Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 302
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Med Virol ; 95(3): e28661, 2023 03.
Article in English | MEDLINE | ID: mdl-36905112

ABSTRACT

Chikungunya virus (CHIKV) infection, a global public health problem, might lead to acute/chronic polyarthritis causing long-term morbidity among infected patients. But, except nonsteroidal anti-inflammatory drugs (NSAIDs) with gastrointestinal, cardiovascular, and immune-related side-effects, no Food and Drug Administration (FDA)-approved analgesic drug is available till date for the treatment of CHIKV-induced arthritis. Curcumin, a plant product with minimal toxicity has been FDA-approved as a Generally Recognized As Safe drug. This study aimed to determine the analgesic and prophylactic effect of curcumin, if any, among CHIKV-induced arthralgic mice. Arthritic pain was evaluated by von Frey assay, locomotory behavior by open-field test, and feet swelling by calipers. Cartilage integrity and proteoglycan loss were evaluated by Safranin O staining followed by Osteoarthritis Research Society International (OARSI), Standardized Microscopic Arthritis Scoring of Histological sections (SMASH) score, and type II collagen loss by immunohistochemistry. Mice were administered high (HD), mid (MD), and low (LD) curcumin doses, before (PT: pretreatment), during (CT: cotreatment) and after (Post-T: posttreatment) CHIKV-infection. Curcumin treatment using PTHD (2000 mg/kg), CTHD , and Post-TMD (1000 mg/kg) significantly alleviated CHIKV-induced arthritic pain by improving pain-threshold, locomotory behavior and reducing feet swelling of infected mice. Also, decreased proteoglycan loss and cartilage erosion with lower OARSI, SMASH scores were observed among these three subgroups compared to infected ones. Compared to infected ones, one- to twofold increased intensity of type II collagen in knee medial femoral condyle and medial tibial plateau regions of these subgroups was observed by immunohistochemical staining. Thus, this study highlighted both the analgesic (CT, Post-T), and prophylactic (PT) activity of curcumin in alleviating CHIKV-induced acute/chronic arthritis within mouse model.


Subject(s)
Arthritis , Chikungunya Fever , Chikungunya virus , Curcumin , Animals , Mice , Chikungunya Fever/drug therapy , Curcumin/pharmacology , Curcumin/therapeutic use , Collagen Type II/therapeutic use , Arthritis/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Disease Models, Animal , Proteoglycans/therapeutic use , Pain/drug therapy
2.
Lipids Health Dis ; 22(1): 120, 2023 Aug 08.
Article in English | MEDLINE | ID: mdl-37553709

ABSTRACT

Obesity is often accompanied by metabolic disorder and insulin resistance, resulting in type 2 diabetes. Based on previous findings, FYGL, a natural hyperbranched proteoglycan extracted from the G. lucidum fruiting body, can decrease blood glucose and reduce body weight in diabetic mice. In this article, the underlying mechanism of FYGL in ameliorating obesity-induced diabetes was further investigated both in vivo and in vitro. FYGL upregulated expression of metabolic genes related to fatty acid biosynthesis, fatty acid ß-oxidation and thermogenesis; downregulated the expression of insulin resistance-related genes; and significantly increased the number of beige adipocytes in db/db mice. In addition, FYGL inhibited preadipocyte differentiation of 3T3-L1 cells by increasing the expression of FABP-4. FYGL not only promoted fatty acid synthesis but also more significantly promoted triglyceride degradation and metabolism by activating the AMPK signalling pathway, therefore preventing fat accumulation, balancing adipocyte production and lipid metabolism, and regulating metabolic disorders and unhealthy obesity. FYGL could be used as a promising pharmacological agent for the treatment of metabolic disorder-related obesity.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Insulin Resistance , Reishi , Mice , Animals , Reishi/metabolism , Lipid Metabolism , Diabetes Mellitus, Experimental/metabolism , Proteoglycans/metabolism , Proteoglycans/pharmacology , Proteoglycans/therapeutic use , Adipocytes/metabolism , Adipogenesis , Obesity/drug therapy , Obesity/genetics , Obesity/metabolism , Fatty Acids/metabolism , 3T3-L1 Cells
3.
Funct Integr Genomics ; 22(6): 1307-1313, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35931836

ABSTRACT

Osteoarthritis (OA) is the most common joint disorder worldwide and a leading cause of pain and disability. However, the pathogenesis of osteoarthritis has not been elucidated. Krüppel-like factor (KLF)-5 is involved in several biological processes, including inflammation and cell differentiation, but its role in OA has not been evaluated. In this study, we investigated the role of KLF-5 in chondrocyte differentiation. KLF-5 overexpression in chondrocytes induced a loss of type II collagen expression and sulfated proteoglycan synthesis at the transcriptional and translational levels. Based on immunofluorescence staining, the ectopic expression of KLF-5 reduced type II collagen expression. In contrast, with KLF-5-transfected cells, KLF-5 siRNA transfection-induced type II expression also blocked dedifferentiation caused by the overexpression of KLF-5. In zebra fish, KLF-5 reduced the sulfated proteoglycan synthesis of ceratobranchial cartilage. Our results suggest that KLF-5 plays a pivotal role in the dedifferentiation of rabbit articular cartilage and zebra fish, providing a basis for therapeutic strategy for osteoarthritis aimed at controlling cartilage destruction.


Subject(s)
Chondrocytes , Osteoarthritis , Animals , Rabbits , Collagen Type II/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Osteoarthritis/genetics , Transcription Factors/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Kruppel-Like Transcription Factors/genetics , Proteoglycans/metabolism , Proteoglycans/therapeutic use , Cells, Cultured
4.
Cell Tissue Res ; 385(1): 265-275, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33837849

ABSTRACT

Glandular epithelial cells (GE) in the endometrium are thought to support the elongation and survival of ruminant embryos by secreting histotrophs. In the present study, the gene expression of bovine endometrial epithelial cells cultured in matrigel was analyzed and examined whether it could be an in vitro model of GE. Bovine endometrial epithelial cells (BEE) and stromal cells (BES) were isolated from the slaughterhouse uteri and cultured in DMEM/F12 + 10% FBS. BEE showed the gland-like structure morphological changes when cultured in 15% matrigel but could not be identified in higher concentrations of the matrigel (30% or 60%). The expression of typical genes expressed in GE, SERPINA14 and GRP, was substantially high in matrigel-cultured BEE than in monolayer (P  <  0.05). P4 and INFα have no significant effect on the SERPINA14 expression of BEE cultured in matrigel without co-culture with BES. On the other hand, when BEE were co-cultured with BES in matrigel culture, the expression of FGF13 was increased by the P4 treatment (P  <  0.05). Furthermore, SERPINA14 and TXN expressions were increased by P4 + IFNα treatment (P  <  0.05). These results demonstrate the appropriate conditions for BEE to form glandular structures in matrigel and the effect of co-culture with BES. The present study highlighted the possible use of matrigel for the culture of BEE to investigate the expression of cell-specific glandular epithelial genes as well as P4 and type-I IFN as factors controlling endometrial function during the implantation period.


Subject(s)
Biocompatible Materials/therapeutic use , Collagen/therapeutic use , Endometrium/physiopathology , Epithelial Cells/metabolism , Gene Expression/genetics , Laminin/therapeutic use , Proteoglycans/therapeutic use , Animals , Cattle , Cells, Cultured , Drug Combinations , Female
5.
J Surg Res ; 261: 74-84, 2021 05.
Article in English | MEDLINE | ID: mdl-33421796

ABSTRACT

BACKGROUND: Although the ability of ß-D-glucan and monophosphoryl lipid A (MPLA) to modulate immune responses has been studied in human primary cells, their effect on sterile inflammation models such as necrotizing pancreatitis has never been investigated. MATERIALS AND METHODS: 85 male New Zealand rabbits were assigned into following groups: A: control, B: pretreatment with ß-D-glucan 3 d before pancreatitis, C: pretreatment with MPLA 3 d before pancreatitis, D: pretreatment with ß-D-glucan and laminarin 3 d before pancreatitis, E: treatment with ß-D-glucan 1 d after pancreatitis, and F: MPLA 1 d after pancreatitis. Pancreatitis was induced by sodium taurocholate injection into the pancreatic duct and parenchyma. Survival was recorded for 21 d. On days 1, 3, and 7, blood was collected for amylase measurement. Peripheral blood mononuclear cells were isolated and stimulated for tumor necrosis factor alpha and interleukin 10 production. Pancreatic necrosis and tissue bacterial load were assessed. RESULTS: 21-d survival was prolonged after pretreatment or treatment with ß-D-glucan; this benefit was lost with laminarin administration. At sacrifice, pancreatic inflammatory alterations were more prominent in the control group. Bacterial load was lower after pretreatment or treatment with ß-D-glucan and MPLA. Tumor necrosis factor alpha production from stimulated peripheral blood mononuclear cells was significantly decreased, whereas interleukin 10 production remained unaltered after pretreatment or treatment with ß-D- glucan. CONCLUSIONS: ß-D-glucan reduces mortality of experimental pancreatitis in vivo. This is mediated through attenuation of cytokine production and prevention of bacterial translocation.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Immunomodulation , Lipid A/analogs & derivatives , Pancreatitis, Acute Necrotizing/drug therapy , Proteoglycans/therapeutic use , Adjuvants, Immunologic/pharmacology , Amylases/blood , Animals , Bacterial Translocation/drug effects , Drug Evaluation, Preclinical , Glucans , Lipid A/pharmacology , Lipid A/therapeutic use , Male , Pancreatitis, Acute Necrotizing/blood , Pancreatitis, Acute Necrotizing/mortality , Proteoglycans/pharmacology , Rabbits , Taurocholic Acid , Tumor Necrosis Factor-alpha/metabolism
6.
Crit Care ; 24(1): 550, 2020 09 05.
Article in English | MEDLINE | ID: mdl-32891170

ABSTRACT

BACKGROUND: (1,3)-ß-D-Glucan has been widely used in clinical practice for the diagnosis of invasive Candida infections. However, such serum biomarker showed potential to guide antimicrobial therapy in order to reduce the duration of empirical antifungal treatment in critically ill septic patients with suspected invasive candidiasis. METHODS: This was a single-centre, randomized, open-label clinical trial in which critically ill patients were enrolled during the admission to the intensive care unit (ICU). All septic patients who presented invasive Candida infection risk factors and for whom an empirical antifungal therapy was commenced were randomly assigned (1:1) in those stopping antifungal therapy if (1,3)-ß-D-glucan was negative ((1,3)-ß-D-glucan group) or those continuing the antifungal therapy based on clinical rules (control group). Serum 1,3-ß-D-glucan was measured at the enrolment and every 48/72 h over 14 days afterwards. The primary endpoint was the duration of antifungal treatment in the first 30 days after enrolment. RESULTS: We randomized 108 patients into the (1,3)-ß-D-glucan (n = 53) and control (n = 55) groups. Median [IQR] duration of antifungal treatment was 2 days [1-3] in the (1,3)-ß-D-glucan group vs. 10 days [6-13] in the control group (between-group absolute difference in means, 6.29 days [95% CI 3.94-8.65], p < 0.001). Thirty-day mortality was similar (28.3% [(1,3)-ß-D-glucan group] vs. 27.3% [control group], p = 0.92) as well as the overall rate of documented candidiasis (11.3% [(1,3)-ß-D-glucan group] vs. 12.7% [control group], p = 0.94), the length of mechanical ventilation (p = 0.97) and ICU stay (p = 0.23). CONCLUSIONS: In critically ill septic patients admitted to the ICU at risk of invasive candidiasis, a (1,3)-ß-D-glucan-guided strategy could reduce the duration of empirical antifungal therapy. However, the safety of this algorithm needs to be confirmed in future, multicentre clinical trial with a larger population. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03117439 , retrospectively registered on 18 April 2017.


Subject(s)
Candidiasis, Invasive/drug therapy , Proteoglycans/administration & dosage , Aged , Antifungal Agents/administration & dosage , Antifungal Agents/therapeutic use , Critical Illness/therapy , Female , Humans , Intensive Care Units/organization & administration , Intensive Care Units/trends , Male , Middle Aged , Proteoglycans/therapeutic use
7.
Biol Pharm Bull ; 43(10): 1542-1550, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32759548

ABSTRACT

The steatosis and resultant oxidative stress and apoptosis play the important roles in the progression of nonalcoholic fatty liver disease (NAFLD), therefore, searching for the effective drugs against NAFLD has been a hot topic. In this work, we investigated a hyperbranched proteoglycan, namely FYGL extracted from Ganoderma lucidum, inhibiting the palmitic acid (PA)-induced steatosis in HepG2 hepatocytes. FYGL compose of hydrophilic polysaccharide and lipophilic protein. Both moieties conclude the reductive residues, such as glucose and cystine, making FYGL capable of anti-oxidation. Herein, we demonstrated that FYGL can significantly inhibit the steatosis, i.e., decrease the contents of triglycerides (TG) and total cholesterol (TC) in hepatic cells on the mechanism of increasing the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC), therefore inhibiting the expressions of sterol regulatory element-binding protein 1 (SREBP1) and fatty acid synthase (FASN), furthermore leading to the carnitine palmitoyl transferase-1 (CPT-1) expression increased against steatosis induced by fatty acids oxidation. Meanwhile, FYGL can alleviate reactive oxygen species (ROS) and malondialdehyde (MDA), promote superoxide dismutase (SOD) and total antioxidant capacity (T-AOC). Moreover, FYGL can prevent the cells from apoptosis by regulating the apoptosis-related protein expressions and alleviating oxidative stress. Notably, FYGL could significantly recover the cells activity and inhibit lactate dehydrogenase (LDH) release which were negatively induced by high concentration PA. These results demonstrated that FYGL has the potential functions to prevent the hepatocytes from lipid accumulation, oxidative stress and apoptosis, therefore against NAFLD.


Subject(s)
Antioxidants/pharmacology , Fungal Polysaccharides/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Proteoglycans/pharmacology , Reishi/chemistry , Antioxidants/therapeutic use , Apoptosis/drug effects , Fungal Polysaccharides/therapeutic use , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/physiology , Humans , Lipogenesis/drug effects , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/drug effects , Palmitic Acid/toxicity , Proteoglycans/therapeutic use
8.
Biomacromolecules ; 20(6): 2230-2240, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31070896

ABSTRACT

Despite the excellent antimicrobial activity, the high toxicity and low selectivity of cationic antimicrobial peptides (AMPs) and their synthetic analogues impede their biomedical applications. In this study, we report a series of cationic peptidopolysaccharides synthesized by thiol-ene click chemistry of grafting antimicrobial polypeptides, methacrylate-ended poly(lysine- random-phenylalanine) (Me-K nF m), onto a thiolated polysaccharide (dextran, Dex) backbone. Their copolymers (Dex- g-K nF m) exhibit potent broad-spectrum antibacterial and antifungal activity against Gram-negative bacteria ( Pseudomonas aeruginosa and Escherichia coli), Gram-positive bacteria [methicillin-resistant Staphylococcus aureus (MRSA) and Staphylococcus epidermidis], and fungi ( Candida albicans) with minimal inhibitory concentrations in the range of 31.25-500 µg·mL-1. More importantly, Dex- g-K nF m copolymers did not induce drug resistance of MRSA up to 17 passages. In addition, these copolymers have an improved hemocompatibility and exhibit good in vitro biocompatibility with murine myoblast (C2C12) cells. Among the synthesized peptidopolysaccharides, DexL- g-K12.5F12.5-50%, as the optimal agent, displayed a selectivity more than 200 times the maximum value of polypeptide molecules. Furthermore, a strong in vivo antimicrobial efficacy with a log reduction above 3 in a mouse bacterial sepsis model has been obtained. These excellent biological properties present a promising prospect for Dex- g-K nF m in biomedical applications.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Antimicrobial Cationic Peptides/chemical synthesis , Click Chemistry , Proteoglycans/chemical synthesis , Animals , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/therapeutic use , Bacterial Infections/drug therapy , Candida albicans/drug effects , Escherichia coli/drug effects , Male , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Microbial Sensitivity Tests , Mycoses/drug therapy , Proteoglycans/pharmacology , Proteoglycans/therapeutic use , Pseudomonas aeruginosa/drug effects
9.
Cell Physiol Biochem ; 49(3): 1163-1179, 2018.
Article in English | MEDLINE | ID: mdl-30196282

ABSTRACT

BACKGROUND/AIMS: Non-alcoholic fatty liver disease (NAFLD) encompasses a series of pathologic changes ranging from steatosis to steatohepatitis, which may progress to cirrhosis and hepatocellular carcinoma. The purpose of this study was to determine whether ganoderma lucidum polysaccharide peptide (GLPP) has therapeutic effect on NAFLD. METHODS: Ob/ ob mouse model and ApoC3 transgenic mouse model were used for exploring the effect of GLPP on NAFLD. Key metabolic pathways and enzymes were identified by metabolomics combining with KEGG and PIUmet analyses and key enzymes were detected by Western blot. Hepatosteatosis models of HepG2 cells and primary hepatocytes were used to further confirm the therapeutic effect of GLPP on NAFLD. RESULTS: GLPP administrated for a month alleviated hepatosteatosis, dyslipidemia, liver dysfunction and liver insulin resistance. Pathways of glycerophospholipid metabolism, fatty acid metabolism and primary bile acid biosynthesis were involved in the therapeutic effect of GLPP on NAFLD. Detection of key enzymes revealed that GLPP reversed low expression of CYP7A1, CYP8B1, FXR, SHP and high expression of FGFR4 in ob/ob mice and ApoC3 mice. Besides, GLPP inhibited fatty acid synthesis by reducing the expression of SREBP1c, FAS and ACC via a FXR-SHP dependent mechanism. Additionally, GLPP reduced the accumulation of lipid droplets and the content of TG in HepG2 cells and primary hepatocytes induced by oleic acid and palmitic acid. CONCLUSION: GLPP significantly improves NAFLD via regulating bile acid synthesis dependent on FXR-SHP/FGF pathway, which finally inhibits fatty acid synthesis, indicating that GLPP might be developed as a therapeutic drug for NAFLD.


Subject(s)
Bile Acids and Salts/metabolism , Lipid Metabolism/drug effects , Proteoglycans/pharmacology , Reishi/metabolism , Signal Transduction/drug effects , Animals , Cholesterol 7-alpha-Hydroxylase/metabolism , Fibroblast Growth Factors/metabolism , Fragile X Mental Retardation Protein/metabolism , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lipid Droplets/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Oleic Acid/pharmacology , Proteoglycans/therapeutic use , Receptors, Cytoplasmic and Nuclear/metabolism
10.
J Surg Res ; 211: 30-38, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28501128

ABSTRACT

BACKGROUND: Effectiveness of protein-bound polysaccharide K (PSK) during adjuvant chemotherapy in gastric cancer patients expressing programmed death-1 ligand 1 (PD-L1) has not been investigated. Investigating this might help in triaging candidates eligible to immunochemotherapy. MATERIALS AND METHODS: In total, 918 patients with stages II and III gastric cancer, undergoing curative gastrectomy, and receiving adjuvant chemotherapy were enrolled in a prospective database, and the patients were retrospectively reviewed. We classified those patients into four cohorts stratified by PD-L1 expression and PSK administration, namely PD-L1, PSK (-,+); PD-L1, PSK (-,-); PD-L1, PSK (+,+); and PD-L1, PSK (+,-). In addition, another independent cohort of 20 patients undergoing radical gastrectomy was prospectively recruited to check their immunological cells of sera before and 2 mo after PSK administration. RESULTS: PSK treatment was an independent prognostic factor for patient's overall survival (P = 0.020), whereas PD-L1 expression per se was not. Administration of PSK prolonged patient survival in stages IIIA and IIIB (P = 0.031) but not in stage II or stage IIIC. Patients with negative expression of PD-L1, treated with PSK had longer survival than those not treated with PSK (P = 0.033). PSK did not affect the survival of patients with positive expression of PD-L1, (P = 0.421). The percentages of natural killer and natural killer T (NKT) cells, but not Th1, Th17, Treg, or IFN-γ+/CD8+ T cells, were significantly increased in PD-L1 (-) patients treated with PSK. However, these findings were not evident in PD-L1 (+) patients. CONCLUSIONS: PSK treatment preferentially confers a survival gain for patients with stage IIIA/IIIB gastric cancer, especially in the PD-L1 (-) subpopulation.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Gastrectomy , Proteoglycans/therapeutic use , Stomach Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/therapeutic use , Chemotherapy, Adjuvant , Female , Fluorouracil/therapeutic use , Follow-Up Studies , Humans , Male , Middle Aged , Prospective Studies , Retrospective Studies , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Stomach Neoplasms/surgery , Treatment Outcome
11.
Biochim Biophys Acta ; 1855(2): 276-300, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25829250

ABSTRACT

Proteoglycans control numerous normal and pathological processes, among which are morphogenesis, tissue repair, inflammation, vascularization and cancer metastasis. During tumor development and growth, proteoglycan expression is markedly modified in the tumor microenvironment. Altered expression of proteoglycans on tumor and stromal cell membranes affects cancer cell signaling, growth and survival, cell adhesion, migration and angiogenesis. Despite the high complexity and heterogeneity of breast cancer, the rapid evolution in our knowledge that proteoglycans are among the key players in the breast tumor microenvironment suggests their potential as pharmacological targets in this type of cancer. It has been recently suggested that pharmacological treatment may target proteoglycan metabolism, their utilization as targets for immunotherapy or their direct use as therapeutic agents. The diversity inherent in the proteoglycans that will be presented herein provides the potential for multiple layers of regulation of breast tumor behavior. This review summarizes recent developments concerning the biology of selected proteoglycans in breast cancer, and presents potential targeted therapeutic approaches based on their novel key roles in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Neovascularization, Pathologic/genetics , Proteoglycans/biosynthesis , Translational Research, Biomedical , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Proteoglycans/antagonists & inhibitors , Proteoglycans/therapeutic use , Signal Transduction/genetics , Tumor Microenvironment/genetics
12.
J Drugs Dermatol ; 15(8): 1001-4, 2016 Aug 01.
Article in English | MEDLINE | ID: mdl-27538002

ABSTRACT

The stress hormone, cortisol, is known to affect the function and cyclic regulation of the hair follicle. When cortisol is present at high levels it has been demonstrated to reduce the synthesis and accelerate the degradation of important skin elements, namely hyaluronan and proteoglycans by approximately 40%. The following discussion outlines the relationship between stress, cortisol, and the effect on the normal function of the hair follicle. As a result of this connection, important correlations have been established in the literature to form a basis for novel, effective treatments of stress-related hair growth disorders.
Amongst various treatment methods and substances, oral supplementation with a specific bioavailable proteoglycan stands out as a promising new therapeutic treatment method.

J Drugs Dermatol. 2016;15(8):1001-1004.


Subject(s)
Alopecia/blood , Hair/growth & development , Hair/metabolism , Hydrocortisone/blood , Stress, Psychological/blood , Alopecia/drug therapy , Alopecia/etiology , Hair/drug effects , Hair Follicle/drug effects , Hair Follicle/metabolism , Hair Follicle/pathology , Humans , Proteoglycans/pharmacology , Proteoglycans/therapeutic use , Stress, Psychological/complications , Stress, Psychological/drug therapy
13.
Postepy Hig Med Dosw (Online) ; 69: 91-7, 2015 Jan 21.
Article in Polish | MEDLINE | ID: mdl-25614677

ABSTRACT

Modern medicine successfully uses multiple immunomodulators of natural origin, that can affect biological reactions and support body's natural defense mechanisms including antitumor activities. Among them is a group of products derived from fungi, including schizophyllan, lentinan, polysaccharide Krestin (PSK), and polysaccharidepeptide (PSP). Present paper is focused on polysaccharidepeptide, which due to the negligible toxicity and numerous benefits for health, is increasingly used in China and Japan as an adjuvant in the treatment of cancer. PSP is a protein-polisaccharide complex with a molecular weight 100 kDa derived from Coriolus versicolor mushroom. The results of numerous studies and clinical trials confirm that it inhibits the growth of cancer cells in in vitro and in vivo settings as well as decreases cancer treatment-related adverse side effects such as fatigue, loss of appetite, nausea, vomiting, and pain. PSP is able to restore weakened immune response observed in patients with cancer during chemotherapy. Its anti-tumor effects seemed to be mediated through immunomodulatory regulation. PSP stimulates cells of the immune system, induces synthesis of cytokines such as interleukin-1ß (IL-1ß), IL-6 and tumor necrosis factor-α (TNF-α), eicosanoids including prostaglandin E2 (PGE2), histamine, reactive oxygen species and nitrogen mediators. There is a growing interest in understanding the mechanisms of PSP action. Because of its unique properties and safety, PSP may become a widely used therapeutic agent in the near future.


Subject(s)
Antineoplastic Agents/therapeutic use , Basidiomycota/chemistry , Immunologic Factors/therapeutic use , Neoplasms/drug therapy , Phytotherapy , Plant Preparations/therapeutic use , Proteoglycans/therapeutic use , China , Humans , Japan
14.
Pharm Res ; 31(9): 2453-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24633416

ABSTRACT

PURPOSE: The purpose of this study was to investigate the effect of locally delivered pancreatic islet with liposomal clodronate (Clodrosome®) as an immunoprotection agent for the treatment of type 1 diabetes. METHOD: The bio-distribution of liposomal clodronate in matrigel was checked by imaging analyzer. To verify the therapeutic efficacy of locally delivered islet with liposomal clodronate using injectable hydrogel, four groups of islet transplanted mice (n = 6 in each group) were prepared: 1) the islet group, 2) the islet-Clodrosome group, 3) the islet-Matrigel group, and 4) the islet-Matrigel-Clodrosome group. Immune cell migration and activation, and pro-inflammatory cytokine secretion was evaluated by immunohistochemistry staining and ELISA assay. RESULTS: Cy5.5 labeled liposomes remained in the matrigel for over 7 days. The median survival time of transplanted islets (Islet-Matrigel-Clodrosome group) was significantly increased (>60 days), compared to other groups. Locally delivered liposomal clodronate in matrigel effectively inhibited the activation of macrophages, immune cell migration and activation, and pro-inflammatory cytokine secretion from macrophages. CONCLUSIONS: Locally co-delivered pancreatic islets and liposomal clodronate using injectable hydrogel effectively cured type 1 diabetes. Especially, the inhibition of macrophage attack in the early stage after local delivery of islets was very important for the successful long-term survival of delivered islets.


Subject(s)
Clodronic Acid/administration & dosage , Collagen/administration & dosage , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation/methods , Laminin/administration & dosage , Proteoglycans/administration & dosage , Animals , Clodronic Acid/therapeutic use , Collagen/therapeutic use , Diabetes Mellitus, Type 1/immunology , Drug Combinations , Inflammation/immunology , Inflammation/prevention & control , Injections , Laminin/therapeutic use , Liposomes , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred C57BL , Proteoglycans/therapeutic use , Rats, Sprague-Dawley
15.
J Cancer Res Clin Oncol ; 149(11): 9191-9200, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37188984

ABSTRACT

OBJECTIVES: Pancreatic ductal adenocarcinoma (PDAC) represents a widespread form of malignant pancreatic neoplasms and a leading oncologic cause of death in Europe and the USA. Despite advances in understanding its molecular biology, the 5-year survival rate remains low at 10%. The extracellular matrix in PDAC contains proteins, including SPOCK2, which are essential for tumorigenicity and drug resistance. The present study aims to explore the possible role of SPOCK2 in the pathogenesis of PDAC. MATERIALS AND METHODS: Expression of SPOCK2 was evaluated in 7 PDAC cell lines and 1 normal pancreatic cell line using quantitative RT-PCR. Demethylation of the gene was carried out using 5-aza-2'-deoxycytidine (5-aza-dC) treatment with subsequent validation Western Blot analysis. In vitro downregulation of SPOCK2 gene was performed using siRNA transfection. MTT and transwell assays were employed to evaluate the impact of the SPOK2 demethylation on the proliferation and migration of PDAC cells. KM Plotter was applied to analyze a correlation between SPOCK2 mRNA expression and the survival of PDAC patients. RESULTS: In contrast to the normal pancreatic cell line, SPOCK2 expression was significantly downregulated in PDAC cell lines. Treatment with 5-aza-dC, led to increase in SPOCK2 expression in the cell lines tested. Importantly, compared with control cells, transfected with SPOCK2 siRNA cells exhibited increased growth rates and more migration ability. Finally, we demonstrated that a high SPOCK2 expression level correlated with longer overall survival of patients with PDAC. CONCLUSION: The expression of SPOCK2 is downregulated in PDAC as a result of hypermethylation of its corresponding gene. SPOCK2 expression as well as the demethylation of its gene could be a potential marker for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Prognosis , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Gene Expression , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Proteoglycans/genetics , Proteoglycans/metabolism , Proteoglycans/therapeutic use , Pancreatic Neoplasms
16.
J Cancer Res Clin Oncol ; 149(17): 16055-16067, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37695389

ABSTRACT

PURPOSE: Glioblastoma is one of the malignant tumors with poor prognosis and no effective treatment is available at present. METHODS: To study the effect of cordycepin combined with temozolomide on glioblastoma, we explored the effect of the combination based on network pharmacology and biological verification. RESULTS: It was found that the drug combination significantly inhibited the cell growth, proliferation, migration and invasion of LN-229 cells. Drug combination inhibited epithelial-mesenchymal transition (EMT) by up-regulating the expression of E-cadherin and suppressing the expression of N-cadherin, Zeb1 and Twist1. Through network pharmacology, we further explored the molecular mechanism of drug combination against glioblastoma, and 36 drug-disease common targets were screened. The GO biological process analysis included 44 items (P < 0.01), which mainly involved the regulation of apoptosis, cell proliferation, cell migration, etc. The enrichment analysis of KEGG pathways included 28 pathways (P < 0.05), and the first four pathways were "MicroRNA in cancer, Proteoglycans in cancer, Pathways in cancer and PI3K-AKT signaling pathway". We detected the expression of important genes in the pathways and PPI network, and the results showed that the drug combination down-regulated NFKB1, MYC, MMP-9, MCL1, CTNNB1, and up-regulated PDCD4. CONCLUSION: Cordycepin combined with temozolomide may down-regulate MYC through "MicroRNA in cancer, Proteoglycans in cancer, Pathways in cancer and PI3K-AKT signaling pathway", which in turn regulate the expression of MCL1, CTNNB1, MMP9, PDCD4, thus regulating cell proliferation, migration and apoptosis in glioblastoma.


Subject(s)
Glioblastoma , MicroRNAs , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/therapeutic use , Cell Line, Tumor , MicroRNAs/genetics , Cell Proliferation , Drug Combinations , Proteoglycans/metabolism , Proteoglycans/pharmacology , Proteoglycans/therapeutic use , RNA-Binding Proteins , Apoptosis Regulatory Proteins/metabolism
17.
Cytotherapy ; 14(5): 584-97, 2012 May.
Article in English | MEDLINE | ID: mdl-22348702

ABSTRACT

BACKGROUND AIMS: Previous studies have reported that scaffold or cell-based transplantation may improve functional recovery following spinal cord injury (SCI), but these results were based on neuronal regeneration and cell replacement. In this study, we investigated whether a combination of Matrigel and neural-induced mesenchymal stem cells (NMSC) improved hindlimb function in dogs with SCI, and what mechanisms were involved. METHODS: We pre-differentiated canine adipose-derived mesenchymal stem cells into NMSC. A total of 12 dogs subjected to SCI procedures were assigned to one of the following three transplantation treatment groups: phosphate-buffered saline (PBS); Matrigel; or Matrigel seeded with NMSC. Treatment occurred 1 week after SCI. Basso, Beattie and Bresnahan (B.B.B.) and Tarlov scores, histopathology, immunofluorescence staining and Western blot analysis were used to evaluate the treatment effects. RESULTS: Compared with dogs administered PBS or Matrigel alone, dogs treated with Matrigel + NMSC showed significantly better functional recovery 8 weeks after transplantation. Histology and immunochemical analysis revealed that the combination of Matrigel + NMSC reduced fibrosis from secondary injury processes and improved neuronal regeneration more than the other treatments. In addition, the combination of Matrigel + NMSC decreased the expression of inflammation and/or astrogliosis markers. Increased expressions of intracellular molecules related to neuronal extension, neuronal markers and neurotrophic factors were also found in the Matrigel + NMSC group. However, the expression of nestin as a neural stem cell marker was increased with Matrigel alone. CONCLUSIONS: The combination of Matrigel + NMSC produced beneficial effects in dogs with regard to functional recovery following SCI through enhancement of anti-inflammation, anti-astrogliosis, neuronal extension and neuronal regeneration effects.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cell Transplantation , Neurons/cytology , Neurons/metabolism , Spinal Cord Injuries/therapy , Animals , Biomarkers/analysis , Cell- and Tissue-Based Therapy , Collagen/therapeutic use , Dogs , Drug Combinations , Gene Expression , Hindlimb/physiopathology , Laminin/therapeutic use , Proteoglycans/therapeutic use , Regenerative Medicine , Spinal Cord Injuries/physiopathology , Treatment Outcome
18.
Cytotherapy ; 14(10): 1235-44, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23066785

ABSTRACT

BACKGROUND AIMS: In this study we investigated the effect of neurotrophin-3 (NT-3) and knockdown of NG2, one of the main inhibitory chondroitin sulfate proteoglycans (CSPG), in the glial scar following spinal cord injury (SCI). METHODS: Short hairpin (sh) RNA were designed to target NG2 and were cloned into a lentiviral vector (LV). A LV was also constructed containing NT-3. LV expressing NT-3, shRNA to NG2 or combinations of both vectors were injected directly into contused rat spinal cords 1 week post-injury. Six weeks post-injection of LV, spinal cords were examined by histology for changes in scar size and by immunohistochemistry for changes in expression of CSPG, NT-3, astrocytes, neurons and microglia/macrophages. Motor function was assessed using the Basso, Beattie and Bresnahan (BBB) locomotor scale. RESULTS: Animals that received the combination treatment of LV shNG2 and LV NT-3 showed reduced scar size. These animals also showed an increase in levels of neurons and NG2, a decrease in levels of astrocytes and a significant functional recovery as assessed using the BBB locomotor scale at 2 weeks post-treatment. CONCLUSIONS: The improvement in locomotor recovery and decrease in scar size shows the potential of this gene therapy approach as a therapeutic treatment for SCI.


Subject(s)
Antigens/therapeutic use , Genetic Therapy , Lentivirus/genetics , Locomotion , Neurotrophin 3/therapeutic use , Proteoglycans/therapeutic use , RNA, Small Interfering/administration & dosage , Spinal Cord Injuries/therapy , Animals , Antigens/genetics , CD11b Antigen/metabolism , Cellular Microenvironment , Chondroitin Sulfate Proteoglycans/metabolism , Cicatrix/pathology , Cicatrix/physiopathology , Female , Gene Transfer Techniques , Genetic Vectors/genetics , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Neurocan , Neurotrophin 3/genetics , Proteoglycans/genetics , Rats , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Staining and Labeling , Tubulin/metabolism
19.
Br J Nutr ; 108(11): 2014-25, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-22453054

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is implicated in the negative regulation of the insulin signalling pathway by dephosphorylating the insulin receptor (IR) and IR substrates. Ganoderma lucidum has traditionally been used for the treatment of diabetes in Chinese medicine; however, its anti-diabetic potency and mechanism in vivo is still unclear. Our previously published study reported a novel proteoglycan PTP1B inhibitor, named Fudan-Yueyang-Ganoderma lucidum (FYGL) from G. lucidum, with a half-maximal inhibitory concentration (IC50) value of 5·12 (sem 0·05) µg/ml, a protein:polyglycan ratio of 17:77 and 78 % glucose in polysaccharide, and dominant amino acid residues of aspartic acid, glycine, glutamic acid, alanine, serine and threonine in protein. FYGL is capable of decreasing plasma glucose in streptozotocin-induced diabetic mice with a high safety of median lethal dose (LD50) of 6 g/kg. In the present study, C57BL/6 db/db diabetic mice were trialed further using FYGL as well as metformin for comparison. Oral treatment with FYGL in db/db diabetic mice for 4 weeks significantly (P < 0·01 or 0·05) decreased the fasting plasma glucose level, serum insulin concentration and the homeostasis model assessment of insulin resistance. FYGL also controlled the biochemistry indices relative to type 2 diabetes-accompanied lipidaemic disorders. Pharmacology research suggests that FYGL decreases the plasma glucose level by the mechanism of inhibiting PTP1B expression and activity, consequently, regulating the tyrosine phosphorylation level of the IR ß-subunit and the level of hepatic glycogen, thus resulting in the improvement of insulin sensitivity. Therefore, FYGL is promising as an insulin sensitiser for the therapy of type 2 diabetes and accompanied dyslipidaemia.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Enzyme Inhibitors/therapeutic use , Hypoglycemic Agents/therapeutic use , Hypolipidemic Agents/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Proteoglycans/therapeutic use , Reishi/chemistry , Animals , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/therapeutic use , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/isolation & purification , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/isolation & purification , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/isolation & purification , Insulin Resistance , Lipid Metabolism/drug effects , Liver/drug effects , Liver/enzymology , Liver/metabolism , Liver Glycogen/metabolism , Male , Mice , Mice, Mutant Strains , Organ Specificity , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Subunits/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Proteoglycans/administration & dosage , Proteoglycans/isolation & purification , Receptor, Insulin/metabolism
20.
Langenbecks Arch Surg ; 397(3): 475-80, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22207390

ABSTRACT

PURPOSE: We previously demonstrated that hepatic ischemia and reperfusion (I/R) injury increased liver metastasis and cancer growth of RCN-H4 cells. Using a rat model of hepatic I/R-induced liver metastasis, we investigated the metastasis-suppressing effect of polysaccharide-K (PSK), a biological response modifier composed of protein-bound polysaccharide. METHODS: Fischer rats underwent 60 min of 70% partial hepatic ischemia. After 60 min of reperfusion, rat colon adenocarcinoma cells (RCN-H4) were inoculated intrasplenically. PSK was administered orally before I/R, after I/R, or before and after I/R. The weights of metastatic lesions of the liver or the numbers of liver metastatic nodules were determined on day 21. The effect of PSK on angiogenesis was studied by a rat cornea model using RCN-H4 cells or a vascular endothelial growth factor (VEGF)-containing pellet and an in vitro VEGF-induced endothelial cell migration assay. RESULTS: PSK administration significantly (p < 0.05) suppressed the I/R-induced increase in hepatic metastasis of RCN-H4 cells. The suppression of I/R-promoted metastasis was observed irrespective of the timing of administration. Furthermore, PSK significantly suppressed angiogenesis induced by RCN-H4 cells (p < 0.05) and the VEGF pellet (p < 0.01). PSK significantly suppressed the VEGF-induced migration of vascular endothelial cells (p < 0.05). CONCLUSION: PSK may suppress metastasis induced by hepatic I/R. The suppression of angiogenesis by PSK may be one of the mechanisms of the inhibition of hepatic metastasis.


Subject(s)
Colonic Neoplasms/pathology , Immunologic Factors/therapeutic use , Neovascularization, Physiologic/drug effects , Proteoglycans/therapeutic use , Stress, Physiological/drug effects , Animals , Cell Line, Tumor , Disease Models, Animal , Liver Neoplasms/physiopathology , Liver Neoplasms/secondary , Male , Rats , Rats, Inbred F344 , Reperfusion Injury/physiopathology , Stress, Physiological/physiology , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL