Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.125
Filter
1.
Phytomedicine ; 134: 156015, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39244942

ABSTRACT

BACKGROUND: Pseudorabies virus (PRV), a member of the family Herpesviridae, is responsible for significant economic losses in the pig industry and has recently been associated with human viral encephalitis, leading to severe neurological symptoms post-recovery. Despite the widespread impact of PRV, there are currently no approved effective drugs for treating PRV-related diseases in humans or pigs. Therefore, the exploration and discovery of safe and effective drugs for the prevention and treatment of PRV infection is of paramount importance. PURPOSE: The objective of this study is to screen and identify natural compounds with antiviral activity against PRV. METHODS: First, we used a strain of PRV with green fluorescent protein (PRV-GFP) to screen a natural product chemical library to identify potential antiviral drugs. Next, we assessed the antiviral abilities of salvianolic acid A (SAA) in vitro using virus titer assay, qPCR, and IFA. We investigated the mechanisms of SAA's antiviral activity through viral attachment, internalization, inactivation, and nuclease digestion assay. Finally, we evaluated the efficacy of SAA in inactivating PRV using mice as the experimental subjects. RESULTS: This study screened 206 natural compounds for anti-PRV activity in vitro, resulting in the identification of seven potential antiviral agents. Notably, SAA emerged as a promising candidate with significant anti-PRV activity. The mechanism of action may be that SAA can directly inactivate the virus by disrupting viral envelope. In vivo experiments have shown that pre-incubation of SAA and PRV can effectively inhibit the infectivity and pathogenicity of PRV in mice. CONCLUSION: This study offers valuable insights into the antiviral properties of SAA, potentially informing strategies for controlling PRV epidemics and treating related diseases in both humans and animals.


Subject(s)
Antiviral Agents , Herpesvirus 1, Suid , Pseudorabies , Herpesvirus 1, Suid/drug effects , Animals , Antiviral Agents/pharmacology , Pseudorabies/drug therapy , Pseudorabies/virology , Mice , Caffeic Acids/pharmacology , Lactates/pharmacology , Virus Internalization/drug effects , Virion/drug effects , Swine , Virus Attachment/drug effects , Cell Line , Female
2.
J Med Life ; 17(6): 574-581, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39296435

ABSTRACT

The mammalian gastrointestinal tract hosts a significant microbial symbiont community, an intriguing feature of this complex organ system. This study aimed to investigate the anti-inflammatory, antioxidant, and protective effects of caffeic acid phenethyl ester (CAPE) against Enterococcus faecalis infection in the stomach at a dose of 106 CFU in Swiss mice. A total of 30 mice were randomly assigned to three groups of ten mice each. Group I was the negative control, Group II was infected orally with E. faecalis for 18 days, and Group III was infected with E. faecalis and treated with CAPE orally at a daily dose of 4 mg/kg for 18 days. We assessed the antioxidant activities of stomach homogenate and the immunohistochemical expressions of the transcription factor nuclear factor kappa B (NF-κB) and proliferating cell nuclear antigen (PCNA). Histopathological examination was performed on the stomachs of all mice. Group II had decreased levels of antioxidant activity and positive expressions of NF-κB and PCNA. Histological observations revealed an increase in mucosal and glandular thickness compared with Group I. Group III, treated with CAPE, showed a significant increase in antioxidant activities and a significant decrease in NF-κB and PCNA immunoreactivities compared with Group II. In addition, Group III showed restoration of the normal thickness of the non-glandular and glandular parts of the stomach. Our results revealed that E. faecalis infection has damaging effects on the stomach and proved that CAPE has promising protective, anti-inflammatory, and antioxidant effects against E. faecalis. Further studies may investigate the potential therapeutic effects of CAPE against E. faecalis infection.


Subject(s)
Antioxidants , Caffeic Acids , Enterococcus faecalis , NF-kappa B , Phenylethyl Alcohol , Animals , Caffeic Acids/pharmacology , Caffeic Acids/therapeutic use , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Phenylethyl Alcohol/therapeutic use , NF-kappa B/metabolism , Enterococcus faecalis/drug effects , Mice , Antioxidants/pharmacology , Gram-Positive Bacterial Infections/drug therapy , Stomach/pathology , Stomach/drug effects , Stomach/microbiology , Male , Proliferating Cell Nuclear Antigen/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/metabolism
3.
Int J Biol Macromol ; 278(Pt 4): 135038, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39256119

ABSTRACT

Rapid hemostasis, potent antimicrobial activity, and efficient wound management are critical factors in enhancing the survival of trauma patients. Chitosan, as a green and sustainable biomaterial with low cost, degradability and biocompatibility, is widely used in the biomedical field. However, chitosan dissolves in an acidic environment, which is not conducive to wound healing. In this study, chitosan was chemically modified to address this limitation. A mussel-inspired hydrogel composed of caffeic acid-grafted chitosan, gallic acid-grafted chitosan, and oxidized microcrystalline cellulose (CHI-C/CSG/OMCC) was designed. This hydrogel exhibits blood-responsive gelation behavior and offers a synergistic combination of tissue adhesion, antimicrobial properties, and tissue repair capabilities. The carboxyl, hydroxyl, phenolic hydroxyl and aldehyde groups within the hydrogel system endowed the hydrogel with excellent adhesion properties (53.1 kPa adhesion strength to porcine skin-adherent tissues), biocompatibility, and excellent antimicrobial properties. Surprisingly, this hydrogel not only achieved rapid and effective hemostasis, but also effectively promoted wound healing in a mouse skin injury model. In addition, its remarkable efficacy in stopping bleeding within approximately 2 min without rebleeding was demonstrated in a porcine model of acute gastrointestinal hemorrhage in the esophagus, stomach, and intestines. This blood-responsive ternary hydrogel offers a promising alternative to wound management materials due to its excellent overall performance and superior efficacy in all phases of wound healing.


Subject(s)
Anti-Bacterial Agents , Bivalvia , Hemostasis , Hydrogels , Wound Healing , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Wound Healing/drug effects , Hemostasis/drug effects , Mice , Bivalvia/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Chitosan/chemistry , Chitosan/pharmacology , Swine , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cellulose/chemistry , Cellulose/pharmacology , Caffeic Acids/pharmacology , Caffeic Acids/chemistry
4.
Carbohydr Polym ; 346: 122648, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245508

ABSTRACT

Uncontrolled non-compressible hemorrhage, which is often accompanied by coagulopathy, is a major cause of mortality following traumatic injuries in civilian and military populations. In this study, coagulopathy-independent injectable catechol-modified chitosan (CS-HCA) hemostatic materials featuring rapid shape recovery were fabricated by combining controlled sodium tripolyphosphate-crosslinking with hydrocaffeic acid (HCA) grafting. CS-HCA exhibited robust mechanical strength and rapid blood-triggered shape recovery. Furthermore, CS-HCA demonstrated superior blood-clotting ability, enhanced blood cell adhesion and activation, and greater protein adsorption than commercial hemostatic gauze and Celox. CS-HCA showed enhanced procoagulant and hemostatic capacities in a lethal liver-perforation wound model in rabbits, particularly in heparinized rabbits. CS-HCA is suitable for mass manufacturing and shows promise as a clinically translatable hemostat.


Subject(s)
Catechols , Chitosan , Hemorrhage , Hemostatics , Chitosan/chemistry , Chitosan/pharmacology , Animals , Rabbits , Catechols/chemistry , Catechols/pharmacology , Hemorrhage/drug therapy , Hemostatics/chemistry , Hemostatics/pharmacology , Blood Coagulation/drug effects , Caffeic Acids/chemistry , Caffeic Acids/pharmacology , Male , Smart Materials/chemistry , Injections
5.
Sci Rep ; 14(1): 20663, 2024 09 05.
Article in English | MEDLINE | ID: mdl-39237669

ABSTRACT

Potatoes (Solanum tuberosum L.) are a significant food crop cultivated around the world. Caffeic acid (CA) can enhance plant growth by promoting antioxidant activity and stimulating root development, contributing to overall plant health and vigor. Cobalt sulfate (CoSO4) boosts plant growth by promoting nitrogen (N) fixation, healthier root development, and chlorophyll synthesis, enhancing photosynthesis and overall plant health. Nanoparticle-coated urea (NPCU) improves nutrient uptake, promoting plant growth efficiency and reducing environmental impact. This study investigates the effects of combining CA, CoSO4, and NPCU as amendments on potatoes with and without NPCU. Four treatments, control, 20 µM CA, 0.15 mg/L CoSO4, and 20 µM CA + 0.15 mg/L CoSO4 with and without NPCU, were applied in four replications using a completely randomized design. Results demonstrate that the combination of CA + CoSO4 with NPCU led to an increase in potato stem length (~ 6%), shoot dry weight (~ 15%), root dry weight (~ 9%), and leaf dry weight (~ 49%) compared to the control in nutrient stress. There was a significant rise in chlorophyll a (~ 27%), chlorophyll b (~ 37%), and total chlorophyll (~ 28%) over the control under nutrient stress also showed the potential of CA + CoSO4 with NPCU. In conclusion, the findings suggest that applying CA + CoSO4 with NPCU is a strategy for alleviating potato nutrient stress.


Subject(s)
Caffeic Acids , Nanoparticles , Solanum tuberosum , Urea , Solanum tuberosum/drug effects , Solanum tuberosum/growth & development , Caffeic Acids/pharmacology , Caffeic Acids/chemistry , Urea/pharmacology , Nanoparticles/chemistry , Cobalt/pharmacology , Cobalt/chemistry , Photosynthesis/drug effects , Chlorophyll/metabolism , Plant Roots/drug effects , Plant Roots/growth & development , Plant Leaves/drug effects
6.
J Nanobiotechnology ; 22(1): 571, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294724

ABSTRACT

Thyroid cancer is one of the most common endocrine malignancies in clinical practice. Traditional surgery and radioactive iodine ablation have poor treatment results for poorly differentiated thyroid cancer, and there is a risk of metastasis and recurrence. In this study, caffeic acid, a natural herbal extract with certain biological activity, has been as precursor to prepare new caffeic acid carbon nanodots via a one-step hydrothermal method. The caffeic acid carbon nanodots retains part of the structure and biological activity of caffeic acid, and have good biocompatibility, water solubility and stability. The construction of the carbon nanodots could effectively improve their bio-absorption rate and the efficacy. In vitro cell experiments showed that low-dose caffeic acid carbon nanodots had a significant inhibitory effect on poorly differentiated papillary thyroid carcinoma BCPAP cells. At low concentrations of 16 µg/mL, the inhibition rate of human thyroid cancer cells BCPAP was ~ 79%. The anti-tumor mechanism was predicted and verified by transcriptome, real-time quantitative PCR and western blot experiments. The caffeic acid carbon nanodots showed to simultaneously downregulate the expression of KRAS, p-BRAF, p-MEK1 and p-ERK1/2, the four continuous key proteins in a MAPK classical signaling pathway. In vivo experiments further confirmed the caffeic acid carbon nanodots could significantly inhibit the tumorigenicity of xenografts in papillary thyroid carcinoma at quite low doses. This piece of work provides a new nanomedicine and therapeutic strategy for highly resistant poorly differentiated papillary thyroid carcinoma.


Subject(s)
Caffeic Acids , Carbon , Mice, Nude , Thyroid Cancer, Papillary , Thyroid Neoplasms , Caffeic Acids/pharmacology , Caffeic Acids/chemistry , Humans , Animals , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Cell Line, Tumor , Carbon/chemistry , Mice , Mice, Inbred BALB C , Drug Resistance, Neoplasm/drug effects , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Female
7.
Toxins (Basel) ; 16(9)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39330861

ABSTRACT

The human colonic commensal enterotoxigenic Bacteroides fragilis (ETBF) is associated with chronic colitis and colon cancer. ETBF colonization induces colitis via the Bacteroides fragilis toxin (BFT). BFT secreted by ETBF cause colon inflammation via E-cadherin cleavage/NF-κB signaling. ETBF promotes colon tumorigenesis via interleukin 17A (IL-17A)/CXCL-dependent inflammation, but its bioactive therapeutics in ETBF-promoted tumorigenesis remain unexplored. In the current study, we investigated the caffeic acid phenethyl ester (CAPE) in the murine model of ETBF colitis and tumorigenesis. In this study, we observed that CAPE treatment mitigated inflammation induced by ETBF in mice. Additionally, our findings indicate that CAPE treatment offers protective effects against ETBF-enhanced colon tumorigenesis in a mouse model of colitis-associated colon cancer induced by azoxymethane (AOM) and dextran sulfate sodium. Notably, the decrease in colon tumorigenesis following CAPE administration correlates with a reduction in the expression of IL-17A and CXCL1 in the gastrointestinal tract. The molecular mechanism for CAPE-induced protection against ETBF-mediated tumorigenesis is mediated by IL-17A/CXCL1, and by NF-κB activity in intestinal epithelial cells. Our findings indicate that CAPE may serve as a preventive agent against the development of ETBF-induced colitis and colorectal cancer (CRC).


Subject(s)
Bacteroides fragilis , Caffeic Acids , Colitis , Phenylethyl Alcohol , Animals , Caffeic Acids/pharmacology , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Bacteroides fragilis/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/microbiology , Mice, Inbred C57BL , Interleukin-17/metabolism , Mice , Carcinogenesis/drug effects , Chemokine CXCL1/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/chemically induced , Colonic Neoplasms/prevention & control , Colonic Neoplasms/pathology , Colonic Neoplasms/microbiology , Male , Colon/drug effects , Colon/pathology , Colon/microbiology , Colon/metabolism , Bacterial Toxins/toxicity , Disease Models, Animal , Azoxymethane/toxicity , Dextran Sulfate , Metalloendopeptidases/metabolism
8.
Aging (Albany NY) ; 16(16): 11926-11938, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39189924

ABSTRACT

Osteoclast activity plays a crucial role in the pathological mechanisms of osteoporosis and bone remodeling. The treatment of these disorders involves the use of pharmacological medicines that work by inhibiting the activity of osteoclasts. Nevertheless, the prevalent and infrequent negative consequences of current antiresorptive and bone anabolic treatments pose significant drawbacks, hence restricting their prolonged administration in patients, particularly those who are elderly and/or suffer from many medical conditions. We are currently in the process of creating a new molecule called N-(4-methoxyphen) methyl caffeamide (MPMCA), which is a derivative of caffeic acid. This compound has shown potential in preventing the production of osteoclasts and causing existing osteoclasts to undergo cell apoptosis. Our investigation discovered that MPMCA hinders osteoclast function via suppressing the MAPK pathways. The expectation is that the findings of this study will stimulate the advancement of a novel approach to treating anti-resorption.


Subject(s)
Apoptosis , Caffeic Acids , Osteoclasts , Osteogenesis , Caffeic Acids/pharmacology , Osteogenesis/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Apoptosis/drug effects , Bone Resorption/therapy , RAW 264.7 Cells , Animals , Mice , Cell Line , Signal Transduction/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism
9.
J Anim Sci ; 1022024 Jan 03.
Article in English | MEDLINE | ID: mdl-39158070

ABSTRACT

Young animals are highly susceptible to intestinal damage due to incomplete intestinal development, making them vulnerable to external stimuli. Weaning stress in piglets, for instance, disrupts the balance of intestinal microbiota and metabolism, triggering intestinal inflammation and resulting in gut damage. Caffeic acid (CA), a plant polyphenol, can potentially improve intestinal health. Here, we evaluated the effects of dietary CA on the intestinal barrier and microbiota using a lipopolysaccharide (LPS)-induced intestinal damage model. Eighteen piglets were divided into three groups: control group (CON), LPS group (LPS), and CA + LPS group (CAL). On the 21st and 28th day, six piglets in each group were administered either LPS (80 µg/kg body weight; Escherichia coli O55:B5) or saline. The results showed that dietary CA improved the intestinal morphology and barrier function, and alleviated the inflammatory response. Moreover, dietary CA also improved the diversity and composition of the intestinal microbiota by increasing Lactobacillus and Terrisporobacter while reducing Romboutsia. Furthermore, the LPS challenge resulted in a decreased abundance of 14 different bile acids and acetate, which were restored to normal levels by dietary CA. Lastly, correlation analysis further revealed the potential relationship between intestinal microbiota, metabolites, and barrier function. These findings suggest that dietary CA could enhance intestinal barrier function and positively influence intestinal microbiota and its metabolites to mitigate intestinal damage in piglets. Consuming foods rich in CA may effectively reduce the incidence of intestinal diseases and promote intestinal health in piglets.


Our study focuses on a major issue affecting young animals. After weaning, piglets are particularly vulnerable to severe intestinal infections due to their immature intestinal systems, leading to damaged barriers and financial losses for the pig industry. We explore the possibility of using caffeic acid (CA), a natural compound found in plants, to promote intestinal health. Our research shows that adding CA to the diet can reduce intestinal inflammation and improve barrier function in weaned piglets challenged by lipopolysaccharide. CA positively affects ileal microbiota by increasing beneficial bacteria like Lactobacillus and Terrisporobacter and decreasing Romboutsia. We also observed differing regulatory effects of CA between the ileum and colon, with opposite changes in primary bile acids. Our findings emphasize the potential of CA as a dietary supplement to improve intestinal barrier function and modulate the inflammatory response by targeting gut microbiota and metabolites. To our knowledge, this is the first to demonstrate the effects of CA on ileal barrier function and microbiota in piglets. Our findings could significantly benefit the pig industry by mitigating financial losses from serious intestinal infections. Additionally, this research may offer key insights into the health of human infants' intestines.


Subject(s)
Caffeic Acids , Gastrointestinal Microbiome , Lipopolysaccharides , Animals , Lipopolysaccharides/pharmacology , Caffeic Acids/pharmacology , Caffeic Acids/administration & dosage , Gastrointestinal Microbiome/drug effects , Swine , Diet/veterinary , Inflammation/veterinary , Swine Diseases/prevention & control , Swine Diseases/microbiology , Intestines/drug effects , Intestines/microbiology , Male , Animal Feed/analysis
10.
Front Immunol ; 15: 1433590, 2024.
Article in English | MEDLINE | ID: mdl-39139557

ABSTRACT

Salvianolic acid A (SalA), a bioactive compound extracted from Salvia miltiorrhiza, has garnered considerable interest for its potential in ameliorating the post-stroke neuroinflammation. This review delineates the possible molecular underpinnings of anti-inflammatory and neuroprotective roles of SalA, offering a comprehensive analysis of its therapeutic efficacy in preclinical studies of ischemic stroke. We explore the intricate interplay between post-stroke neuroinflammation and the modulatory effects of SalA on pro-inflammatory cytokines, inflammatory signaling pathways, the peripheral immune cell infiltration through blood-brain barrier disruption, and endothelial cell function. The pharmacokinetic profiles of SalA in the context of stroke, characterized by enhanced cerebral penetration post-ischemia, makes it particularly suitable as a therapeutic agent. Preliminary clinical findings have demonstrated that salvianolic acids (SA) has a positive impact on cerebral perfusion and neurological deficits in stroke patients, warranting further investigation. This review emphasizes SalA as a potential anti-inflammatory agent for the advancement of innovative therapeutic approaches in the treatment of ischemic stroke.


Subject(s)
Anti-Inflammatory Agents , Caffeic Acids , Neuroinflammatory Diseases , Stroke , Humans , Animals , Caffeic Acids/therapeutic use , Caffeic Acids/pharmacology , Stroke/drug therapy , Stroke/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Lactates/therapeutic use , Lactates/pharmacology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects
11.
Int J Med Mushrooms ; 26(10): 1-8, 2024.
Article in English | MEDLINE | ID: mdl-39171627

ABSTRACT

Mucosal vaccination is a promising strategy for combating infectious diseases caused by pathogenic microbes, as it can generate antigen-specific immune responses in both systemic and mucosal compartments. In our recent study, we developed a nasal vaccine system for Streptococcus pneumoniae infections in mice using enzymatically polymerized polyphenols such as caffeic acid. However, the efficacy of this mucosal vaccine system is approximately 70%, indicating a need for improvement. To address this issue, we hypothesized that incorporating a mucoadhesive agent that enhances mucosal absorption into a polyphenol-based mucosal vaccine system would improve vaccine efficacy. Contrary to our expectations, we found that adding a mucoadhesive agent, hydrophobically modified hydroxypropylmethylcellulose, to the vaccine system reduced the stimulation of antigen-specific antibody responses in both the mucosal (more than 90% reduction; P < 0.05) and systemic compartments (more than 80% reduction; P < 0.05). Although the addition of the mucoadhesive agent may have interfered with the interaction between the mucosal epithelium and the vaccine system, the underlying mechanism remains unclear, and further research is needed to fully understand the mechanisms involved.


Subject(s)
Administration, Intranasal , Caffeic Acids , Animals , Caffeic Acids/administration & dosage , Caffeic Acids/pharmacology , Mice , Mice, Inbred BALB C , Female , Immunity, Mucosal/drug effects , Antibody Formation/drug effects , Pneumococcal Infections/prevention & control , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology
12.
Mol Biol Rep ; 51(1): 931, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39177837

ABSTRACT

Propolis is a natural product used in cancer treatment, which is produced by bees via different sources. The chemical composition of Propolis is determined based on the climatic and geographical conditions, as well as harvesting time and method. This compound has been the subject of numerous investigational endeavors due to its expansive therapeutic capacity which includes antibacterial, anti-fungal, anti-inflammatory, anti-oxidant, anti-viral, and anti-cancer effects. The growing incidence rate of different cancers necessitates the need for developing novel preventive and therapeutic strategies. Chemotherapy, radiotherapy, and stem cell therapy have proved effective in cancer treatment, regardless of the adverse events associated with these modalities. Clinical application of natural compounds such as Propolis may confer promise as an adjuvant therapeutic intervention, particularly in certain subpopulations of patients that develop adverse events associated with anticancer regimens. The diverse biologically active compounds of propolis are believed to confer anti-cancer potential by modulation of critical signaling cascades such as caffeic acid phenethyl ester, Galangin, Artepillin C, Chrysin, Quercetin, Caffeic acid, Nymphaeols A and C, Frondoside A, Genistein, p-coumaric acid, and Propolin C. This review article aims to deliver a mechanistic account of anti-cancer effects of propolis and its components. Propolis can prevent angiogenesis by downregulating pathways involving Jun-N terminal kinase, ERK1/2, Akt and NF-ƘB, while counteracting metastatic progression of cancer by inhibiting Wtn2 and FAK, and MAPK and PI3K/AKT signaling pathways. Moreover, propolis or its main components show regulatory effects on cyclin D, CDK2/4/6, and their inhibitors. Additionally, propolis-induced up-regulation of p21 and p27 may result in cell cycle arrest at G2/M or G0/G1. The broad anti-apoptotic effects of propolis are mediated through upregulation of TRAIL, Bax, p53, and downregulation of the ERK1/2 signaling pathway. Considering the growing body of evidence regarding different anti-cancers effects of propolis and its active components, this natural compound could be considered an effective adjuvant therapy aimed at reducing related side effects associated with chemotherapy and radiotherapy.


Subject(s)
Neoplasms , Propolis , Signal Transduction , Propolis/pharmacology , Propolis/chemistry , Propolis/therapeutic use , Humans , Signal Transduction/drug effects , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Flavonoids/pharmacology , Flavonoids/therapeutic use , Biological Products/pharmacology , Biological Products/therapeutic use , Caffeic Acids/pharmacology , Caffeic Acids/therapeutic use , Caffeic Acids/chemistry , Phenylethyl Alcohol/analogs & derivatives , Phenylpropionates
13.
Molecules ; 29(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39202808

ABSTRACT

Natural caffeic acid (CA) and its analogues have been studied for their potential applications in the treatment of various inflammatory and infectious skin diseases. However, the molecular mechanism underlying the effects of the CA remains largely unknown. Here, we report that CA and its two analogues, caffeic acid phenethyl ester (CAPE) and caffeic acid methyl caffeate (CAMC), inhibit TRPV3 currents in their concentration- and structure-dependent manners with IC50 values ranging from 102 to 410 µM. At the single-channel level, CA reduces the channel open probability and open frequency without alteration of unitary conductance. CA selectively inhibits TRPV3 relative to other subtypes of thermo-TRPs, such as TRPA1, TRPV1, TRPV4, and TRPM8. Molecular docking combined with site-specific mutagenesis reveals that a residue T636 in the Pore-loop is critical for CA binding to TRPV3. Further in vivo evaluation shows that CA significantly reverses TRPV3-mediated skin inflammation induced by skin sensitizer carvacrol. Altogether, our findings demonstrate that CA exerts its anti-inflammatory effects by selectively inhibiting TRPV3 through binding to the pocket formed by the Pore-loop and the S6. CA may serve as a lead for further modification and identification of specific TRPV3 channel inhibitors.


Subject(s)
Caffeic Acids , Molecular Docking Simulation , TRPV Cation Channels , Caffeic Acids/pharmacology , Caffeic Acids/chemistry , TRPV Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , Humans , Animals , Mice , Skin/metabolism , Skin/drug effects , Skin/pathology , Cymenes/pharmacology , Cymenes/chemistry , HEK293 Cells , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Inflammation/drug therapy , Inflammation/metabolism
14.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125618

ABSTRACT

Caffeic acid phenethyl ester (CAPE) is a phenolic natural product with a wide range of biological activities, including anticancer activity; however, the ester group of CAPE is metabolically labile. The corresponding amide, CAPA, has improved metabolic stability but limited anticancer activity relative to CAPE. We report the synthesis using flow and on-water Wittig reaction approaches of five previously reported and five novel CAPA analogues. All of these analogues lack the reactive catechol functionality of CAPA and CAPE. Cytotoxicity studies of CAPE, CAPA, and these CAPA analogues in HeLa and BE(2)-C cells were carried out. Surprisingly, we found that CAPA is cytotoxic against the neuroblastoma BE(2)-C cell line (IC50 = 12 µM), in contrast to the weak activity of CAPA against HeLa cells (IC50 = 112 µM), and the literature reports of the absence of activity for CAPA against a variety of other cancer cell lines. One novel CAPA analogue, 3f, was identified as having cytotoxic activity similar to CAPE in HeLa cells (IC50 = 63 µM for 3f vs. 32 µM for CAPE), albeit with lower activity against BE(2)-C cells (IC50 = 91 µM) than CAPA. A different CAPA analogue, 3g, was found to have similar effects against BE(2)-C cells (IC50 = 92 µM). These results show that CAPA is uniquely active against neuroblastoma cells and that specific CAPA analogues that are predicted to be more metabolically stable than CAPE can reproduce CAPA's activity against neuroblastoma cells and CAPE's activity against HeLa cells.


Subject(s)
Antineoplastic Agents , Caffeic Acids , Phenylethyl Alcohol , Humans , Caffeic Acids/pharmacology , Caffeic Acids/chemistry , Caffeic Acids/chemical synthesis , HeLa Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Phenylethyl Alcohol/chemistry , Phenylethyl Alcohol/chemical synthesis , Water/chemistry , Cell Line, Tumor , Amides/pharmacology , Amides/chemistry , Cell Survival/drug effects
15.
Biomed Pharmacother ; 178: 117158, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39042963

ABSTRACT

Triple-negative breast cancer (TNBC) still one of the most challenging sub-type in breast cancer clinical. Caffeic acid (CA) derived from effective components of traditional Chinese herbal medicine has been show potential against TNBCs. Our research has found that CA can inhibit the proliferation of TNBC cells while also suppressing the size of cancer stem cell spheres. Additionally, it reduces reactive oxygen species (ROS) levels and disruption of mitochondrial membrane potential. Simultaneously, CA influences the stemness of TNBC cells by reducing the expression of the stem cell marker protein CD44. Furthermore, we have observed that CA can modulate the FOXO1/FIS signaling pathway, disrupting mitochondrial function, inducing mitochondrial autophagy, and exerting anti-tumor activity. Additionally, changes in the immune microenvironment were detected using a mass cytometer, we found that CA can induce M1 polarization of macrophages, enhancing anti-tumor immune responses to exert anti-tumor activity. In summary, CA can be considered as a lead compound for further research in targeting TNBC.


Subject(s)
Caffeic Acids , Cell Proliferation , Signal Transduction , Triple Negative Breast Neoplasms , Animals , Female , Humans , Mice , Autophagy/drug effects , Caffeic Acids/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Forkhead Box Protein O1/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment/drug effects
16.
PLoS One ; 19(7): e0307501, 2024.
Article in English | MEDLINE | ID: mdl-39037973

ABSTRACT

The histone deacetylase 2 (HDAC2), an enzyme involved in gene regulation, is a potent drug target for the treatment of colon cancer. Phytocompounds having anticancer properties show the ability to interact with HDAC2 enzyme. Among the compounds, docking scores of caffeic acid (CA) and p-coumaric acid (pCA) with HDAC2 showed good binding efficacy of -5.46 kcal/mol and -5.16 kcal/mol, respectively, with small inhibition constants. The higher binding efficacy of CA compared to pCA can be credited to the presence of an extra oxygen atom in the CA molecule, which forms an additional hydrogen bond with Tyr297. The HDAC2 in complex with these molecules was found to be stable by analyzing RMSD, RMSF, Rg, and SASA values obtained through MD simulations. Furthermore, CA and pCA exhibited low MM/GBSA free energies of -16.32 ± 2.62 kcal/mol and -17.01 ± 2.87 kcal/mol, respectively. The HOMO and LUMO energy gaps, dipole moments, global reactivity descriptor values, and MEP surfaces showed the reactivity of the molecules. The favourable physicochemical and pharmacokinetic properties, along with absence of toxicity of the molecules determined using ADMET analysis, suggested both the acids to be regarded as effective drugs in the treatment of colon cancer.


Subject(s)
Colonic Neoplasms , Histone Deacetylase 2 , Molecular Docking Simulation , Molecular Dynamics Simulation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Humans , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/metabolism , Histone Deacetylase 2/chemistry , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Caffeic Acids/chemistry , Caffeic Acids/pharmacology , Propionates/chemistry , Propionates/pharmacology , Coumaric Acids/chemistry , Coumaric Acids/pharmacology , Hydrogen Bonding , Density Functional Theory
17.
Chem Biol Interact ; 400: 111162, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39047806

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) affects approximately a quarter of the population and, to date, there is no approved drug therapy for this condition. Individuals with type 2 diabetes mellitus (T2DM) are at a significantly elevated risk of developing NAFLD, underscoring the urgency of identifying effective NAFLD treatments for T2DM patients. Salvianolic acid A (SAA) is a naturally occurring phenolic acid that is an important component of the water-soluble constituents isolated from the roots of Salvia miltiorrhiza Bunge. SAA has been demonstrated to possess anti-inflammatory and antioxidant stress properties. Nevertheless, its potential in ameliorating diabetes-associated NAFLD has not yet been fully elucidated. In this study, diabetic ApoE-/- mice were employed to establish a NAFLD model via a Western diet. Following this, they were treated with different doses of SAA (10 mg/kg, 20 mg/kg) via gavage. The study demonstrated a marked improvement in liver injury, lipid accumulation, inflammation, and the pro-fibrotic phenotype after the administration of SAA. Additionally, RNA-seq analysis indicated that the primary pathway by which SAA alleviates diabetes-induced NAFLD involves the cascade pathways of lipid metabolism. Furthermore, SAA was found to be effective in the inhibition of lipid accumulation, mitochondrial dysfunction and ferroptosis. A functional enrichment analysis of RNA-seq data revealed that SAA treatment modulates the AMPK pathway and IGFBP-1. Further experimental results demonstrated that SAA is capable of inhibiting lipid accumulation through the activation of the AMPK pathway and IGFBP-1.


Subject(s)
AMP-Activated Protein Kinases , Caffeic Acids , Insulin-Like Growth Factor Binding Protein 1 , Lactates , Non-alcoholic Fatty Liver Disease , Signal Transduction , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Animals , Mice , Lactates/pharmacology , Lactates/therapeutic use , Lactates/chemistry , AMP-Activated Protein Kinases/metabolism , Caffeic Acids/pharmacology , Caffeic Acids/chemistry , Caffeic Acids/therapeutic use , Insulin-Like Growth Factor Binding Protein 1/metabolism , Insulin-Like Growth Factor Binding Protein 1/genetics , Male , Signal Transduction/drug effects , Mice, Inbred C57BL , Liver/drug effects , Liver/metabolism , Liver/pathology , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Lipid Metabolism/drug effects , Mice, Knockout
18.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000270

ABSTRACT

The combination of a polyphenol, quercetin, with dasatinib initiated clinical trials to evaluate the safety and efficacy of senolytics in idiopathic pulmonary fibrosis, a lung disease associated with the presence of senescent cells. Another approach to senotherapeutics consists of controlling inflammation related to cellular senescence or "inflammaging", which participates, among other processes, in establishing pulmonary fibrosis. We evaluate whether polyphenols such as caffeic acid, chlorogenic acid, epicatechin, gallic acid, quercetin, or resveratrol combined with different senotherapeutics such as metformin or rapamycin, and antifibrotic drugs such as nintedanib or pirfenidone, could present beneficial actions in an in vitro model of senescent MRC-5 lung fibroblasts. A senescent-associated secretory phenotype (SASP) was evaluated by the measurement of interleukin (IL)-6, IL-8, and IL-1ß. The senescent-associated ß-galactosidase (SA-ß-gal) activity and cellular proliferation were assessed. Fibrosis was evaluated using a Picrosirius red assay and the gene expression of fibrosis-related genes. Epithelial-mesenchymal transition (EMT) was assayed in the A549 cell line exposed to Transforming Growth Factor (TGF)-ß in vitro. The combination that demonstrated the best results was metformin and caffeic acid, by inhibiting IL-6 and IL-8 in senescent MRC-5 cells. Metformin and caffeic acid also restore cellular proliferation and reduce SA-ß-gal activity during senescence induction. The collagen production by senescent MRC-5 cells was inhibited by epicatechin alone or combined with drugs. Epicatechin and nintedanib were able to control EMT in A549 cells. In conclusion, caffeic acid and epicatechin can potentially increase the effectiveness of senotherapeutic drugs in controlling lung diseases whose pathophysiological component is the presence of senescent cells and fibrosis.


Subject(s)
Cellular Senescence , Fibroblasts , Lung , Polyphenols , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , Cellular Senescence/drug effects , Polyphenols/pharmacology , Lung/pathology , Lung/drug effects , Lung/metabolism , A549 Cells , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Metformin/pharmacology , Caffeic Acids/pharmacology , Indoles/pharmacology , Senotherapeutics/pharmacology , Cell Line , Senescence-Associated Secretory Phenotype/drug effects , Sirolimus/pharmacology , Interleukin-8/metabolism , Interleukin-8/genetics , Transforming Growth Factor beta/metabolism , Pyridones
19.
Bioorg Chem ; 150: 107603, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38968905

ABSTRACT

Inhibition of LSD1 was proposed as promising and attractive therapies for treating osteoporosis. Here, we synthesized a series of novel TCP-(MP)-Caffeic acid analogs as potential LSD1 inhibitors to assess their inhibitory effects on osteoclastogenesis by using TRAP-staining assay and try to explore the preliminary SAR. Among them, TCP-MP-CA (11a) demonstrated osteoclastic bone loss both in vitro and in vivo, showing a significant improvement in the in vivo effects compared to the LSD1 inhibitor GSK-LSD1. Additionally, we elucidated a mechanism that 11a and its precursor that 11e directly bind to LSD1/CoREST complex through FAD to inhibit LSD1 demethylation activity and influence its downstream IκB/NF-κB signaling pathway, and thus regulate osteoclastic bone loss. These findings suggested 11a or 11e as potential novel candidates for treating osteoclastic bone loss, and a concept for further development of TCP-(MP)-Caffeic acid analogs for therapeutic use in osteoporosis clinics.


Subject(s)
Caffeic Acids , Osteoclasts , Osteoclasts/drug effects , Osteoclasts/metabolism , Caffeic Acids/pharmacology , Caffeic Acids/chemistry , Caffeic Acids/chemical synthesis , Animals , Structure-Activity Relationship , Mice , Molecular Structure , Dose-Response Relationship, Drug , Drug Discovery , Humans , Osteoporosis/drug therapy , Bone Resorption/drug therapy , RAW 264.7 Cells , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis
20.
Zhonghua Yu Fang Yi Xue Za Zhi ; 58(7): 1011-1019, 2024 Jul 06.
Article in Chinese | MEDLINE | ID: mdl-39034785

ABSTRACT

Objective: To investigate the effects and possible mechanisms of caffeic acid phenethyl ester (CAPE) on the replication, amplification, and fibre formation of prions (PrPSc). Methods: The CCK8 assay was used to detect the cell viability of the prion-infected cell model SMB-S15 after CAPE treatment for 3 days and 7 days and the maximum safe concentration of CAPE for SMB-S15 was obtained. The cells were treated with a concentration within a safe range, and the content of PrPSc in the cells before and after CAPE treatment was analyzed by western blot. Protein misfolding cycle amplification (PMCA) and western blot were used to assess changes in PrPSc level in amplification products following CAPE treatment. Real-time-quaking induced conversion assay (RT-QuIC) technology was employed to explore the changes in fibril formation before and after CAPE treatment. The binding affinity between CAPE and murine recombinant full-length prion protein was determined using a molecular interaction assay. Results: CCK8 cell viability assay results demonstrated that treatment with 1 µmol/L CAPE for 3 and 7 days did not exhibit statistically significant differences in cell viability compared to the control group (all P<0.05). However, when the concentration of CAPE exceeded 1 µmol/L, a significant reduction in cell viability was observed in cells treated with CAPE for 3 and 7 days, compared to the control group (all P<0.05). Thus, 1 µmol/L was determined as the maximum safe concentration of CAPE treatment for SMB-S15 cells. The western blot results revealed that treatment with CAPE for both 3 and 7 days led to a detectable reduction in the levels of PrPSc in SMB-S15 cells (all P<0.05). The products of PMCA experiments were assessed using western blot. The findings revealed a significant decrease in the levels of PrPSc (relative grey value) in the PMCA amplification products of adapted-strains SMB-S15, 139A, and ME7 following treatment with CAPE, as compared to the control group (all P<0.05). The RT-QuIC experimental results demonstrated a reduction in fibril formation (as indicated by ThT peak values) in CAPE-treated mouse-adapted strains 139A, ME7, and SMB-S15, as well as in SMB-S15 cells infected with prions. Furthermore, CAPE exhibited varying degrees of inhibition towards different seed fibrils formation, with statistically significant differences observed (all P<0.05). Notably, CAPE exhibited a more pronounced inhibitory effect on ME7 seed fibrils. Molecular interaction analyses demonstrated significant binding between CAPE and murine recombinant prion protein, and the association constant was (2.92±0.41)×10-6 mol/L. Conclusions: CAPE inhibits PrPSc replication, amplification, and fibril formation in vitro possibly due to specific interactions with the prion protein at the molecular level.


Subject(s)
Caffeic Acids , Phenylethyl Alcohol , Animals , Caffeic Acids/pharmacology , Mice , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Cell Survival/drug effects , PrPSc Proteins/metabolism , Prions , Cell Line , Prion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL