Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 971
Filter
1.
J Nanobiotechnology ; 22(1): 538, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39227962

ABSTRACT

The risk of ulcerative colitis (UC) is increasing worldwide with limited success using classical drugs, which has underscored the development of novel agents. Recently, carrier-free molecular assembly has been proven to be an effective drug delivery system, but it has yet to be examined for UC drug development using phytochemicals. Based on traditional Chinese medicine compatibility and potential medicinal uses, a pair of natural compounds, berberine (BBR) and magnolol (MAG), were found to self-assemble into nanostructures in aqueous solutions. Spectral analysis revealed that the assembly mechanisms of BBR and MAG were mediated through charge interactions and π-π stacking. Pharmacokinetic studies and animal imaging showed that BBR-MAG self-assembly (BM) effectively promoted the oral bioavailability and biodistribution of BBR in the colon. BM exhibited superior effects in regulating inflammatory factors, maintaining colon barrier integrity, and regulating gut microbiota in a dextran sulfate sodium salt-induced colitis mouse model. Additionally, no apparent signs of toxicity were observed, suggesting that BM has a favorable safety profile. This study presents a new strategy for UC management and highlights the cooperative effects of combined phytochemicals.


Subject(s)
Berberine , Biphenyl Compounds , Colitis, Ulcerative , Lignans , Nanostructures , Animals , Colitis, Ulcerative/drug therapy , Berberine/chemistry , Berberine/pharmacology , Berberine/therapeutic use , Lignans/chemistry , Lignans/pharmacology , Lignans/therapeutic use , Mice , Biphenyl Compounds/chemistry , Nanostructures/chemistry , Male , Dextran Sulfate/chemistry , Colon/drug effects , Colon/pathology , Disease Models, Animal , Tissue Distribution , Mice, Inbred C57BL , Gastrointestinal Microbiome/drug effects , Biological Availability
2.
Biomed Pharmacother ; 179: 117292, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39151314

ABSTRACT

A type of colorectal cancer (CRC),Colitis-associated colorectal cancer (CAC), is closely associated with chronic inflammation and gut microbiota dysbiosis. Berberine (BBR) has a long history in the treatment of intestinal diseases, which has been reported to inhibit colitis and CRC. However, the mechanism of its action is still unclear. Here, this study aimed to explore the potential protective effects of BBR on azoxymethane (AOM)/dextransulfate sodium (DSS)-induced colitis and tumor mice, and to elucidate its potential molecular mechanisms by microbiota, genes and metabolic alterations. The results showed that BBR inhibited the gut inflammation and improved the function of mucosal barrier to ameliorate AOM/DSS-induced colitis. And BBR treatment significantly reduced intestinal tumor development and ki-67 expression of intestinal tissue along with promoted apoptosis. Through microbiota analysis based on the 16 S rRNA gene, we found that BBR treatment improved intestinal microbiota imbalance in AOM/DSS-induced colitis and tumor mice, which were characterized by an increase of beneficial bacteria, for instance Akkermanisa, Lactobacillus, Bacteroides uniformis and Bacteroides acidifaciens. In addition, transcriptome analysis showed that BBR regulated colonic epithelial signaling pathway in CAC mice particularly by tryptophan metabolism and Wnt signaling pathway. Notably, BBR treatment resulted in the enrichment of amino acids metabolism and microbiota-derived SCFA metabolites. In summary, our research findings suggest that the gut microbiota-amino acid metabolism-Wnt signaling pathway axis plays critical role in maintaining intestinal homeostasis, which may provide new insights into the inhibitory effects of BBR on colitis and colon cancer.


Subject(s)
Azoxymethane , Berberine , Colitis-Associated Neoplasms , Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Metabolomics , Transcriptome , Berberine/pharmacology , Berberine/therapeutic use , Animals , Gastrointestinal Microbiome/drug effects , Transcriptome/drug effects , Mice , Azoxymethane/toxicity , Colitis/microbiology , Colitis/chemically induced , Colitis/drug therapy , Colitis-Associated Neoplasms/microbiology , Colitis-Associated Neoplasms/drug therapy , Colitis-Associated Neoplasms/pathology , Male , Mice, Inbred C57BL , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/drug therapy , Dysbiosis , Disease Models, Animal
3.
Int Immunopharmacol ; 140: 112856, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39121609

ABSTRACT

BACKGROUND: Wound healing pivots on a finely orchestrated inflammatory cascade, critical for tissue repair. Chronic wounds, compounded by persistent inflammation and susceptibility to infection, pose formidable clinical challenges. Nanofiber dressings offer promising avenues for wound care, yet their interaction with inflammation and infection remains elusive. We aim to delineate the inflammatory cascade preceding wound closure and assess Cu@Bbc nanofibers' therapeutic efficacy in mitigating inflammation and combating infection. Their unique attributes suggest promise in modulating inflammation, fostering tissue regeneration, and preventing microbial colonization. Investigating the intricate interplay between nanofiber scaffolds, inflammation, and infection may unveil mechanisms of enhanced wound healing. Our findings could stimulate the development of tailored dressings, urgently needed for effective wound management amidst immune dysregulation, infection, and inflammation. METHODS: In this investigation, we synthesized Cu@Bbc nanofibers, incorporating curcumin and berberine chloride, for wound healing applications. We evaluated their individual and combined antibacterial, anti-biofilm, and antioxidant activities, alongside binding affinity with pro-inflammatory cytokines through molecular docking. Morphological characterization was conducted via SEM, FTIR assessed functional groups, and wettability contact angle measured hydrophobic properties. The physical properties, including tensile strength, swelling behavior, and thermal stability, were evaluated using tensile testing, saline immersion method and thermogravimetric analysis. Biodegradability of the nanofibers was assessed through a soil burial test. Biocompatibility was determined via MTT assay, while wound healing efficacy was assessed with in vitro scratch assays. Controlled drug release and antibacterial activity against MRSA were examined, with in vivo assessment in a zebrafish model elucidating inflammatory responses and tissue remodeling. RESULTS: In this study, the synergistic action of curcumin and berberine chloride exhibited potent antibacterial efficacy against MRSA, with significant anti-mature biofilm disruption. Additionally, the combination demonstrated heightened antioxidant potential. Molecular docking studies revealed strong binding affinity with pro-inflammatory cytokines, suggesting a role in expediting the inflammatory response crucial for wound healing. Morphological analysis confirmed nanofiber quality, with drug presence verified via FTIR spectroscopy. Cu@Bbc demonstrated higher tensile strength, optimal swelling behavior, and robust thermal stability as evaluated through tensile testing and thermogravimetric analysis. Additionally, the Cu@Bbc nanofiber showed enhanced biodegradability, as confirmed by the soil burial test. Biocompatibility assessments showed favorable compatibility, while in vitro studies demonstrated potent antibacterial activity. In vivo zebrafish experiments revealed accelerated wound closure, re-epithelialization, and heightened immune response, indicative of enhanced wound healing. CONCLUSION: In summary, our investigation highlights the efficacy of Cu@Bbc nanofibers, laden with curcumin and berberine chloride, in displaying robust antibacterial and antioxidant attributes while also modulating immune responses and inflammatory cascades essential for wound healing. These results signify their potential as multifaceted wound dressings for clinical implementation.


Subject(s)
Anti-Bacterial Agents , Berberine , Curcumin , Methicillin-Resistant Staphylococcus aureus , Nanofibers , Staphylococcal Infections , Wound Healing , Zebrafish , Animals , Methicillin-Resistant Staphylococcus aureus/drug effects , Curcumin/pharmacology , Curcumin/chemistry , Curcumin/therapeutic use , Berberine/pharmacology , Berberine/chemistry , Berberine/therapeutic use , Wound Healing/drug effects , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology , Nanofibers/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/chemistry , Drug Synergism , Molecular Docking Simulation , Cytokines/metabolism , Biofilms/drug effects , Humans , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
4.
Biochem Pharmacol ; 228: 116509, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39214450

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy, primarily due to the intrinsic development of chemoresistance. The most apparent histopathological feature associated with chemoresistance is the alterations in extracellular matrix (ECM) proteins. Natural dietary botanicals such as berberine (BBR) and emodin (EMO) have been shown to possess chemo-preventive potential by regulating ECM in various cancers. Herein, we further investigated the potential synergistic effects of BBR and EMO in enhancing anticancer efficacy by targeting ECM proteins in pancreatic cancer. Genomewide transcriptomic profiling identified that LAMB3 was significantly upregulated in PDAC tissue and highly associated with poor overall survival (OS, hazard ratio [HR], 2.99, 95 % confidence interval [CI], 1.46-6.15; p = 0.003) and progress-free survival (PFS, HR, 2.59; 95 % CI, 1.30-5.18; p = 0.007) in PDAC. A systematic series of functional experiments in BxPC-3 and MIA-PaCa-2 cells revealed that the combination of BBR and EMO exhibited synergistic anti-tumor potential, as demonstrated by cell proliferation, clonogenicity, migration, and invasion assays (p < 0.05-0.001). The combination also altered the expression of key proteins involved in apoptosis, EMT, and EGFR/ERK1,2/AKT signaling. These findings were further supported by patient-derived organoids (PDOs), where the combined treatment resulted in fewer and smaller organoids compared to each compound individually (p < 0.05-0.001). Our results suggest that BBR combined with EMO exerts synergistic anti-cancer effects by modulating the EGFR-signaling pathway through interference with LAMB3 in PDAC.


Subject(s)
Berberine , Drug Synergism , Emodin , ErbB Receptors , Pancreatic Neoplasms , Signal Transduction , Berberine/pharmacology , Berberine/therapeutic use , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Emodin/pharmacology , Emodin/therapeutic use , ErbB Receptors/metabolism , ErbB Receptors/genetics , Signal Transduction/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Animals , Female , Male
5.
Int Immunopharmacol ; 141: 112931, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39146781

ABSTRACT

Uterine inflammation affects 8% of women in the United States and 32% in developing nations, often caused by uncontrolled inflammation and oxidative stress. This condition significantly impacts women's health, productivity, and quality of life, and increases the risk of related morbidities leading to higher healthcare costs. Research now focuses on natural antioxidants and anti-inflammatory, particularly berberine (BBR), an isoquinoline alkaloid known for its antioxidant, anti-inflammatory, and antiapoptotic activities. The present study sought to examine the potential therapeutic efficacy of BBR against uterine inflammation induced by the intrauterine infusion of an iodine (I2) mixture in an experimental setting. Female Sprague Dawley rats (n = 6) were divided into five groups, control, sham, I2, I2 and BBR 10 mg/kg, and I2 and BBR 25 mg/kg-treated groups. Compared to I2 infusion, BBR treatment effectively restored normal uterine histopathology and reduced inflammatory markers such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), nuclear factor- kappa B (NF-κB), monocyte chemoattractant protein 1 (MCP1), and myeloperoxidase (MPO). It lowered oxidative markers like malondialdehyde (MDA), and increased antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD). It balanced apoptotic genes by upregulating B-cell lymphoma 2 (Bcl-2) and downregulating Bcl-2-associated X protein (Bax). Furthermore, BBR reduced the expression of Toll-like receptor 2 (TLR-2), phosphorylated phosphatidylinositol 3­kinase (p-PI3K), and phosphorylated protein kinase B (p-AKT) in the rats treated with intrauterine I2. Ultimately, the therapeutic benefits of BBR can be attributed, to some extent, to its antioxidant, anti-inflammatory, and antiapoptotic properties, in addition to its ability to modulate the TLR-2/p-PI3K/p-AKT axis.


Subject(s)
Anti-Inflammatory Agents , Berberine , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Signal Transduction , Toll-Like Receptor 2 , Uterus , Animals , Female , Berberine/pharmacology , Berberine/therapeutic use , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/genetics , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Uterus/drug effects , Uterus/pathology , Uterus/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Oxidative Stress/drug effects , Apoptosis/drug effects , Humans , Cytokines/metabolism , Inflammation/drug therapy
7.
Int J Mol Sci ; 25(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39125943

ABSTRACT

The rising incidence of colorectal cancer (CRC) and gastric cancer (GC) worldwide, coupled with the limited effectiveness of current chemotherapeutic agents, has prioritized the search for new therapeutic options. Natural substances, which often exhibit cytostatic properties, hold significant promise in this area. This review evaluates the anticancer properties of three natural alkaloids-berberine, sanguinarine, and chelerythrine-against CRC and GC. In vivo and in vitro studies have demonstrated that these substances can reduce tumor volume and inhibit the epithelial-mesenchymal transition (EMT) of tumors. At the molecular level, these alkaloids disrupt key signaling pathways in cancer cells, including mTOR, MAPK, EGFR, PI3K/AKT, and NF-κB. Additionally, they exhibit immunomodulatory effects, leading to the induction of programmed cell death through both apoptosis and autophagy. Notably, these substances have shown synergistic effects when combined with classical cytostatic agents such as cyclophosphamide, 5-fluorouracil, cetuximab, and erlotinib. Furthermore, berberine has demonstrated the ability to restore sensitivity in individuals originally resistant to cisplatin GC. Given these findings, natural compounds emerge as a promising option in the chemotherapy of malignant gastrointestinal tumors, particularly in cases with limited treatment options. However, more research is necessary to fully understand their therapeutic potential.


Subject(s)
Benzophenanthridines , Berberine , Colorectal Neoplasms , Stomach Neoplasms , Humans , Benzophenanthridines/pharmacology , Benzophenanthridines/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Berberine/pharmacology , Berberine/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Animals , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Signal Transduction/drug effects , Alkaloids/pharmacology , Alkaloids/therapeutic use
8.
Physiol Rep ; 12(15): e16146, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39107107

ABSTRACT

This study compared the capacity of two different models of HIIT [high-(HC) and low-(LC) compression], with or without the use of berberine (BBR), on NOD-like receptor pyrin domain-containing protein-3 (NLRP3), H19, interleukin (IL)-1ß, high-sensitivity C-reactive protein (hs-CRP), and insulin resistance markers. Fifty-four middle-aged men with overweight or obesity and prediabetes [fasting blood glucose (FBG) 110-180 mg/dL] were randomly and equally assigned to the HC, LC, HC + BBR, LC + BBR, BBR, and non-exercising control (CON) groups. The HC (2:1 work-to-rest) and LC (1:1 work-to-rest) home-based training programs included 2-4 sets of 8 exercises at 80%-95% HRmax, twice a week for 8 weeks. Participants in the berberine groups received approximately 1000 mg daily. All exercise interventions led to a significant reduction in hs-CRP, IL-1ß, insulin, FBG, and insulin resistance index (HOMA-IR) versus CON. Notably, there was a significant reduction in FBG and HOMA-IR with the BBR group compared to the baseline. Both NLRP3 and H19 experienced a significant drop only with LC in comparison to the baseline. While both exercise protocols were beneficial overall, LC uniquely exhibited more anti-inflammatory effects, as indicated by reductions in H19 and NLRP3. However, the addition of berberine to the exercise programs did not demonstrate additional benefits.


Subject(s)
Berberine , Prediabetic State , Humans , Male , Berberine/pharmacology , Berberine/administration & dosage , Berberine/therapeutic use , Prediabetic State/blood , Middle Aged , Insulin Resistance , High-Intensity Interval Training/methods , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adult , Biomarkers/blood , C-Reactive Protein/metabolism , Dietary Supplements , Interleukin-1beta/blood , Inflammation/blood
9.
J Physiol Pharmacol ; 75(3)2024 Jun.
Article in English | MEDLINE | ID: mdl-39042390

ABSTRACT

Globally, the metabolic dysfunction-associated fatty liver disease (MAFLD) holds the position as the most widespread chronic liver condition. Berberine (BBR) shows promise as a natural compound for managing obesity, hepatic steatosis, and metabolic disorders. The study aimed to investigate the effectiveness of BBR in addressing factors linked to MAFLD. This is a randomized, double-blind, and placebo-controlled clinical trial. Seventy individuals with MAFLD were enrolled in this study and randomly assigned in a 1:1 ratio to two groups. BBR (1500 mg/day) or placebo was administrated orally for 12 weeks. Selected anthropometric, hepatic, and metabolic parameters were assessed. After a 12-week intervention, the BBR group demonstrated a statistically significant decrease in alanine transaminase (ALT) p=0.0105, and de Ritis ratio p=0.0011 compared to the control group. In both groups we observed a decrease in trunk fat (kg) - BBR group p=0.0185, and placebo group p=0.0323. After three months, a significant divergence between the BBR and placebo groups was evident in the alteration of Δ total cholesterol (TC) p=0.0009, favoring the BBR group. Nevertheless, there were no significant differences detected in other lipid and glucose parameters. In the BBR group, we found significant correlations between changes and amelioration of certain variables: Δ body mass index (BMI) correlated with ΔALT (r=0.47; p=0.0089) and D aspartate aminotransferase (AST) (r=0.47; p=0.0081) levels; Δ trunk fat with Δ fatty liver index (FLI) (r=0.55; p=0.0337), Δ homeostasis model assessment for insulin resistant index (HOMA-IR) (r=0.37; p=0.0020), and AST (r=0.42; p=0.0202); D the de Ritis ratio correlated with Δ fibrosis-4 index (FIB-4) levels (r=0.59; p=0.0011); and ΔFLI correlated with ΔHOMA-IR (r=0.37; p=0.0409) and Δ visceral adiposity index (VAI) (r=0.54; p=0.0019), while no significant differences were observed in the Placebo group. The results show that BBR appears to be a bioactive compound that positively impacts MAFLD, however, additional research with extended intervention durations is required to fully assess its efficacy and potential clinical use.


Subject(s)
Berberine , Liver , Humans , Berberine/therapeutic use , Berberine/pharmacology , Double-Blind Method , Male , Female , Middle Aged , Adult , Liver/metabolism , Liver/drug effects , Alanine Transaminase/blood , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Anthropometry
10.
Asian J Androl ; 26(5): 500-509, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39012524

ABSTRACT

ABSTRACT: Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is highly prevalent worldwide and poses a significant threat to men's health, particularly affecting young men. However, the exact causes and mechanisms behind CP/CPPS remain unclear, leading to challenges in its treatment. In this research, a CP/CPPS rat model was established with complete Freund's adjuvant (CFA), and berberine hydrochloride was administered through daily gavage to assess its therapeutic effects. The alterations in the gut microbiome induced by CP/CPPS and berberine hydrochloride were investigated through 16S ribosomal RNA sequencing of cecum content and colonic epithelial cells. To investigate the impact of the gut microbiome on CP/CPPS, a pseudo germ-free rat model was established, and fecal microbiome transplantation (FMT) was performed on these rats. In all, berberine hydrochloride demonstrated effective reduction of inflammation and oxidative stress in the prostate, offering significant therapeutic advantages for CP/CPPS. Through analysis of the gut microbiome using 16S ribosome RNA sequencing, distinct differences were observed between CP/CPPS rats and control rats, and Clostridium butyricum was identified as a key bacteria. Pseudo germ-free rats that underwent FMT from CP/CPPS rats or rats treated with berberine hydrochloride displayed varying levels of inflammatory cytokine production, oxidative stress, and activity of associated signaling pathways. In conclusion, the therapeutic potential of berberine hydrochloride in addressing CP/CPPS is highly significant. The gut microbiome has emerged as a critical factor in the development of CP/CPPS and plays a pivotal role in mediating the therapeutic effects of berberine hydrochloride.


Subject(s)
Berberine , Gastrointestinal Microbiome , Prostatitis , Rats, Sprague-Dawley , Signal Transduction , Berberine/pharmacology , Berberine/therapeutic use , Male , Animals , Prostatitis/microbiology , Prostatitis/drug therapy , Gastrointestinal Microbiome/drug effects , Rats , Signal Transduction/drug effects , Pelvic Pain/drug therapy , Pelvic Pain/therapy , Fecal Microbiota Transplantation , Disease Models, Animal , Oxidative Stress/drug effects , Chronic Pain/drug therapy , Prostate/drug effects , Prostate/microbiology , RNA, Ribosomal, 16S/genetics
11.
Gen Physiol Biophys ; 43(4): 353-366, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38953577

ABSTRACT

This study aimed to assess the prophylactic effects of Berberine on experimentally induced lung sepsis and examine its effects on selected cytokines, genes, and protein expression besides the histopathological evaluation. Berberine significantly reduced the wet/dry lung ratio, the broncho-alveolar lavage fluid (BALF) protein, cells, neutrophils percentage, and cytokines levels. In addition, pretreatment with Berberine decreased the myeloperoxidase (MPO) and malondialdehyde (MDA) levels and decreased gene expression of nuclear factor kappa B (NF-κB), monocyte chemoattractant protein-1 (MCP-1), and the intracellular adhesion molecule 1 (ICAM-1) by RT-qPCR analysis, revealing Berberine's antioxidant and anti-inflammatory mode of action. Western blot analysis revealed increased peroxisome proliferator-activated receptor gamma (PPAR-γ) expression in the Berberine pretreated group compared to the cecal ligation and puncture (CLP) group, in which the histopathological examination evidenced this improvement. In conclusion, Berberine improved lung sepsis via its PPAR-γ mediated antioxidant and anti-inflammatory effects.


Subject(s)
Acute Lung Injury , Berberine , PPAR gamma , Sepsis , Signal Transduction , Berberine/pharmacology , Berberine/therapeutic use , Animals , PPAR gamma/metabolism , Sepsis/metabolism , Sepsis/drug therapy , Rats , Acute Lung Injury/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/prevention & control , Male , Signal Transduction/drug effects , Up-Regulation/drug effects , Rats, Wistar , Rats, Sprague-Dawley
12.
PLoS One ; 19(7): e0306783, 2024.
Article in English | MEDLINE | ID: mdl-39058681

ABSTRACT

BACKGROUND: The prevention of obesity represents a major health and socio-economic challenge. Nutraceuticals are regularly highlighted for their beneficial effects in preventing the metabolic disturbances associated with obesity. However, few studies have described the combined action of nutraceutical mixtures combining polyphenols with alkaloids. OBJECTIVE: The aim of this study was to evaluate the effects of long-term dietary supplementation with a mixture of Berberine, Citrus and Apple extracts (BCA) in the primary prevention of obesity and its metabolic and vascular complications in the obese Zucker rat, a spontaneous model of genetic obesity and insulin resistance. METHODS: Sixteen 8-week-old obese Zucker male rats were randomly divided into two groups: all rats received oral gavage daily either with water, untreated obese (U-ObZ) or BCA (BCA-ObZ) mixture for thirteen weeks. Morphological and metabolic parameters were measured along the study. Cumulative concentration-response curves to insulin, acetylcholine and phenylephrine were determined on isolated thoracic aorta. Colon permeability measurements were performed using the Ussing chamber technique. Fecal samples collected at the beginning and the end of the protocol were used as a template for amplification of the V3-V4 region of the 16S rDNA genes. RESULTS: BCA supplementation reduced weight gain (p<0.05) and food intake (p<0.05) in the BCA-ObZ group rats compared to the U-ObZ group rats. It also improved glucose tolerance (p<0.001) and decreased fasting insulin and Homeostasis model assessment index (p<0.05). Through ex vivo experiments, the BCA mixture enhanced significantly aortic insulin relaxation (p<0.01), reduced α1-adrenoceptor-mediated vasoconstriction (p<0.01), and decreased distal colon permeability. Moreover, short-chain fatty acid producers such as Bacteroides, Blautia, and Akkermansia were found to be increased by the BCA mixture supplementation. CONCLUSION: The results showed that a 13-week-supplementation with BCA mixture prevented weight gain and improved glucose metabolism in obese Zucker rats. We also demonstrated that BCA supplementation improved vascular function, colonic barrier permeability and gut microbiota profile.


Subject(s)
Berberine , Citrus , Dietary Supplements , Malus , Obesity , Plant Extracts , Rats, Zucker , Animals , Berberine/pharmacology , Berberine/therapeutic use , Male , Obesity/metabolism , Obesity/prevention & control , Obesity/drug therapy , Rats , Citrus/chemistry , Plant Extracts/pharmacology , Malus/chemistry , Insulin Resistance , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Insulin/blood , Insulin/metabolism
13.
Acta Pharmacol Sin ; 45(10): 2107-2118, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38862818

ABSTRACT

Diabetic cardiomyopathy (DCM) is a complication of diabetes mellitus characterized by heart failure and cardiac remodeling. Previous studies show that tetrahydroberberrubine (THBru) retrogrades cardiac aging by promoting PHB2-mediated mitochondrial autophagy and prevents peritoneal adhesion by suppressing inflammation. In this study we investigated whether THBru exerted protective effect against DCM in db/db mice and potential mechanisms. Eight-week-old male db/db mice were administered THBru (25, 50 mg·kg-1·d-1, i.g.) for 12 weeks. Cardiac function was assessed using echocardiography. We showed that THBru administration significantly improved both cardiac systolic and diastolic function, as well as attenuated cardiac remodeling in db/db mice. In primary neonatal mouse cardiomyocytes (NMCMs), THBru (20, 40 µM) dose-dependently ameliorated high glucose (HG)-induced cell damage, hypertrophy, inflammatory cytokines release, and reactive oxygen species (ROS) production. Using Autodock, surface plasmon resonance (SPR) and DARTS analyses, we revealed that THBru bound to the domain of the receptor for advanced glycosylation end products (RAGE), subsequently leading to inactivation of the PI3K/AKT/NF-κB pathway. Importantly, overexpression of RAGE in NMCMs reversed HG-induced inactivation of the PI3K/AKT/NF-κB pathway and subsequently counteracted the beneficial effects mediated by THBru. We conclude that THBru acts as an inhibitor of RAGE, leading to inactivation of the PI3K/AKT/NF-κB pathway. This action effectively alleviates the inflammatory responses and oxidative stress in cardiomyocytes, ultimately leading to ameliorated DCM.


Subject(s)
Berberine , Diabetic Cardiomyopathies , Inflammation , Myocytes, Cardiac , Receptor for Advanced Glycation End Products , Animals , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/metabolism , Male , Receptor for Advanced Glycation End Products/metabolism , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Mice , Inflammation/drug therapy , Inflammation/metabolism , Berberine/pharmacology , Berberine/therapeutic use , Berberine/analogs & derivatives , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Cells, Cultured , Signal Transduction/drug effects , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism
14.
Biochem Pharmacol ; 226: 116381, 2024 08.
Article in English | MEDLINE | ID: mdl-38909786

ABSTRACT

The escalating prevalence of obesity presents formidable challenges, necessitating the development of effective therapeutic strategies. In this study, we aimed to elucidate the preventive effects on obesity of tetrahydroberberrubine (THBru), a derivative of berberine (BBR) and to unravel its underlying mechanism. Using an obese mouse model induced by a high-fat diet (HFD), THBru was found to markedly ameliorate obesity, as evidenced by reduced body weight, decreased Lee's index, diminished fat mass in epididymal white adipose tissue (WAT) and brown adipose tissue (BAT), alongside improved dyslipidemia. Notably, at the same dose, THBru exhibited superior efficacy compared to BBR. RNA-sequencing and gene set enrichment analysis indicated THBru activated thermogenesis, which was further confirmed in WAT, BAT, and 3T3-L1 cells. Bioinformatics analysis of RNA-sequencing data revealed the candidate gene Pgc1α, a key regulator involved in thermogenesis. Moreover, THBru was demonstrated to elevate the expression of PGC1α by stabilizing its mRNA in WAT, BAT and 3T3-L1 cells. Furthermore, PGC1α knockdown blocked the pro-thermogenic and anti-obesity action of THBru both in vivo and in vitro. This study unravels the preventive effects of THBru on obesity through the activation of PGC1α-mediated thermogenesis, thereby delineating its potential therapeutic implications for obesity and associated disorders.


Subject(s)
3T3-L1 Cells , Adipose Tissue, Brown , Adipose Tissue, White , Berberine , Diet, High-Fat , Mice, Inbred C57BL , Obesity , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Thermogenesis , Animals , Thermogenesis/drug effects , Mice , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Male , Berberine/pharmacology , Berberine/analogs & derivatives , Berberine/therapeutic use , Obesity/prevention & control , Obesity/metabolism , Obesity/drug therapy , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Diet, High-Fat/adverse effects , Anti-Obesity Agents/pharmacology
15.
Sci Rep ; 14(1): 14924, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38942824

ABSTRACT

Oxyberberine (OBB) is a significant natural compound, with excellent hepatoprotective properties. However, the poor water solubility of OBB hinders its release and absorption thus resulting in low bioavailability. To overcome these drawbacks of OBB, amorphous spray-dried powders (ASDs) of OBB were formulated. The dissolution, characterizations, and pharmacokinetics of OBB-ASDs formulation were investigated, and its hepatoprotective action was disquisitive in the D-GalN/LPS-induced acute liver injury (ALI) mouse model. The characterizations of OBB-ASDs indicated that the crystalline form of OBB active pharmaceutical ingredients (API) was changed into an amorphous form in OBB-ASDs. More importantly, OBB-ASDs showed a higher bioavailability than OBB API. In addition, OBB-ASDs treatment restored abnormal histopathological changes, improved liver functions, and relieved hepatic inflammatory mediators and oxidative stress in ALI mice. The spray drying techniques produced an amorphous form of OBB, which could significantly enhance the bioavailability and exhibit excellent hepatoprotective effects, indicating that the OBB-ASDs can exhibit further potential in hepatoprotective drug delivery systems. Our results provide guidance for improving the bioavailability and pharmacological activities of other compounds, especially insoluble natural compounds. Meanwhile, the successful development of OBB-ASDs could shed new light on the research process of poorly soluble medicine.


Subject(s)
Berberine , Biological Availability , Toll-Like Receptor 4 , Animals , Toll-Like Receptor 4/metabolism , Mice , Berberine/pharmacology , Berberine/chemistry , Berberine/therapeutic use , Male , Solubility , Liver/metabolism , Liver/drug effects , Liver/pathology , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Disease Models, Animal , Oxidative Stress/drug effects , Protective Agents/pharmacology , Protective Agents/chemistry , Lipopolysaccharides , Powders , Drug Delivery Systems
16.
Comput Biol Med ; 178: 108804, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38941899

ABSTRACT

Chronic atrophic gastritis (CAG), characterized by inflammation and erosion of the gastric lining, is a prevalent digestive disorder and considered a precursor to gastric cancer (GC). Coptis chinensis France (CCF) is renowned for its potent heat-clearing, detoxification, and anti-inflammatory properties. Zuojin Pill (ZJP), a classic Chinese medicine primarily composed of CCF, has demonstrated effectiveness in CAG treatment. This study aims to elucidate the potential mechanism of CCF treatment for CAG through a multifaceted approach encompassing network pharmacology, molecular docking, molecular dynamics simulation and experimental verification. The study identified three major active compounds of CCF and elucidated key pathways, such as TNF signaling, PI3K-Akt signaling and p53 signaling. Molecular docking revealed interactions between these active compounds and pivotal targets like PTGS2, TNF, MTOR, and TP53. Additionally, molecular dynamics simulation validated berberine as the primary active compound of CCF, which was further confirmed through experimental verification. This study not only identified berberine as the primary active compound of CCF but also provided valuable insights into the molecular mechanisms underlying CCF's efficacy in treating CAG. Furthermore, it offers a reference for refining therapeutic strategies for CAG management.


Subject(s)
Coptis , Drugs, Chinese Herbal , Gastritis, Atrophic , Molecular Dynamics Simulation , Network Pharmacology , Humans , Coptis/chemistry , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/pharmacology , Gastritis, Atrophic/drug therapy , Gastritis, Atrophic/metabolism , Molecular Docking Simulation , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Berberine/chemistry , Berberine/therapeutic use , Berberine/pharmacology , Tumor Necrosis Factor-alpha/metabolism
17.
ACS Biomater Sci Eng ; 10(7): 4347-4358, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38841860

ABSTRACT

In order to improve the effectiveness of tumor treatment and reduce the toxic side effects of drugs, we formed carrier-free multifunctional nanoparticles (BI NPs) by noncovalent interaction of berberine hydrochloride and IR780. BI NPs possessed the synergistic effects of promoting apoptosis, inhibiting proliferation and metastasis of tumors, and phototherapeutic treatment. Dispersive and passive targeting ability retention (EPR) effects of BI NPs on tumor sites in vivo could be monitored by fluorescence imaging. In addition, BI NPs exhibited effective reactive oxygen species (ROS) generation and photothermal conversion capabilities, photodynamic therapy (PDT), and photothermal therapy (PTT). Importantly, BI NPs inhibit tumor suppression through the AMPK/PI3K/AKT signaling pathway to inhibit tumor proliferation and metastasis. BI NPs not only have efficient in vivo multimodal therapeutic effects but also have good biosafety and potential clinical applications.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular , Cell Proliferation , Liver Neoplasms , Nanomedicine , Nanoparticles , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Humans , Cell Proliferation/drug effects , Animals , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Nanomedicine/methods , Mice , Reactive Oxygen Species/metabolism , Photochemotherapy/methods , Berberine/pharmacology , Berberine/chemistry , Berberine/therapeutic use , Photothermal Therapy , Mice, Inbred BALB C , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use
18.
Int Immunopharmacol ; 137: 112422, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38880024

ABSTRACT

The rising prevalence of autoimmune diseases poses a significant challenge to global public health. Continual exploration of natural compounds for effective treatments for autoimmune diseases is crucial. Berberine, a benzylisoquinoline alkaloid, is a bioactive component found in various medicinal plants, exhibiting diverse pharmacological properties. This review aims to consolidate the current understanding of berberine's pharmacological effects and mechanisms in addressing four autoimmune diseases: rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, and psoriasis. Overall, as a traditional Chinese medicinal preparation, berberine shows promise as an effective and safe treatment for autoimmune diseases. However, further comprehensive studies, particularly clinical trials, are essential to elucidate additional mechanisms and molecular targets, as well as to assess the efficacy and safety of berberine in treating these autoimmune diseases.


Subject(s)
Autoimmune Diseases , Berberine , Humans , Berberine/therapeutic use , Berberine/pharmacology , Animals , Autoimmune Diseases/drug therapy
19.
Phytother Res ; 38(8): 3986-4008, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38864713

ABSTRACT

Obesity and Type 2 diabetes are prevalent metabolic dysfunctions that present significant health challenges worldwide. Available cures for these ailments have constraints with accompanying unwanted effects that persistently exist. Compounds originated from plants have recently been introduced as hopeful remedies to treat metabolic disorders because of their diverse pharmacological activities. This detailed observation gives an introduction into the treatment capacity of plant-derived compounds regarding metabolic syndromes while analyzing various groups alongside their performance in this field despite unique mechanisms designed by nature itself. Interestingly, this study provides some examples including curcumin, resveratrol, quercetin, berberine, epigallocatechin gallate (EGCG), and capsaicin, which highlights potential therapeutic impacts for future testing. However, current clinical trials inspecting human studies investigating efficacies concerning metabolism challenge present limitations. Finally, the review weighs up bad reactions possibly inflicted after administering plant-originated materials though suggestive insights will be provided later. Above all, it outlines the chance to identify novel therapies encapsulated within natural substances based upon recent developments could hold significant promise toward managing misplaced metabolisms globally.


Subject(s)
Metabolic Diseases , Humans , Metabolic Diseases/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Catechin/analogs & derivatives , Catechin/pharmacology , Animals , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Obesity/drug therapy , Berberine/pharmacology , Berberine/therapeutic use , Phytotherapy , Phytochemicals/pharmacology , Resveratrol/pharmacology , Resveratrol/therapeutic use , Quercetin/pharmacology , Quercetin/analogs & derivatives , Capsaicin/pharmacology , Capsaicin/therapeutic use
20.
J Cell Mol Med ; 28(12): e18407, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38894630

ABSTRACT

Chronic intermittent hypoxia (CIH) is associated with an increased risk of cardiovascular diseases. Previously, we have shown that berberine (BBR) is a potential cardioprotective agent. However, its effect and mechanism on CIH-induced cardiomyopathy remain uncovered. This study was designed to determine the effects of BBR against CIH-induced cardiac damage and to explore the molecular mechanisms. Mice were exposed to 5 weeks of CIH with or without the treatment of BBR and adeno-associated virus 9 (AAV9) carrying SIRT6 or SIRT6-specific short hairpin RNA. The effect of BBR was evaluated by echocardiography, histological analysis and western blot analysis. CIH caused the inactivation of myocardial SIRT6 and AMPK-FOXO3a signalling. BBR dose-dependently ameliorated cardiac injury in CIH-induced mice, as evidenced by increased cardiac function and decreased fibrosis. Notably, SIRT6 overexpression mimicked these beneficial effects, whereas infection with recombinant AAV9 carrying SIRT6-specific short hairpin RNA abrogated them. Mechanistically, BBR reduced oxidative stress damage and preserved mitochondrial function via activating SIRT6-AMPK-FOXO3a signalling, enhancing mitochondrial biogenesis as well as PINK1-Parkin-mediated mitophagy. Taken together, these data demonstrate that SIRT6 activation protects against the pathogenesis of CIH-induced cardiac dysfunction. BBR attenuates CIH-induced myocardial injury by improving mitochondrial biogenesis and PINK1-Parkin-dependent mitophagy via the SIRT6-AMPK-FOXO3a signalling pathway.


Subject(s)
Berberine , Forkhead Box Protein O3 , Hypoxia , Signal Transduction , Sirtuins , Berberine/pharmacology , Berberine/therapeutic use , Animals , Sirtuins/metabolism , Sirtuins/genetics , Signal Transduction/drug effects , Hypoxia/metabolism , Mice , Male , Forkhead Box Protein O3/metabolism , Forkhead Box Protein O3/genetics , Oxidative Stress/drug effects , Mice, Inbred C57BL , AMP-Activated Protein Kinases/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Mitophagy/drug effects , Ventricular Remodeling/drug effects , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL