Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Phytomedicine ; 121: 155113, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37748388

ABSTRACT

BACKGROUND: Accumulating evidence suggested increasing energy expenditure is a feasible strategy for combating obesity, and browning of white adipose tissue (WAT) to promote thermogenesis might be one of the attractive ways. Hydroxy-α-sanshool (HAS), a natural amide alkaloid extracted from the fruits of Zanthoxylum bungeanum Maxim, possesses lots of benefits in lipid metabolism regulation. METHODS: The anti-obesity effect of HAS was investigated by establishing an animal model of obesity and a 3T3-L1 differentiation cell model. Effects of HAS on the whole-body fat and liver of obese mice, and the role of HAS in inducing browning of white fat were studied by Micro CT, Metabolic cage detection, Cell mitochondrial pressure detection, transmission electron microscopy and cold exposure assays. Furthermore, the Real-time PCR (qPCR), digital PCR (dPCR), western blot, Co-immunoprecipitation (Co-IP), molecular docking, drug affinity responsive target stability (DARTS), Cellular thermal shift assay (CETSA) and other methods were used to investigate the target and mechanisms of HAS. RESULTS: We found that treatment with HAS helped mice combat obesity caused by a high fat diet (HFD) and improve metabolic characteristics. In addition, our results suggested that the anti-obesity effect of HAS is related to increase energy consumption and thermogenesis via induction of browning of WAT. The further investigations uncovered that HAS can up-regulate UCP-1 expression, increase mitochondria number, and elevate the cellular oxygen consumption rates (OCRs) of white adipocytes. Importantly, the results indicated that browning effects of HAS is closely associated with SIRT1-dependent PPAR-γ deacetylation through activating the TRPV1/AMPK pathway, and TRPV1 is the potential drug target of HAS for the browning effects of WAT. CONCLUSIONS: Our results suggested the HAS can promote browning of WAT via regulating AMPK/SIRT-1/PPARγ signaling, and the potential drug target of HAS is the membrane receptor of TRPV1.


Subject(s)
PPAR gamma , Zanthoxylum , Mice , Animals , PPAR gamma/metabolism , Fruit , Molecular Docking Simulation , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, White , Obesity/drug therapy , Obesity/metabolism , Polyunsaturated Alkamides/pharmacology , Diet, High-Fat/adverse effects , 3T3-L1 Cells , TRPV Cation Channels/metabolism , TRPV Cation Channels/pharmacology
2.
Aging (Albany NY) ; 14(13): 5376-5389, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35696640

ABSTRACT

The mushroom Ganoderma lucidum is a traditional Chinese medicine and G. lucidum spore oil (GLSO) is the lipid fraction isolated from Ganoderma spores. We examined the effect of GLSO on burn wound healing in mice. Following wounding, GLSO was applied on the wounds twice daily. Repair analysis was performed by Sirius-Red-staining at different time points. Cell proliferation and migration assays were performed to verify the effect of GLSO on growth. Network pharmacology analysis to identify possible targets was also carried out, followed by Western blotting, nuclear translocation, cell proliferation, and immunofluorescence assays for in-depth investigation of the mechanism. Our study showed that GLSO significantly promoted cell proliferation, and network pharmacology analysis suggested that GLSO might act through transient receptor potential vanilloid receptor 1 (TRPV1)/SMAD signaling. Furthermore, GLSO elevated SMAD2/3 expression in skin burn and promoted its nuclear translocation, and TRPV1 expression was also increased upon exposure to GLSO. Cell proliferation and immunofluorescence assays with TRPV1 inhibitor showed that GLSO accelerated skin burn wound healing through TRPV1 and SMADs signaling, which provides a foundation for clinical application of GLSO in the healing of deep skin burns.


Subject(s)
Burns , Reishi , Animals , Burns/drug therapy , Cell Proliferation , Mice , Oils/pharmacology , Smad Proteins , TRPV Cation Channels/pharmacology , Wound Healing
3.
Inflammation ; 42(5): 1777-1787, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31190106

ABSTRACT

Electroacupuncture (EA) pretreatment, electrical stimulation using metal needle at specific acupoints in advance, possesses the potential to prevent cerebral ischemia-reperfusion injury (CIRI). Transient receptor potential vanilloid 1 (TRPV-1) has been indicated to take part in cerebral protection of EA; however, the detailed mechanisms remain unclear. The aim of this study was to investigate whether neuroprotection of EA pretreatment against CIRI is associated with TRPV-1 and explore the underlying mechanisms. Middle cerebral artery occlusion (MCAO) was performed to induce CIRI after EA pretreatment at Baihui (GV20), bilateral Shenshu (BL23), and Sanyinjiao (SP6) acupoints in rats. Neurological deficit scores, infarct volumes, oxidative stress damage, inflammatory cytokine production, MAPK signaling activation, and the expression of TRPV-1 were assessed. EA pretreatment lowered neurological deficit scores, reduced infarct volumes, impeded oxidative stress injury, inhibited inflammatory cytokine production, curbed P38 phosphorylation, and suppressed TRPV-1 expression in MCAO rats. Attributing to inhibition of TRPV-1 expression, AMG-517 (TRPV-1 antagonist) showed the synergistic effect with EA pretreatment on the neuroprotection against ischemia-reperfusion injury. However, TRPV-1 agonists capsaicin significantly abrogated the neuroprotective effects of EA pretreatment in MCAO rats accompanying enhancement of TRPV-1 expression. These findings indicated EA pretreatment exerted neuroprotection in rats with cerebral ischemia-reperfusion injury, which at least partially were associated with TRPV1-mediated anti-oxidant stress and anti-inflammation via inhibiting P38 MAPK activation.


Subject(s)
Brain Ischemia/prevention & control , Electroacupuncture/methods , Reperfusion Injury/prevention & control , TRPV Cation Channels/physiology , Animals , Combined Modality Therapy , Inflammation/prevention & control , Neuroprotection/drug effects , Oxidative Stress/drug effects , Rats , TRPV Cation Channels/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Nutrients ; 11(1)2019 Jan 21.
Article in English | MEDLINE | ID: mdl-30669695

ABSTRACT

This study aimed to evaluate the effect of Lactobacillus reuteri DSM 17938 (DSM) on ethanol-induced gastric injury, and if its possible mechanism of action is related to inhibiting the transient receptor potential vanilloid type 1 (TRPV1). We evaluated the effect of supplementing 108 CFU•g body wt-1•day-1 of DSM on ethanol-induced gastric injury. DSM significantly reduced the ulcer area (1.940 ± 1.121 mm²) with 3 days of pretreatment. The effects of DSM supplementation were reversed by Resiniferatoxin (RTX), TRPV1 agonist (3 nmol/kg p.o.). Substance P (SP) (1 µmol/L per 20 g) plus 50% ethanol resulted in hemorrhagic lesions, and DSM supplementation did not reverse the lesion area induced by administering SP. TRPV1 staining intensity was lower, SP, malondialdehyde (MDA) and nitrite levels were reduced, and restored normal levels of antioxidant parameters (glutathione and superoxide dismutase) in the gastric mucosa in mice treated with DSM. In conclusion, DSM exhibited gastroprotective activity through decreased expression of TRPV1 receptor and decreasing SP levels, with a consequent reduction of oxidative stress.


Subject(s)
Ethanol/adverse effects , Gastric Mucosa/pathology , Limosilactobacillus reuteri/growth & development , Probiotics/therapeutic use , Stomach Ulcer/prevention & control , Substance P/antagonists & inhibitors , TRPV Cation Channels/antagonists & inhibitors , Animals , Antioxidants/metabolism , Diterpenes/pharmacology , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastritis/chemically induced , Gastritis/metabolism , Gastritis/prevention & control , Glutathione/metabolism , Limosilactobacillus reuteri/classification , Malondialdehyde/metabolism , Mice , Protective Agents/therapeutic use , Species Specificity , Stomach/microbiology , Stomach/pathology , Stomach Ulcer/chemically induced , Stomach Ulcer/metabolism , Stomach Ulcer/pathology , Superoxide Dismutase/metabolism , TRPV Cation Channels/pharmacology
5.
Phytother Res ; 32(6): 1108-1118, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29464796

ABSTRACT

In this study, a series of 20 structurally similar vanilloids (Vn) were tested for their antiproliferative effects against 12 human cancer cells: human breast (MCF-7 and MDA-MB-231), cervical (HeLa), ovarian (Caov-3), lung (A549), liver (HepG2), colorectal (HT-29 and HCT116), nasopharyngeal (CNE-1 and HK-1), and leukemic (K562 and CEM-SS) cancer cells. Among all the tested vanilloids, Vn16 (6-shogaol) exhibited the most potent cytotoxic effects against human colorectal cancer cells (HT-29). The apoptotic induction effects exhibited by Vn16 on HT-29 cells were confirmed using dual staining fluorescence microscopy and enzyme-linked immunosorbent assay. The effects of Vn16 on regulation of 43 apoptotic-related markers were determined in HT-29. The results suggested that 8 apoptotic markers (caspase 8, BAD, BAX, second mitochondrial-derived activator, caspase 3, survivin, bcl-2, and cIAP-2) were either upregulated or downregulated. These results further support the chemopreventive properties of foods that contain vanilloids.


Subject(s)
Antineoplastic Agents/therapeutic use , Inhibitor of Apoptosis Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , TRPV Cation Channels/therapeutic use , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , TRPV Cation Channels/pharmacology
6.
Invest New Drugs ; 35(3): 324-333, 2017 06.
Article in English | MEDLINE | ID: mdl-28150073

ABSTRACT

Introduction This was an open-label, dose escalation (3 + 3 design), Phase I study of SOR-C13 in patients with advanced tumors of epithelial origin. Primary objectives were to assess safety/tolerability and pharmacokinetics. Secondary goals were to assess pharmacodynamics and efficacy of SOR-C13. Methods SOR-C13 was administered IV QD on days 1-3 and 8-10 of a 21-day cycle. Doses were 2.75 and 5.5 mg/kg (20-min infusion) and 1.375, 2.75, 4.13 and 6.2 mg/kg (90-min infusion). Toxicity was assessed by National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. Dose limiting toxicity (DLT) was assessed within the first treatment cycle. Tumors were evaluated, using Response Evaluation Criteria in Solid Tumors (RECIST) 1.1, after two cycles. Results Twenty-three patients were treated. No drug-related serious adverse events occurred. DLTs occurred in six patients: asymptomatic, drug-related, transient Grade 2 hypocalcemia (4 patients), and unrelated Grade 3 anemia and Grade 3 atrial fibrillation, 1 patient each. Calcium and vitamin D supplementation eliminated further Grade 2 hypocalcemia. One Grade 3 treatment emergent adverse event, urticaria, was definitely related to SOR-C13. Four possibly drug-related, Grade 3 events (alanine aminotransferase and aspartate aminotransferase elevation, headache, and hypokalemia) were observed. Of 22 evaluable patients, 54.5% showed stable disease ranging from 2.8 to 12.5 months. The best response was a 27% reduction in a pancreatic tumor with a 55% reduction in CA19-9 levels at 6.2 mg/kg. Conclusion SOR-C13 was safe and tolerated up to 6.2 mg/kg. The Maximal Tolerated Dose (MTD) was not established. Stable disease suggested antitumor activity.


Subject(s)
Antineoplastic Agents , Calcium Channel Blockers , Neoplasms/drug therapy , TRPV Cation Channels/antagonists & inhibitors , Adult , Aged , Alanine Transaminase/blood , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Aspartate Aminotransferases/blood , Calcium Channel Blockers/adverse effects , Calcium Channel Blockers/pharmacokinetics , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels/genetics , Female , Headache/chemically induced , Humans , Hypocalcemia/chemically induced , Hypokalemia/chemically induced , Keratin-18/blood , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Peptides/adverse effects , Peptides/pharmacokinetics , Peptides/pharmacology , Peptides/therapeutic use , RNA, Messenger/blood , TRPV Cation Channels/adverse effects , TRPV Cation Channels/genetics , TRPV Cation Channels/pharmacokinetics , TRPV Cation Channels/pharmacology , TRPV Cation Channels/therapeutic use , Treatment Outcome , Urticaria/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL