B knockdown inhibited eNOS phosphorylation but did not affect gCTRP9-induced AMP-activated protein kinase phosphorylation. HG impaired N-cadherin expression, but treatment with gCTRP9 restored N-cadherin expression after HG stimulation. gCTRP9 restored HG-impaired EPC functions through both adiponectin receptor 1 and N-cadherin-mediated AMP-activated protein kinase /protein kinase B/eNOS signaling. Nude mice that received EPCs treated with gCTRP9 under HG medium showed a significant enhancement of the reendothelialization capacity compared with those with EPCs incubated under HG conditions. CONCLUSIONS: CTRP9 promotes EPC migration, adhesion, and tube formation and restores these functions under HG conditions through eNOS-mediated signaling mechanisms. Therefore, CTRP9 modulation could eventually be used for vascular healing after injury.


Subject(s)
Adiponectin , Endothelial Progenitor Cells , Glycoproteins , Proto-Oncogene Proteins c-akt , Mice , Animals , Proto-Oncogene Proteins c-akt/metabolism , Endothelial Progenitor Cells/metabolism , Complement C1q/metabolism , Complement C1q/pharmacology , AMP-Activated Protein Kinases/metabolism , Cytokines/metabolism , Nitric Oxide Synthase Type III/metabolism , Mice, Nude , Receptors, Adiponectin/metabolism , Nitrites , Cell Movement , Glucose/pharmacology , Glucose/metabolism , Cadherins/metabolism , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology , Nitric Oxide/metabolism , Cells, Cultured
2.
Chin J Integr Med ; 30(4): 322-329, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37861963

ABSTRACT

OBJECTIVE: To investigate the mechanistic basis for the anti-proliferation and anti-invasion effect of tumor necrosis factor-related apoptosis-induced ligand (TRAIL) and celastrol combination treatment (TCCT) in glioblastoma cells. METHODS: Cell counting kit-8 was used to detect the effects of different concentrations of celastrol (0-16 µmol/L) and TRAIL (0-500 ng/mL) on the cell viability of glioblastoma cells. U87 cells were randomly divided into 4 groups, namely control, TRAIL (TRAIL 100 ng/mL), Cel (celastrol 0.5 µmol/L) and TCCT (TRAIL 100 ng/mL+ celastrol 0.5 µmol/L). Cell proliferation, migration, and invasion were detected by colony formation, wound healing, and Transwell assays, respectively. Quantitative reverse transcription polymerase chain reaction and Western blotting were performed to assess the levels of epithelial-mesenchymal transition (EMT) markers (zona occludens, N-cadherin, vimentin, zinc finger E-box-binding homeobox, Slug, and ß-catenin). Wnt pathway was activated by lithium chloride (LiCl, 20 mol/L) and the mechanism for action of TCCT was explored. RESULTS: Celastrol and TRAIL synergistically inhibited the proliferation, migration, invasion, and EMT of U87 cells (P<0.01). TCCT up-regulated the expression of GSK-3ß and down-regulated the expression of ß-catenin and its associated proteins (P<0.05 or P<0.01), including c-Myc, Cyclin-D1, and matrix metalloproteinase (MMP)-2. In addition, LiCl, an activator of the Wnt signaling pathway, restored the inhibitory effects of TCCT on the expression of ß-catenin and its downstream genes, as well as the migration and invasion of glioblastoma cells (P<0.05 or P<0.01). CONCLUSIONS: Celastrol and TRAIL can synergistically suppress glioblastoma cell migration, invasion, and EMT, potentially through inhibition of Wnt/ß-catenin pathway. This underlies a novel mechanism of action for TCCT as an effective therapy for glioblastoma.


Subject(s)
Glioblastoma , Pentacyclic Triterpenes , Wnt Signaling Pathway , Humans , Glioblastoma/drug therapy , Glioblastoma/pathology , beta Catenin/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Ligands , Cell Line, Tumor , Apoptosis , Tumor Necrosis Factors/pharmacology , Cell Proliferation , Cell Movement , Epithelial-Mesenchymal Transition
3.
Cancer Biol Med ; 20(11)2023 11 02.
Article in English | MEDLINE | ID: mdl-37921408

ABSTRACT

OBJECTIVE: Immature vasculature lacking pericyte coverage substantially contributes to tumor growth, drug resistance, and cancer cell dissemination. We previously demonstrated that tumor necrosis factor superfamily 15 (TNFSF15) is a cytokine with important roles in modulating hematopoiesis and vascular homeostasis. The main purpose of this study was to explore whether TNFSF15 might promote freshly isolated myeloid cells to differentiate into CD11b+ cells and further into pericytes. METHODS: A model of Lewis lung cancer was established in mice with red fluorescent bone marrow. After TNFSF15 treatment, CD11b+ myeloid cells and vascular pericytes in the tumors, and the co-localization of pericytes and vascular endothelial cells, were assessed. Additionally, CD11b+ cells were isolated from wild-type mice and treated with TNFSF15 to determine the effects on the differentiation of these cells. RESULTS: We observed elevated percentages of bone marrow-derived CD11b+ myeloid cells and vascular pericytes in TNFSF15-treated tumors, and the latter cells co-localized with vascular endothelial cells. TNFSF15 protected against CD11b+ cell apoptosis and facilitated the differentiation of these cells into pericytes by down-regulating Wnt3a-VEGFR1 and up-regulating CD49e-FN signaling pathways. CONCLUSIONS: TNFSF15 facilitates the production of CD11b+ cells in the bone marrow and promotes the differentiation of these cells into pericytes, which may stabilize the tumor neovasculature.


Subject(s)
Neoplasms , Pericytes , Animals , Humans , Mice , Cell Differentiation , Endothelial Cells , Mice, Inbred C57BL , Myeloid Cells/metabolism , Neoplasms/metabolism , Pericytes/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/pharmacology , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology
4.
Am J Chin Med ; 51(7): 1627-1651, 2023.
Article in English | MEDLINE | ID: mdl-37638827

ABSTRACT

The tumor microenvironment (TME) plays an important role in the development of tumors. Immunoregulatory cells and cytokines facilitate cancer cells to avoid immune surveillance. Overexpression of immune checkpoint molecules such as CTLA-4 and PD-1/PD-L1 inhibits immune function and enables cancer cells to avoid clearance by the immune system. Thus, minimizing tumor immunosuppression could be an important strategy for cancer therapy. Currently, many immune checkpoint-targeted drugs, such as PD-1/PD-L1 inhibitors, have been approved for marketing and have shown unique advantages in the clinical treatment of cancers. The concept of "strengthening resistance to eliminate pathogenic factors" in traditional Chinese medicine (TCM) is consistent with the immunotherapy of cancer. According to previous studies, the role of TCM in tumor immunotherapy is mainly associated with the positive regulation of natural killer cells, CD8/CD4 T cells, dendritic cells, M2 macrophages, interleukin-2, tumor necrosis factor-[Formula: see text], and IFN-[Formula: see text], as well as with the negative regulation of Tregs, myeloid-derived suppressor cells, cancer-associated fibroblasts, PD-1/PD-L1, transforming growth factor-[Formula: see text], and tumor necrosis factor-[Formula: see text]. This paper summarizes the current research on the effect of TCM targeting the TME, and further introduces the research progress on studying the effects of TCM on immune checkpoints. Modern pharmacological studies have demonstrated that TCM can directly or indirectly affect the TME by inhibiting the overexpression of immune checkpoint molecules and enhancing the efficacy of tumor immunotherapy. TCM with immunomodulatory stimulation could be the key factor to achieve benefits from immunotherapy for patients with non-inflammatory, or "cold", tumors.


Subject(s)
B7-H1 Antigen , Neoplasms , Humans , B7-H1 Antigen/pharmacology , Medicine, Chinese Traditional , Immune Checkpoint Proteins/pharmacology , Programmed Cell Death 1 Receptor , Neoplasms/pathology , Immunotherapy , Tumor Necrosis Factors/pharmacology , Tumor Microenvironment
5.
Hepatology ; 77(5): 1639-1653, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36626628

ABSTRACT

BACKGROUND AND AIMS: Biliary atresia (BA), a congenital cholestatic liver disease, commonly culminates in end-stage liver disease. We previously demonstrated in BA that Prominin-1 ( Prom1 )-expressing hepatic progenitor cells (HPCs) expand within regions of developing fibrosis, giving rise to cholangiocytes within biliary ductular reactions. Null mutation of Prom1 or ablation of cells expressing Prom1 significantly diminishes fibrogenesis. FN14, the receptor for TNF-like weak inducer of apoptosis (TWEAK), is expressed by HPCs. TWEAK/FN14 signaling promotes fibrosis in multiple organ systems. Therefore, we hypothesized that TWEAK/FN14 signaling mediates Prom1 -expressing HPC proliferation leading to profibrogenic ductular reactions in BA. APPROACH AND RESULTS: The experimental mouse model of BA mediated by perinatal rhesus rotavirus (RRV) infection resulted in increased co-expression of Fn14 in Prom1 -expressing HPCs within regions of ductular reactions. FN14 antagonist L524-0366 decreased ductular reactions, biliary fibrosis and periportal fibroblast activation in RRV injury. L524-0366 inhibition also demonstrated loss of downstream noncanonical NF-kB signaling expression in RRV injury. Murine HPC organoids demonstrated accelerated organoid growth and proliferation when treated with recombinant TWEAK. Increased organoid proliferation with recombinant TWEAK was lost when also treated with L524-0366. Analysis of a large publicly available RNA sequencing database of BA and normal control patients revealed significant increases in expression of PROM1 , FN14 , and genes downstream of TNF signaling and noncanonical NF-κB signaling pathways in BA infants. Infants who failed to achieve bile drainage after hepatoportoenterostomy had higher relative levels of FN14 expression. CONCLUSION: TWEAK/FN14 signaling activation in Prom1 -expressing HPCs contributes to proliferation of profibrogenic ductular reactions in BA.


Subject(s)
Biliary Atresia , Rotavirus Infections , Rotavirus , Animals , Mice , AC133 Antigen/genetics , Biliary Atresia/metabolism , Fibrosis , Rotavirus/metabolism , Stem Cells/metabolism , Transcription Factors , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology
6.
J Am Heart Assoc ; 12(3): e027589, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36718875

ABSTRACT

Background Angiotensin II type 1 receptor blockers (ARBs) have been shown to limit the growth of abdominal aortic aneurysm (AAA), but their efficacy is controversial. This study aimed to investigate the molecular mechanism underlying the protective effect of ARBs against AAA progression. Methods and Results Olmesartan, an ARB, was administered to wild-type and osteoprotegerin-knockout (Opg-KO) mice starting 2 weeks before direct application of CaCl2 to aortas to induce AAA. The protective effect of olmesartan against AAA in wild-type and Opg-KO mice was compared at 6 weeks after AAA induction. Olmesartan prevented AAA progression in Opg-KO mice, including excessive aortic dilatation and collapse of tunica media, but not in wild-type mice. Deficiency of the Opg gene is known to cause excessive activation of the tumor necrosis factor-related apoptosis-inducing ligand-induced c-Jun N-terminal kinase/matrix metalloproteinase 9 pathway, resulting in prolonged AAA progression. Olmesartan attenuated the upregulation of phosphorylated c-Jun N-terminal kinase and matrix metalloproteinase 9 expression in the aortic wall of Opg-KO mice. In cultured vascular smooth muscle cells, tumor necrosis factor-related apoptosis-inducing ligand-induced c-Jun N-terminal kinase phosphorylation and matrix metalloproteinase 9 expression were inhibited by angiotensin (1-7), the circulating levels of which are increased by ARBs. Furthermore, administering an angiotensin (1-7) antagonist to Opg-KO mice diminished the protective effect of olmesartan against AAA progression. Conclusions Olmesartan prevented AAA progression in Opg-KO mice by upregulating angiotensin (1-7), suggesting that angiotensin (1-7) may be a key factor that mediates the protective effect of ARBs.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Aortic Aneurysm, Abdominal , Animals , Mice , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/prevention & control , Disease Models, Animal , JNK Mitogen-Activated Protein Kinases/metabolism , Ligands , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology , Up-Regulation
7.
Turk J Gastroenterol ; 34(3): 211-220, 2023 03.
Article in English | MEDLINE | ID: mdl-36511604

ABSTRACT

BACKGROUND: The purpose of the present research was to explore the therapeutic impact of raw lacquer extract from Toxicodendron vernicifluum on colorectal cancer cells and to investigate the outcome of raw lacquer extract and ONC201 co-treatment on the activity of colorectal cancer cells. METHODS: The cells of HCT116 were treated with raw lacquer extract, ONC201, or co-treatment. Subsequently, MTT, trypan blue staining, colony formation, annexin V/propidium iodide staining, wound healing, and transwell assays were performed to assess the effects of raw lacquer extract, ONC201 and the synthesis effect of co-treatment on cell activity, survival, proliferation, apoptosis, migration, and invasion in HCT116 cells. Western blotting and immunostaining assay were also performed to detect the expressions of tumor necrosis factor-related apoptosis-inducing ligand, death receptor-5, cleaved caspase-8, p-mTOR/mTOR, and p-S6K/S6K in cells. RESULTS: The results showed that ONC201 and raw lacquer extract had effective anti-cancer effects on HCT116 cells. ONC201 and raw lacquer extract treatment on colorectal cancer cells inhibited cell viability and growth, as well as induced cell apoptosis and cell death of HCT116. The migration and invasion of HCT116 cells were also inhibited. Significantly, raw lacquer extract and ONC201 cotreatment further enhanced the anti-colorectal cancer cell activity in HCT116 cells. Western blotting and immunostaining assay showed that raw lacquer extract in combination with ONC201 induced tumor necrosis factor-related apoptosis-inducing ligand/death receptor-5 expression activation, inhibited the expression of cleaved caspase-8/procaspase-8, and reduced the expression of p-mTOR/mTOR and p-S6K/S6K. CONCLUSION: These results indicated that raw lacquer extract in combination with ONC201 enhanced the inhibitory effects on colorectal cancer cell activity.


Subject(s)
Colorectal Neoplasms , Toxicodendron , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Toxicodendron/metabolism , Caspase 8/metabolism , Caspase 8/pharmacology , Caspase 8/therapeutic use , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Lacquer , Ligands , TOR Serine-Threonine Kinases/metabolism , Apoptosis , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology , Tumor Necrosis Factors/therapeutic use , Cell Proliferation
8.
J Ethnopharmacol ; 303: 115929, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36379416

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Prostate cancer originates from cells inside a gland, which begin to grow out of control. In the world, prostate cancer is the most common cancer in the male population. New therapeutic strategies are needed for this tumor which still has a high mortality. A. arborescens leaves and aerial parts have various ethnopharmacological uses such as anti-spasmodic, and their decoctions were used to resolve urticaria, neuralgia and several lung diseases. Often this species has been also used to treat different inflammatory-related diseases such as cancer. AIM OF THE STUDY: In a continuation of our research on essential oils from medicinal plants, we have selected, two essential oils from Artemisia arborescens L. (Compositae), an aromatic shrub widely used in traditional medicine. We evaluated their pro-apototic effect on androgen-sensitive (LNCaP) and androgen-insensitive (DU-145) human prostate cancer cells. In this study, we also evaluated the anti-Signal transducer and transcription factor 3 (STAT-3) activity of both essential oils in the human prostate cancer cell lines, and the treatment with Tumor necrosis factor (TNF)-Related Apoptosis (TRAIL). MATERIALS AND METHODS: The cells were exposed to essential oils for 72 h and cell viability and cell membrane integrity were evaluated. Genomic DNA and the activity of caspase-3 was tested to confirm the cell death for apoptosis. Western blot analysis was employed to evaluate the expression of Bcl-2, Bax, cleaved caspase-3, cleaved caspase-9, Hsp70, STAT-3 and SOD proteins. Assays to evaluate reactive oxygen species (ROS) and GSH levels were also performed. RESULTS: The results showed the capacity of two essential oils to activate an apoptotic process increasing the inhibition of Hsp70 and STAT-3 protein expression. In addition, our natural products sensitize LNCaP cells to Tumor necrosis factor (TNF)-Related Apoptosis (TRAIL)-induced apoptosis. CONCLUSIONS: In summary, our study provides a further contribution to the hypothesis of the use of essential oils, from traditional medicinal plants, for the treatment of tumors, and suggests that the combination of our samples with other anti-prostate cancer therapies could be used to affect prostate cancer.


Subject(s)
Artemisia , Oils, Volatile , Prostatic Neoplasms , Male , Humans , Caspase 3/metabolism , Oils, Volatile/pharmacology , Oils, Volatile/therapeutic use , Androgens/pharmacology , Apoptosis , Prostatic Neoplasms/metabolism , Tumor Necrosis Factors/pharmacology , Tumor Necrosis Factors/therapeutic use , Cell Line, Tumor
9.
Yakugaku Zasshi ; 142(10): 1045-1053, 2022.
Article in Japanese | MEDLINE | ID: mdl-36184438

ABSTRACT

Sialic acid-binding lectin from Rana catesbeiana (cSBL) is a multifunctional protein with both lectin and ribonuclease activity and is, therefore, called a leczyme. It exerts cancer cell-selective antitumor effects on a variety of cancer cells in vitro and in vivo under conditions where no undesired side effects are observed. cSBL elicits antitumor effects by degrading cellular RNA and subsequently inducing apoptosis via a pathway mediated by mitochondria and endoplasmic reticulum stress. Further, it exerts synergistic antitumor effects with other molecules such as tumor necrosis factor-related apoptosis-inducing ligand and pemetrexed. Recent studies have revealed that long-term treatment of cancer cells with cSBL causes significant pleiotropic changes in the expression profiles of several genes, including multiple genes involved in metabolic pathways. Furthermore, cSBL reduces the expression of some cancer-related molecules such as human epidermal growth factor receptors, aldo-keto reductase 1B10, and ATP-binding cassette transporter C2. The information described above is expected to lead to useful applications, such as effective regimens comprising cSBL and other drugs. These findings reveal favorable properties of cSBL as an anticancer drug, which may contribute to the development of new therapeutic strategies for cancer treatment.


Subject(s)
Antineoplastic Agents , Neoplasms , ATP-Binding Cassette Transporters , Aldo-Keto Reductases/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , EGF Family of Proteins/metabolism , EGF Family of Proteins/pharmacology , Humans , Lectins/chemistry , Ligands , N-Acetylneuraminic Acid/pharmacology , Neoplasms/drug therapy , Pemetrexed , RNA/pharmacology , Rana catesbeiana/metabolism , Ribonucleases/metabolism , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology
10.
Food Funct ; 13(20): 10587-10600, 2022 Oct 17.
Article in English | MEDLINE | ID: mdl-36156620

ABSTRACT

Chronic kidney disease (CKD) with diverse aetiologies is emerging as a challenging kidney disorder associated with inflammation and interstitial fibrosis. Carvacrol (CVL) is a bioactive monoterpenoid found abundantly in oregano, thyme, and bergamot, having diverse pharmacological benefits. However, the effect of CVL against fibrotic changes in the kidneys is poorly defined. In the current study, a robust mouse model of renal fibrosis induced through unilateral ureteral obstruction (UUO) is used to investigate the anti-fibrotic activity of CVL. The mice were treated with two different oral doses of CVL (25 mg kg-1 and 50 mg kg-1 body weight) for 14 consecutive days. The UUO induction resulted in impaired renal function, severe histological damage, and collagen deposition in the obstructed kidney. Our findings revealed profound activation of transforming growth factor-ß1 (TGF-ß1) and NF-κB (p65) signaling along with the downregulation of antioxidant proteins, nuclear factor-erythroid factor 2-related factor 2 (Nrf2), NAD(P)H: quinone oxidoreductase 1 (NQO1), and superoxide dismutase (SOD) in the obstructed kidney. CVL administration markedly recovered antioxidant proteins and kidney histological changes. In addition, CVL blunted the NF-κB (p65) phosphorylation and reduced the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, and cyclooxygenase 2 (COX-2) compared to the UUO control group. CVL also alleviated the increased fibrotic protein levels of TGF-ß1, pSmad2/3, collagen I, collagen III, fibronectin, and myofibroblast activation and epithelial-mesenchymal transition (EMT) markers, including alpha-smooth muscle actin (α-SMA), E-cadherin, and vimentin in the kidneys. Findings from in vitro study also confirmed that CVL inhibits the EMT process in TGF-ß1 stimulated renal tubular epithelial cells (NRK 52E cells). Collectively, our findings indicate that CVL administration attenuates kidney fibrosis by targeting oxidative stress and inflammation.


Subject(s)
Kidney Diseases , Ureteral Obstruction , Actins/metabolism , Animals , Antioxidants/metabolism , Cadherins/metabolism , Collagen/metabolism , Cyclooxygenase 2/metabolism , Cymenes , Fibronectins/metabolism , Fibrosis , Inflammation/metabolism , Interleukin-6/metabolism , Kidney , Kidney Diseases/metabolism , Mice , NAD/metabolism , NAD/pharmacology , NAD/therapeutic use , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidoreductases/metabolism , Quinones/pharmacology , Superoxide Dismutase/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology , Tumor Necrosis Factors/therapeutic use , Ureteral Obstruction/complications , Ureteral Obstruction/pathology , Ureteral Obstruction/therapy , Vimentin/metabolism
11.
Yonsei Med J ; 63(10): 956-965, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36168249

ABSTRACT

PURPOSE: Cerebral ischemia-reperfusion (IR) injury is a severe secondary injury induced by reperfusion after stroke. Didymin has been reported to have a protective effect on intracerebral hemorrhage. However, the underlying mechanism of didymin on regulating cerebral IR injury remains largely unknown. MATERIALS AND METHODS: A rat cerebral IR model and oxygen-glucose deprivation/reperfusion (OGD/R) model in PC12 cells were established. Hematoxylin and eosin (H&E) was used to detect the pathological changes in brain tissues, and TUNEL staining was performed to detect apoptosis of brain tissues. MTT and flow cytometry were used to measure the viability and apoptosis of PC12 cells. QRT-PCR and western blot were used to detect inflammation cytokines in PC12 cells. Western blot was used to measure the expression of PPAR-γ, RXRA, Bax, c-caspase-3, and Bcl-2. RESULTS: Didymin pretreatment decreased apoptotic rates, reduced levels of Bax and c-caspase-3, and increased Bcl-2 level in vivo and in vitro. Additionally, didymin pretreatment increased viability and decreased the inflammation levels [interleukin (IL)-1ß, IL-6, tumor necrosis factor (TNF)-α, and monocyte chemotactic protein (MCP)-1] of OGD/R treated PC12 cells. Moreover, didymin activated the peroxisome proliferator-activated receptors (PPAR) signaling pathway and increased the expression of PPAR-γ and RXRA in OGD/R treated PC12 cells. Inhibition of PPAR-γ eliminated the protective effect of didymin on OGD/R treated cells. CONCLUSION: Didymin protected neuron cells against IR injury in vitro and in vivo by activation of the PPAR pathway. Didymin may be a candidate drug for IR treatment.


Subject(s)
Interleukin-6 , Reperfusion Injury , Animals , Apoptosis , Caspase 3/metabolism , Cytokines/metabolism , Eosine Yellowish-(YS)/pharmacology , Eosine Yellowish-(YS)/therapeutic use , Flavonoids , Glucose/metabolism , Glycosides , Hematoxylin/pharmacology , Hematoxylin/therapeutic use , Inflammation/drug therapy , Monocyte Chemoattractant Proteins/pharmacology , Monocyte Chemoattractant Proteins/therapeutic use , Oxygen/metabolism , PPAR gamma/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/pharmacology , Proto-Oncogene Proteins c-bcl-2/therapeutic use , Rats , Reperfusion Injury/metabolism , Signal Transduction , Tumor Necrosis Factors/pharmacology , Tumor Necrosis Factors/therapeutic use , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/pharmacology
12.
Microbiol Res ; 265: 127200, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36162148

ABSTRACT

The incidence of intra-abdominal candidiasis (IAC), characterized by high morbidity and mortality, has become a serious concern. The limitations of current antifungal drugs on the market underscores the importance of the development of novel antifungal agents. In the present study, the antifungal activity of vitamin D3 (VD3) against various Candida species was investigated. In vitro, the broth microdilution method and solid plate assay confirmed that VD3 inhibited the growth of Candida spp. in a broad-spectrum, dose-dependent manner. VD3 also had a significant antifungal effect on the initiation, development, and maturation phases of biofilm formation in Candida albicans. The mechanism of VD3 action was explored by transcriptomics and reverse transcription quantitative PCR (RT-qPCR) analysis, and showed that VD3 affects ribosome biogenesis, coenzyme metabolism, and carbon metabolism. These results suggested that VD3 may have multitarget effects against C. albicans. In the murine IAC model, VD3 reduced the fungal burden in the liver, kidneys, and small intestine. Further histopathological analysis and quantification of plasma cytokine levels confirmed that VD3 treatment significantly decreased the infiltration of inflammatory cells and the levels of plasma interferon (IFN)-γ and tumor necrosis factor (TNF)-α. Taken together, these findings suggest a new antifungal mechanism for VD3 and indicate that VD3 could be an effective therapeutic agent for use in IAC treatment.


Subject(s)
Candida albicans , Candidiasis , Animals , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Candida , Candidiasis/drug therapy , Candidiasis/microbiology , Carbon , Cholecalciferol/pharmacology , Coenzymes/pharmacology , Cytokines , Interferons/pharmacology , Mice , Microbial Sensitivity Tests , Tumor Necrosis Factors/pharmacology
13.
World J Gastroenterol ; 28(32): 4635-4648, 2022 Aug 28.
Article in English | MEDLINE | ID: mdl-36157920

ABSTRACT

BACKGROUND: Obstructive jaundice (OJ) is caused by bile excretion disorder after partial or complete bile duct obstruction. It may cause liver injury through various mechanisms. Traditional Chinese medicine (TCM) has a lot of advantages in treating OJ. The recovery of liver function can be accelerated by combining Chinese medicine treatment with existing clinical practice. Yinchenhao decoction (YCHD), a TCM formula, has been used to treat jaundice. Although much progress has been made in recent years in understanding the mechanism of YCHD in treating OJ-induced liver injury, it is still not clear. AIM: To investigate chemical components of YCHD that are effective in the treatment of OJ and predict the mechanism of YCHD. METHODS: The active components and putative targets of YCHD were predicted using a network pharmacology approach. Gene Ontology biological process and Kyoto Encyclopedia of Genes and Genomes path enrichment analysis were carried out by cluster profile. We predicted the biological processes, possible targets, and associated signaling pathways that YCHD may involve in the treatment of OJ. Thirty male Sprague-Dawley rats were randomly divided into three groups, each consisting of 10 rats: the sham group (Group S), the OJ model group (Group M), and the YCHD-treated group (Group Y). The sham group only received laparotomy. The OJ model was established by ligating the common bile duct twice in Groups M and Y. For 1 wk, rats in Group Y were given a gavage of YCHD (3.6 mL/kg) twice daily, whereas rats in Groups S and M were given the same amount of physiological saline after intragastric administration daily. After 7 d, all rats were killed, and the liver and blood samples were collected for histopathological and biochemical examinations. Total bilirubin (TBIL), direct bilirubin (DBIL), alanine aminotransferase (ALT), and aspartate transaminase (AST) levels in the blood samples were detected. The gene expression levels of inducible nitric oxide synthase (iNOS) and endothelial nitric oxide synthase (eNOS), and the nucleus positive rate of NF-E2 related factor 2 (Nrf2) protein were measured. Western blot analyses were used to detect the protein and gene expression levels of Nrf2, Kelch-like ECH-associated protein 1, NAD(P)H quinone dehydrogenase 1 (NQO1), and glutathione-S-transferase (GST) in the liver tissues. One-way analysis of variance was used to evaluate the statistical differences using the statistical package for the social sciences 23.0 software. Intergroup comparisons were followed by the least significant difference test and Dunnett's test. RESULTS: The effects of YCHD on OJ involve biological processes such as DNA transcription factor binding, RNA polymerase II specific regulation, DNA binding transcriptional activator activity, and nuclear receptor activity. The protective effects of YCHD against OJ were closely related to 20 pathways, including the hepatitis-B, the mitogen-activated protein kinase, the phosphatidylinositol 3-kinase/protein kinase B, and tumor necrosis factor signaling pathways. YCHD alleviated the swelling and necrosis of hepatocytes. Following YCHD treatment, the serum levels of TBIL (176.39 ± 17.03 µmol/L vs 132.23 ± 13.88 µmol/L, P < 0.01), DBIL (141.41 ± 14.66 µmol/L vs 106.43 ± 10.88 µmol/L, P < 0.01), ALT (332.07 ± 34.34 U/L vs 269.97 ± 24.78 U/L, P < 0.05), and AST (411.44 ± 47.64 U/L vs 305.47 ± 29.36 U/L, P < 0.01) decreased. YCHD promoted the translocation of Nrf2 into the nucleus (12.78 ± 0.99 % vs 60.77 ± 1.90 %, P < 0.001). After YCHD treatment, we found a decrease in iNOS (0.30 ± 0.02 vs 0.20 ± 0.02, P < 0.001) and an increase in eNOS (0.18 ± 0.02 vs 0.32 ± 0.02, P < 0.001). Meanwhile, in OJ rats, YCHD increased the expressions of Nrf2 (0.57 ± 0.03 vs 1.18 ± 0.10, P < 0.001), NQO1 (0.13 ± 0.09 vs 1.19 ± 0.07, P < 0.001), and GST (0.12 ± 0.02 vs 0.50 ± 0.05, P < 0.001), implying that the potential mechanism of YCHD against OJ-induced liver injury was the upregulation of the Nrf2 signaling pathway. CONCLUSION: OJ-induced liver injury is associated with the Nrf2 signaling pathway. YCHD can reduce liver injury and oxidative damage by upregulating the Nrf2 pathway.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Jaundice, Obstructive , Animals , Male , Rats , Alanine Transaminase/metabolism , Aspartate Aminotransferases/metabolism , Bilirubin/pharmacology , Drugs, Chinese Herbal , Glutathione/metabolism , Jaundice, Obstructive/drug therapy , Jaundice, Obstructive/pathology , Kelch-Like ECH-Associated Protein 1/metabolism , Liver/pathology , Mitogen-Activated Protein Kinases/metabolism , NAD/metabolism , NAD/pharmacology , NF-E2-Related Factor 2/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinones/metabolism , Quinones/pharmacology , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , RNA Polymerase II , Signal Transduction , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology
14.
Life Sci ; 307: 120904, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36029850

ABSTRACT

Most of the published experiments about carbimazole (CMZ)-induced testicular injury are constructed in normal healthy animals, which lakes the translational identification. Despite metformin (MET) having advantageous effects on injured testicles, its impact on thyroid function is arguable. In the current levothyroxine (LT4)/CMZ model, Wistar rats were primed by LT4 for sixty days. CMZ was then given individually or simultaneously with different doses of MET, 100, 200, and 400 mg, daily for thirty days. Serum was assessed for thyroid profile panel, sex hormones, and gonadotropin levels. Testicular tissues were examined for steroidogenesis, spermatogenesis, inflammation, and apoptosis. Histopathology of thyroid and testes were examined, besides thyroidal nuclear factor (NF)-kB expression. MET in a dose-response manner improved the LT4/CMZ-induced testicular toxicity by increasing the steroidogenic acute regulatory protein (StAR), and 17-ß-hydroxysteroid dehydrogenase (17ßHSD) activities, the proliferating cell nuclear antigen (PCNA), sperm count and motility, sex hormones, and gonadotropin levels. MET-400 mg markedly decreased the elevated NF-kB expressions, tumour necrosis factor (TNF)-α, caspase-3, and BAX, and increased BCL-2. LT4/CMZ could be used as translational animal modelling. MET displayed a dose-dependent ameliorative effect on the LT4/CMZ model without significant harmful effects on thyroid functions. MET-testicular protective roles in diabetics with thyroidal diseases should be explored.


Subject(s)
Metformin , Testis , Animals , Carbimazole/metabolism , Carbimazole/pharmacology , Caspase 3/metabolism , Gonadal Steroid Hormones/metabolism , Gonadotropins/metabolism , Male , Metformin/metabolism , Metformin/pharmacology , NF-kappa B/metabolism , Oxidative Stress , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Wistar , Semen/metabolism , Spermatogenesis , Testosterone/metabolism , Thyroxine/metabolism , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology , bcl-2-Associated X Protein/metabolism
15.
Tohoku J Exp Med ; 258(2): 143-148, 2022 Sep 29.
Article in English | MEDLINE | ID: mdl-35965095

ABSTRACT

Spinal cord injury (SCI) is commonly associated with neuropathic pain, which affects large population. Thus, the presented investigation evaluates the beneficial effect of epifriedelinol against SCI-associated neuropathic pain. SCI injury was induced in rats by clip-compression and rats were treated with epifriedelinol 100 and 200 mg/kg, i.p. for 21 days after the induction of SCI. The effect of epifriedelinol was assessed on neuropathic pain by mechanical allodynia and locomotor function. Level of inflammatory cytokines were assessed in the neuronal tissue using enzyme linked immunosorbent assay (ELISA) and expression of caspase-3 and Bcl2 protein were assessed by western blot assay. Data of investigation reveals that epifriedelinol reduces mechanical allodynia in SCI injured rats. Moreover, it also improves locomotor function in SCI injured rats. There was significant decrease in level of interleukin (IL)-1ß, IL-6 and tumor necrosis factor (TNF)-α in the neuronal tissues of epifriedelinol-treated group than negative control group. Moreover, treatment with epifriedelinol ameliorates the altered expression of caspase 3, Bcl2 and GluN1 and level of glutamate in neuronal tissue of SCI-injured rats. In conclusion, data reveal that epifriedelinol treatment protects neuropathic pain associated with spinal cord injury by downregulating the N-methyl-D-aspartate (NMDA) receptor function.


Subject(s)
Neuralgia , Spinal Cord Injuries , Animals , Apoptosis , Caspase 3/metabolism , Caspase 3/pharmacology , Down-Regulation , Glutamates/metabolism , Glutamates/pharmacology , Hyperalgesia/complications , Hyperalgesia/drug therapy , Interleukin-6 , N-Methylaspartate/metabolism , N-Methylaspartate/pharmacology , Neuralgia/drug therapy , Neuralgia/metabolism , Neuralgia/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord Injuries/complications , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology
16.
Neurotox Res ; 40(5): 1272-1291, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35781221

ABSTRACT

We aimed to evaluate the effects of 1,2-diacetylbenzene (DAB) and curcumin on neuroinflammation induced by DAB via triggering receptor expressed on myeloid cells 1 (TREM-1), Toll-like receptor 4 (TLR4), and NLR family pyrin domain containing 3 (NLP3)/calcium-dependent activator protein for secretion 1 (CAPS1)/interleukin 1 beta (IL1B) pathways; tau hyperphosphorylation; reactive oxygen species (ROS); and advanced glycation end-product (AGE) in microglia cells; and explore the molecular mechanisms involved in the key genes induced by DAB and targeted by curcumin in silico analysis. In this study, Western blot, quantitative polymerase chain reaction, and immunocytochemistry were used as the key methods in vitro. In silico analysis, GeneMANIA, ToppFun feature, Metascape, CHEA3, Cytoscape, Autodock, and MIENTURNET were the core approaches used. Curcumin inhibited both the DAB-induced TREM-1/DAP12/NLRP3/caspase-1/IL1B pathway and the TLR4/NF-κB pathway. In BV2 cells, curcumin inhibited ROS, AGE, hyperphosphorylation, glycogen synthase kinase-3ß (GSK-3ß), and ß-amyloid while activating nuclear factor erythroid 2-related factor 2 (Nrf2) expression. In silico studies showed that tumor necrosis factor (TNF), IL6, NFKB1, IL10, and IL1B, as well as MTF1 and ZNF267, were shown to be important genes and transcription factors in the pathogenesis of cognitive impairment produced by DAB and curcumin. Three significant miRNAs (hsa-miR-26a-5p, hsa-miR-203a-3p, and hsa-miR-155-5p) implicated in the etiology of DAB-induced cognitive impairment and targeted by curcumin were also identified. Inflammation and cytokine-associated pathways, Alzheimer's disease, and cognitive impairment were characterized as the most significant biological processes implicated in genes, miRNAs, and transcription factors induced by DAB and targeted by curcumin. Our findings provide new insight into fundamental molecular mechanisms implicated in the pathogenesis of cognitive impairment caused by DAB, particularly the effects of neuroinflammation. Furthermore, this study suggests that curcumin might be a promising therapeutic molecule for cognitive impairment treatment through modulating neuroinflammatory responses.


Subject(s)
Curcumin , MicroRNAs , Benzene/pharmacology , Calmodulin/metabolism , Calmodulin/pharmacology , Caspases/metabolism , Curcumin/pharmacology , Diacetyl/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology
17.
Nat Metab ; 4(3): 359-373, 2022 03.
Article in English | MEDLINE | ID: mdl-35288721

ABSTRACT

Both host genetics and the gut microbiome have important effects on human health, yet how host genetics regulates gut bacteria and further determines disease susceptibility remains unclear. Here, we find that the gut microbiome pattern of participants with active tuberculosis is characterized by a reduction of core species found across healthy individuals, particularly Akkermansia muciniphila. Oral treatment of A. muciniphila or A. muciniphila-mediated palmitoleic acid strongly inhibits tuberculosis infection through epigenetic inhibition of tumour necrosis factor in mice infected with Mycobacterium tuberculosis. We use three independent cohorts comprising 6,512 individuals and identify that the single-nucleotide polymorphism rs2257167 'G' allele of type I interferon receptor 1 (encoded by IFNAR1 in humans) contributes to stronger type I interferon signalling, impaired colonization and abundance of A. muciniphila, reduced palmitoleic acid production, higher levels of tumour necrosis factor, and more severe tuberculosis disease in humans and transgenic mice. Thus, host genetics are critical in modulating the structure and functions of gut microbiome and gut microbial metabolites, which further determine disease susceptibility.


Subject(s)
Gastrointestinal Microbiome , Tuberculosis , Animals , Disease Susceptibility , Fatty Acids, Monounsaturated , Humans , Immunity , Mice , Receptor, Interferon alpha-beta , Tuberculosis/genetics , Tumor Necrosis Factors/pharmacology , Verrucomicrobia
18.
Front Immunol ; 12: 648815, 2021.
Article in English | MEDLINE | ID: mdl-34305888

ABSTRACT

Multiple lines of evidence have demonstrated that cigarette smoke or Chronic Obstructive Pulmonary Disease upregulates angiotensin-converting enzyme 2, the cellular receptor for the entry of the severe acute respiratory syndrome coronavirus 2, which predisposes individuals to develop severe Coronavirus disease 2019. The reason for this observation is unknown. We recently reported that the loss of function of Miz1 in the lung epithelium in mice leads to a spontaneous COPD-like phenotype, associated with upregulation of angiotensin-converting enzyme 2. We also reported that cigarette smoke exposure downregulates Miz1 in lung epithelial cells and in mice, and Miz1 is also downregulated in the lungs of COPD patients. Here, we provide further evidence that Miz1 directly binds to and represses the promoter of angiotensin-converting enzyme 2 in mouse and human lung epithelial cells. Our data provide a potential molecular mechanism for the upregulation of angiotensin-converting enzyme 2 observed in smokers and COPD patients, with implication in severe Coronavirus disease 2019.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , Kruppel-Like Transcription Factors/metabolism , Receptors, Virus/genetics , Transcription, Genetic , Alveolar Epithelial Cells/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Animals , BTB-POZ Domain , Cell Line , Cigarette Smoking/adverse effects , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/genetics , Mice , Promoter Regions, Genetic , Protein Binding , Receptors, Virus/metabolism , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/metabolism , Transcription, Genetic/drug effects , Tumor Necrosis Factors/pharmacology , Virus Internalization
19.
Eur J Immunol ; 49(4): 638-652, 2019 04.
Article in English | MEDLINE | ID: mdl-30707446

ABSTRACT

Conventional DCs are a heterogeneous population that bridge the innate and adaptive immune systems. The lung DC population comprises CD103+ XCR1+ DC1s and CD11b+ DC2s; their various combined functions cover the whole spectrum of immune responses needed to maintain homeostasis. Here, we report that in vivo exposure to LPS leads to profound alterations in the proportions of CD103+ XCR1+ DCs in the lung. Using ex vivo LPS and TNF stimulations of murine lung and spleen-isolated DCs, we show that this is partly due to a direct downregulation of the GM-CSF-induced DC CD103 expression. Furthermore, we demonstrate that LPS-induced systemic inflammation alters the transcriptional signature of DC precursors toward a lower capacity to differentiate into XCR1+ DCs. Also, we report that TNF prevents the capacity of pre-DCs to express CD103 upon maturation. Overall, our results indicate that exposure to LPS directly impacts the capacity of pre-DCs to differentiate into XCR1+ DCs, in addition to lowering their capacity to express CD103. This leads to decreased proportions of CD103+ XCR1+ DCs in the lung, favoring CD11b+ DCs, which likely plays a role in the break in homeostasis following LPS exposure, and in determining the nature of the immune response to LPS.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Integrin alpha Chains/metabolism , Lipopolysaccharides/immunology , Lung/immunology , Lung/metabolism , Animals , Antigens, CD/genetics , Biomarkers , Bone Marrow/immunology , Bone Marrow/metabolism , Cells, Cultured , Dendritic Cells/drug effects , Gene Expression , Immunophenotyping , Inflammation Mediators/metabolism , Integrin alpha Chains/genetics , Lung/pathology , Mice , Pneumonia/etiology , Pneumonia/metabolism , Pneumonia/pathology , Signal Transduction/drug effects , Tumor Necrosis Factors/pharmacology
20.
Int J Mol Sci ; 19(5)2018 May 11.
Article in English | MEDLINE | ID: mdl-29751683

ABSTRACT

The first Food and Drug Administration-(FDA)-approved drugs were small, chemically-manufactured and highly active molecules with possible off-target effects, followed by protein-based medicines such as antibodies. Conventional antibodies bind a specific protein and are becoming increasingly important in the therapeutic landscape. A very prominent class of biologicals are the anti-tumor necrosis factor (TNF) drugs that are applied in several inflammatory diseases that are characterized by dysregulated TNF levels. Marketing of TNF inhibitors revolutionized the treatment of diseases such as Crohn's disease. However, these inhibitors also have undesired effects, some of them directly associated with the inherent nature of this drug class, whereas others are linked with their mechanism of action, being pan-TNF inhibition. The effects of TNF can diverge at the level of TNF format or receptor, and we discuss the consequences of this in sepsis, autoimmunity and neurodegeneration. Recently, researchers tried to design drugs with reduced side effects. These include molecules with more specificity targeting one specific TNF format or receptor, or that neutralize TNF in specific cells. Alternatively, TNF-directed biologicals without the typical antibody structure are manufactured. Here, we review the complications related to the use of conventional TNF inhibitors, together with the anti-TNF alternatives and the benefits of selective approaches in different diseases.


Subject(s)
Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Tumor Necrosis Factor Inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Disease Susceptibility , Humans , Immunomodulation , Inflammation Mediators/metabolism , Protein Binding , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism , Tumor Necrosis Factors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...