Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.821
Filter
1.
PeerJ ; 12: e17447, 2024.
Article in English | MEDLINE | ID: mdl-38832029

ABSTRACT

Objective: To investigate the effect of GnRH agonist (GnRH-a) down-regulation prior to hormone replacement treatment (HRT) to prepare the endometrium in frozen embryo transfer (FET) cycles in women of different ages. Methods: This was a retrospective study, and after excluding patients with adenomyosis, endometriosis, severe endometrial adhesions, polycystic ovary syndrome (PCOS), and repeated embryo implantation failures, a total of 4,091 HRT cycles were collected. Patients were divided into group A (<35 years old) and group B (≥35 years old), and each group was further divided into HRT and GnRHa-HRT groups. The clinical outcomes were compared between groups. Results: There was no statistically significant difference in clinical outcomes between the HRT and GnRHa-HRT groups among women aged <35 years. In women of advanced age, higher rates of clinical pregnancy and live birth were seen in the GnRHa-HRT group. Logistic regression analysis showed that female age and number of embryos transferred influenced the live birth rate in FET cycles, and in women aged ≥ 35 years, the use of GnRH-a down-regulation prior to HRT improved pregnancy outcomes. Conclusions: In elderly woman without adenomyosis, endometriosis, PCOS, severe uterine adhesions, and RIF, hormone replacement treatment with GnRH agonist for pituitary suppression can improve the live birth rate of FET cycles.


Subject(s)
Down-Regulation , Embryo Transfer , Gonadotropin-Releasing Hormone , Hormone Replacement Therapy , Humans , Female , Embryo Transfer/methods , Retrospective Studies , Adult , Gonadotropin-Releasing Hormone/agonists , Pregnancy , Down-Regulation/drug effects , Hormone Replacement Therapy/methods , Age Factors , Pregnancy Outcome/epidemiology , Pregnancy Rate , Embryo Implantation/drug effects
2.
PLoS One ; 19(5): e0302906, 2024.
Article in English | MEDLINE | ID: mdl-38718039

ABSTRACT

Osteoarthritis is the most prevalent type of degenerative arthritis. It is characterized by persistent pain, joint dysfunction, and physical disability. Pain relief and inflammation control are prioritised during osteoarthritis treatment Mume Fructus (Omae), a fumigated product of the Prunus mume fruit, is used as a traditional medicine in several Asian countries. However, its therapeutic mechanism of action and effects on osteoarthritis and articular chondrocytes remain unknown. In this study, we analyzed the anti-osteoarthritis and articular regenerative effects of Mume Fructus extract on rat chondrocytes. Mume Fructus treatment reduced the interleukin-1ß-induced expression of matrix metalloproteinase 3, matrix metalloproteinase 13, and a disintegrin and metalloproteinase with thrombospondin type 1 motifs 5. Additionally, it enhanced collagen type II alpha 1 chain and aggrecan accumulation in rat chondrocytes. Furthermore, Mume Fructus treatment regulated the inflammatory cytokine levels, mitogen-activated protein kinase phosphorylation, and nuclear factor-kappa B activation. Overall, our results demonstrated that Mume Fructus inhibits osteoarthritis progression by inhibiting the nuclear factor-kappa B and mitogen-activated protein kinase pathways to reduce the levels of inflammatory cytokines and prevent cartilage degeneration. Therefore, Mume Fructus may be a potential therapeutic option for osteoarthritis.


Subject(s)
Cartilage, Articular , Chondrocytes , Interleukin-1beta , NF-kappa B , Osteoarthritis , Plant Extracts , Animals , Chondrocytes/drug effects , Chondrocytes/metabolism , Interleukin-1beta/metabolism , Rats , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , NF-kappa B/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Plant Extracts/pharmacology , Prunus/chemistry , Rats, Sprague-Dawley , Down-Regulation/drug effects , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/genetics , Collagen Type II/metabolism , Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 3/genetics , Fruit/chemistry , Aggrecans/metabolism , ADAMTS5 Protein/metabolism , ADAMTS5 Protein/genetics , Cells, Cultured , Male , MAP Kinase Signaling System/drug effects
3.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 739-747, 2024 Apr 20.
Article in Chinese | MEDLINE | ID: mdl-38708508

ABSTRACT

OBJECTIVE: To explore the inhibitory effect of Sidaxue, a traditional Miao herbal medicine formula, on articular bone and cartilage destruction and synovial neovascularization in rats with collagen-induced arthritis (CIA). METHODS: In a SD rat model of CIA, we tested the effects of daily gavage of Sidaxue at low, moderate and high doses (10, 20, and 40 g/kg, respectively) for 21 days, with Tripterygium glycosides (GTW) as the positive control, on swelling in the hind limb plantar regions by arthritis index scoring. Pathologies in joint synovial membrane of the rats were observed with HE staining, and serum TNF-α and IL-1ß levels were detected with ELISA. The expressions of NF-κB p65, matrix metalloproteinase 1 (MMP1), MMP2 and MMP9 at the mRNA and protein levels in the synovial tissues were detected using real-time PCR and Western blotting. Network pharmacology analysis was conducted to identify the important target proteins in the pathways correlated with the therapeutic effects of topical Sidaxue treatment for RA, and the core target proteins were screened by topological analysis. RESULTS: Treatment with GTW and Sidaxue at the 3 doses all significantly alleviated plantar swelling, lowered arthritis index scores, improved cartilage and bone damage and reduced neovascularization in CIA rats (P<0.05), and the effects of Sidaxue showed a dose dependence. Both GTW and Sidaxue treatments significantly lowered TNF-α, IL-1ß, NF-κB p65, MMP1, MMP2, and MMP9 mRNA and protein expressions in the synovial tissues of CIA rats (P<0.05). Network pharmacological analysis identified MMPs as the core proteins associated with topical Sidaxue treatment of RA. CONCLUSION: Sidaxue alleviates articular bone and cartilage damages and reduces synovial neovascularization in CIA rats possibly by downregulating MMPs via the TNF-α/IL-1ß/NF-κB-MMP1, 2, 9 signaling pathway, and MMPs probably plays a key role in mediating the effect of Sidaxue though the therapeutic pathways other than oral administration.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Drugs, Chinese Herbal , Matrix Metalloproteinase 1 , Rats, Sprague-Dawley , Synovial Membrane , Tumor Necrosis Factor-alpha , Animals , Rats , Arthritis, Rheumatoid/drug therapy , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/pharmacology , Matrix Metalloproteinase 1/metabolism , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1beta/metabolism , Matrix Metalloproteinase 2/metabolism , Down-Regulation/drug effects , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/metabolism , Tripterygium/chemistry , Transcription Factor RelA/metabolism
4.
Drug Des Devel Ther ; 18: 1531-1546, 2024.
Article in English | MEDLINE | ID: mdl-38737331

ABSTRACT

Purpose: Lung adenocarcinoma currently ranks the leading causes of cancer-related mortality worldwide. Many anti-inflammation herbs, like tetramethylpyrazine, have shown their anti-tumor potentials. Here, we evaluated the role of a novel chalcone derivative of tetramethylpyrazine ((E) -1- (E) -1- (2-hydroxy-5-chlorophenyl) -3- (3,5,6-trimethylpyrazin-2-yl) -2-propen-1, HCTMPPK) in lung adenocarcinoma. Methods: The effects of HCTMPPK on cell proliferation, apoptosis, and invasion were investigated by in-vitro assays, including CCK-8, colony formation assay, flow cytometry, transwell assay, and wound-healing assay. The therapeutic potential of HCTMPPK in vivo was evaluated in xenograft mice. To figure out the target molecules of HCTMPPK, a network pharmacology approach and molecular docking studies were employed, and subsequent experiments were conducted to confirm these candidate molecules. Results: HCTMPPK effectively suppressed the proliferative activity and migration, as well as enhanced the apoptosis of A549 cells in a concentration-dependent manner. Consistent with this, tumor growth was inhibited by HCTMPPK significantly in vivo. Regarding the mechanisms, HCTMPPK down-regulated Bcl-2 and MMP-9 and up-regulating Bax and cleaved-caspase-3. Subsequently, we identified 601 overlapping DEGs from LUAD patients in TCGA and GEO database. Then, 15 hub genes were identified by PPI network and CytoHubba. Finally, MELK was verified to be the HCTMPPK targeted site, through the molecular docking studies and validation experiments. Conclusion: Overall, our study indicates HCTMPPK as a potential MELK inhibitor and may be a promising candidate for the therapy of lung cancer.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Proliferation , Down-Regulation , Drug Screening Assays, Antitumor , Lung Neoplasms , Pyrazines , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Pyrazines/pharmacology , Pyrazines/chemistry , Cell Proliferation/drug effects , Animals , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Down-Regulation/drug effects , Chalcone/pharmacology , Chalcone/chemistry , Molecular Structure , Dose-Response Relationship, Drug , Structure-Activity Relationship , Molecular Docking Simulation , Mice, Nude , Mice, Inbred BALB C , A549 Cells , Cell Movement/drug effects , Chalcones/pharmacology , Chalcones/chemistry , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Neoplasms, Experimental/metabolism , Tumor Cells, Cultured
5.
Cells ; 13(10)2024 May 11.
Article in English | MEDLINE | ID: mdl-38786044

ABSTRACT

Breast cancer includes tumor subgroups with morphological, molecular, and clinical differences. Intrinsic heterogeneity especially characterizes breast tumors with a triple negative phenotype, often leading to the failure of even the most advanced therapeutic strategies. To improve breast cancer treatment, the use of natural agents to integrate conventional therapies is the subject of ever-increasing attention. In this context, garlic (Allium sativum) shows anti-cancerous potential, interfering with the proliferation, motility, and malignant progression of both non-invasive and invasive breast tumor cells. As heterogeneity could be at the basis of variable effects, the main objective of our study was to evaluate the anti-tumoral activity of a garlic extract in breast cancer cells with a triple negative phenotype. Established triple negative breast cancer (TNBC) cell lines from patient-derived xenografts (PDXs) were used, revealing subtype-dependent effects on morphology, cell cycle, and invasive potential, correlated with the peculiar down-modulation of Akt signaling, a crucial regulator in solid tumors. Our results first demonstrate that the effects of garlic on TNBC breast cancer are not unique and suggest that only more precise knowledge of the mechanisms activated by this natural compound in each tumor will allow for the inclusion of garlic in personalized therapeutic approaches to breast cancer.


Subject(s)
Garlic , Proto-Oncogene Proteins c-akt , Signal Transduction , Triple Negative Breast Neoplasms , Humans , Garlic/chemistry , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Female , Cell Line, Tumor , Animals , Phenotype , Cell Proliferation/drug effects , Mice , Plant Extracts/pharmacology , Down-Regulation/drug effects , Cell Movement/drug effects , Xenograft Model Antitumor Assays
6.
J Ethnopharmacol ; 331: 118284, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38735420

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ginkgo biloba leaf and seed have been traditionally used in ancient China for the treatment of cough and asthma. However, there is limited literature available on the anti-COPD effects and mechanisms of Ginkgo biloba. AIMS OF THE STUDY: The aim of this study was to comprehensively investigate the therapeutic potential of ginkgo extracts in COPD through a combination of in vivo and in vitro functional experiments. Transcriptomic analyses were also employed to uncover novel molecular mechanisms underlying the therapeutic effects of ginkgetin in COPD. MATERIALS AND METHODS: The therapeutic efficacy of ginkgo extracts was assessed in a COPD model. The anti-inflammatory effects of ginkgetin and its underlying molecular mechanisms were examined in A549 cells treated with cigarette smoke extract (CSE). Additionally, transcriptomic analyses were conducted to identify novel molecular pathways influenced by ginkgetin. These findings were further validated using quantitative real-time polymerase chain reaction (qPCR) and Western blot techniques. RESULTS: The ethyl acetate extract of Ginkgo biloba L. seeds and ginkgetin treatment significantly reduced cytokine production in COPD mice. Following drug administration, lung function improved in different groups. The transcriptome data strongly supports the inhibitory effect of ginkgetin on CSE-induced inflammation through the downregulation of the c/EBPß signaling pathway and subsequent inhibition of CCL2 expression. CONCLUSION: Our results demonstrate that ginkgetin, one of the biflavones found in Ginkgo biloba, exhibits inhibitory effects on smoke-induced airway inflammation. This effect is achieved through the downregulation of the c/EBPß signaling pathway and the reduction of CCL2 expression.


Subject(s)
Biflavonoids , Chemokine CCL2 , Down-Regulation , Ginkgo biloba , Pulmonary Disease, Chronic Obstructive , Signal Transduction , Animals , Pulmonary Disease, Chronic Obstructive/drug therapy , Biflavonoids/pharmacology , Biflavonoids/therapeutic use , Humans , Signal Transduction/drug effects , Ginkgo biloba/chemistry , Down-Regulation/drug effects , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Mice , Male , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Smoke/adverse effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , A549 Cells , Mice, Inbred C57BL , Disease Models, Animal , Lung/drug effects , Lung/metabolism , Lung/pathology , Ginkgo Extract
7.
Cell Death Dis ; 15(5): 322, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719798

ABSTRACT

Metastatic dissemination from the primary tumor is a complex process that requires crosstalk between tumor cells and the surrounding milieu and involves the interplay between numerous cellular-signaling programs. Epithelial-mesenchymal transition (EMT) remains at the forefront of orchestrating a shift in numerous cellular programs, such as stemness, drug resistance, and apoptosis that allow for successful metastasis. Till date, there is limited success in therapeutically targeting EMT. Utilizing a high throughput screen of FDA-approved compounds, we uncovered a novel role of the topoisomerase inhibitor, Teniposide, in reversing EMT. Here, we demonstrate Teniposide as a potent modulator of the EMT program, specifically through an IRF7-NMI mediated response. Furthermore, Teniposide significantly reduces the expression of the key EMT transcriptional regulator, Zinc Finger E-Box Binding Homeobox 2 (ZEB2). ZEB2 downregulation by Teniposide inhibited RNA polymerase I (Pol I) activity and rRNA biogenesis. Importantly, Teniposide treatment markedly reduced pulmonary colonization of breast cancer cells. We have uncovered a novel role of Teniposide, which when used at a very low concentration, mitigates mesenchymal-like invasive phenotype. Overall, its ability to target EMT and rRNA biogenesis makes Teniposide a viable candidate to be repurposed as a therapeutic option to restrict breast cancer metastases.


Subject(s)
Breast Neoplasms , Down-Regulation , Epithelial-Mesenchymal Transition , RNA Polymerase I , Teniposide , Zinc Finger E-box Binding Homeobox 2 , Epithelial-Mesenchymal Transition/drug effects , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Female , Zinc Finger E-box Binding Homeobox 2/metabolism , Zinc Finger E-box Binding Homeobox 2/genetics , Cell Line, Tumor , Down-Regulation/drug effects , RNA Polymerase I/metabolism , Teniposide/pharmacology , Animals , Mice , Gene Expression Regulation, Neoplastic/drug effects
8.
Chemosphere ; 359: 142332, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38754493

ABSTRACT

Perfluorooctanesulfonic acid (PFOS) is a widely recognized environment pollutant known for its high bioaccumulation potential and a long elimination half-life. Several studies have shown that PFOS can alter multiple biological pathways and negatively affect human health. Considering the direct exposure to the gastrointestinal (GI) tract to environmental pollutants, PFOS can potentially disrupt intestinal homeostasis. However, there is limited knowledge about the effect of PFOS exposure on normal intestinal tissues, and its contribution to GI-associated diseases remains to be determined. In this study, we examined the effect of PFOS exposure on the gene expression profile of intestinal tissues of C57BL/6 mice using RNAseq analysis. We found that PFOS exposure in drinking water significantly downregulates mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2), a rate-limiting ketogenic enzyme, in intestinal tissues of mice. We found that diets containing the soluble fibers inulin and pectin, which are known to be protective against PFOS exposure, were ineffective in reversing the downregulation of HMGCS2 expression in vivo. Analysis of intestinal tissues also demonstrated that PFOS exposure leads to upregulation of proteins implicated in colorectal carcinogenesis, including ß-catenin, c-MYC, mTOR and FASN. Consistent with the in vivo results, PFOS exposure leads to downregulation of HMGCS2 in mouse and human normal intestinal organoids in vitro. Furthermore, we show that shRNA-mediated knockdown of HMGCS2 in a human normal intestinal cell line resulted in increased cell proliferation and upregulation of key proliferation-associated proteins such as cyclin D, survivin, ERK1/2 and AKT, along with an increase in lipid accumulation. In summary, our results suggest that PFOS exposure may contribute to pathological changes in normal intestinal cells via downregulation of HMGCS2 expression and upregulation of pro-carcinogenic signaling pathways that may increase the risk of colorectal cancer development.


Subject(s)
Alkanesulfonic Acids , Carcinogenesis , Down-Regulation , Fluorocarbons , Hydroxymethylglutaryl-CoA Synthase , Mice, Inbred C57BL , Animals , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Hydroxymethylglutaryl-CoA Synthase/metabolism , Hydroxymethylglutaryl-CoA Synthase/genetics , Mice , Down-Regulation/drug effects , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Up-Regulation/drug effects , Environmental Pollutants/toxicity , Intestines/drug effects , Humans , Intestinal Mucosa/metabolism
9.
Int Immunopharmacol ; 134: 112175, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38733821

ABSTRACT

BACKGROUND: Our previous study has revealed that asiaticoside (AC) promotes endoplasmic reticulum stress and antagonizes proliferation and migration of gastric cancer (GC) via miR-635/HMGA1 axis. However, the effect and mechanism of AC on other progressions of GC, such as ferroptosis and immune escape, are still unknown. METHODS: AGS and HGC27 cells were incubated with 1, 2 and 4 µM of AC for 24 h. Mice xenografted with AGS cells were intragastrically injected with AC. The effect and mechanism of AC on GC were determined by the measurement of the ferrous iron level, the ROS level and the glutathione peroxidase (GSH) content, flow cytometry, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry and western blotting assays. RESULTS: AC increased the Fe2+ level and the ROS level, but decreased the expression of GPX4 and SLC7A11 and the GSH level. Besides, AC enhanced the percent of CD8+ T cells and the IFN-γ concentration, but reduced the PD-L1 expression and the IL-10 level. Mechanically, AC downregulated the relative levels of ß-catenin, active-ß-catenin, p-GSK3ß/GSK3ß, cyclin D1 and c-Myc in GC cells, which were rescued with the application of LiCl (an activator of Wnt/ß-catenin pathway) in AGS cells. Moreover, activation of Wnt/ß-catenin pathway by LiCl or the ß-catenin overexpression inverted the effect of AC on ferroptosis and immune escape in GC cells. In vivo, AC treatment declined the tumor size and weight, the level of GPX4, SLC7A11, PD-L1 and IFN-γ, and the expression of Wnt/ß-catenin pathway. CONCLUSION: AC enhanced ferroptosis and repressed immune escape by downregulating the Wnt/ß-catenin signaling in GC.


Subject(s)
Down-Regulation , Ferroptosis , Stomach Neoplasms , Triterpenes , Tumor Escape , Wnt Signaling Pathway , Ferroptosis/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Animals , Humans , Wnt Signaling Pathway/drug effects , Cell Line, Tumor , Triterpenes/pharmacology , Triterpenes/therapeutic use , Tumor Escape/drug effects , Mice , Down-Regulation/drug effects , Mice, Inbred BALB C , beta Catenin/metabolism , Xenograft Model Antitumor Assays , Mice, Nude , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Male , Reactive Oxygen Species/metabolism , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects
10.
Biomed Pharmacother ; 175: 116736, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38739992

ABSTRACT

AIMS: The xanthone dimer 12-O-deacetyl-phomoxanthone A (12-ODPXA) was extracted from the secondary metabolites of the endophytic fungus Diaporthe goulteri. The 12-ODPXA compound exhibited anticancer properties in murine lymphoma; however, the anti-ovarian cancer (OC) mechanism has not yet been explored. Therefore, the present study evaluated whether 12-ODPXA reduces OC cell proliferation, metastasis, and invasion by downregulating pyruvate dehydrogenase kinase (PDK)4 expression. METHODS: Cell counting kit-8, colony formation, flow cytometry, wound healing, and transwell assays were performed to examine the effects of 12-ODPXA on OC cell proliferation, apoptosis, migration, and invasion. Transcriptome analysis was used to predict the changes in gene expression. Protein expression was determined using western blotting. Glucose, lactate, and adenosine triphosphate (ATP) test kits were used to measure glucose consumption and lactate and ATP production, respectively. Zebrafish xenograft models were constructed to elucidate the anti-OC effects of 12-ODPXA. RESULTS: The 12-ODPXA compound inhibited OC cell proliferation, migration, invasion, and glycolysis while inducing cell apoptosis via downregulation of PDK4. In vivo experiments showed that 12-ODPXA suppressed tumor growth and migration in zebrafish. CONCLUSION: Our data demonstrate that 12-ODPXA inhibits ovarian tumor growth and metastasis by downregulating PDK4, revealing the underlying mechanisms of action of 12-ODPXA in OC.


Subject(s)
Apoptosis , Cell Movement , Cell Proliferation , Down-Regulation , Ovarian Neoplasms , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Xanthones , Zebrafish , Animals , Female , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Humans , Xanthones/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Cell Movement/drug effects , Apoptosis/drug effects , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Xenograft Model Antitumor Assays/methods , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Metastasis , Neoplasm Invasiveness
11.
Biomed Pharmacother ; 175: 116725, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744219

ABSTRACT

Qualitative alterations in type I collagen due to pathogenic variants in the COL1A1 or COL1A2 genes, result in moderate and severe Osteogenesis Imperfecta (OI), a rare disease characterized by bone fragility. The TGF-ß signaling pathway is overactive in OI patients and certain OI mouse models, and inhibition of TGF-ß through anti-TGF-ß monoclonal antibody therapy in phase I clinical trials in OI adults is rendering encouraging results. However, the impact of TGF-ß inhibition on osteogenic differentiation of mesenchymal stem cells from OI patients (OI-MSCs) is unknown. The following study demonstrates that pediatric skeletal OI-MSCs have imbalanced osteogenesis favoring the osteogenic commitment. Galunisertib, a small molecule inhibitor (SMI) that targets the TGF-ß receptor I (TßRI), favored the final osteogenic maturation of OI-MSCs. Mechanistically, galunisertib downregulated type I collagen expression in OI-MSCs, with greater impact on mutant type I collagen, and concomitantly, modulated the expression of unfolded protein response (UPR) and autophagy markers. In vivo, galunisertib improved trabecular bone parameters only in female oim/oim mice. These results further suggest that type I collagen is a tunable target within the bone ECM that deserves investigation and that the SMI, galunisertib, is a promising new candidate for the anti-TGF-ß targeting for the treatment of OI.


Subject(s)
Collagen Type I , Down-Regulation , Mesenchymal Stem Cells , Osteogenesis Imperfecta , Osteogenesis , Pyrazoles , Quinolines , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/drug therapy , Osteogenesis/drug effects , Osteogenesis/genetics , Animals , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Down-Regulation/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Female , Quinolines/pharmacology , Mice , Child , Pyrazoles/pharmacology , Male , Cell Differentiation/drug effects , Mutation , Disease Models, Animal , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Child, Preschool , Cells, Cultured , Transforming Growth Factor beta/metabolism , Unfolded Protein Response/drug effects , Signal Transduction/drug effects
12.
Biomed Pharmacother ; 175: 116734, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38754264

ABSTRACT

Spinal cord injury (SCI) is a type of central nervous system (CNS) injury in which ferroptosis is becoming a promising target for treatment. Alpha-tocopherol (Vitamin E, Vit E) is a compound with anti-ferroptosis activity. The mechanism of alpha-tocopherol in regulating ferroptosis after SCI has not been deeply studied. In this study, rats with SCI were treated by Alpha-tocopherol based on bioinformatic analysis and molecular docking prediction. Behavioral tests and histological findings showed that Alpha-tocopherol promoted neural function recovery and tissue repairment in rats with SCI. Subsequently, regulatory effects of Alpha-tocopherol on Alox15 and ferroptosis were detected and then localized by immunofluorescence. In vitro, alpha-tocopherol improved the ROS accumulation, iron overload, lipid peroxidation and mitochondrial dysfunction. The effects of Alpha-tocopherol on the expression of Alox15, Ptgs2 and 4Hne were validated in vitro. Finally, the inhibitory effects of Alpha-tocopherol on Alox15 and ferroptosis were weakened by the mutation of 87th residue of Alox15. In summary, alpha-tocopherol could alleviate SCI-induced ferroptosis by downregulating Alox15 to promote neural function recovery in rats with SCI. Findings in this study could help further our understanding on SCI-induced ferroptosis and provide a novel insight for treating SCI.


Subject(s)
Arachidonate 15-Lipoxygenase , Down-Regulation , Ferroptosis , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord Injuries , alpha-Tocopherol , Animals , Ferroptosis/drug effects , alpha-Tocopherol/pharmacology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Recovery of Function/drug effects , Down-Regulation/drug effects , Rats , Arachidonate 15-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Lipid Peroxidation/drug effects , Male , Reactive Oxygen Species/metabolism , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 12-Lipoxygenase/genetics , Disease Models, Animal , Molecular Docking Simulation
13.
Eur J Med Chem ; 272: 116460, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38704943

ABSTRACT

It has been reported that 4,5-dihydropyrazole and thiazole derivatives have many biological functions, especially in the aspect of anti-inflammation. According to the strategy of pharmacophore combination, we introduced thiazolinone and dihydropyrazole moiety into steroid skeleton to design and synthesize a novel series of D-ring substituted steroidal 4,5-dihydropyrazole thiazolinone derivatives, and assessed their in vitro anti-inflammatory profiles against Lipopolysaccharide (LPS)-induced inflammation in RAW 264.7 macrophage cells. The anti-inflammatory activities assay demonstrated that compound 12e was considered as the most effective anti-inflammatory drug, which suppressed the expression of pro-inflammatory mediators including nitric oxide (NO), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), it also dose-dependently inhibited the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in LPS-induced RAW 264.7 macrophage cells. Furthermore, the results of the Western blot analysis showed a correlation between the inhibition of the Nuclear factor-kappa B (NF-κB) and Mitogen-activated protein kinases (MAPKs) signaling pathways and the suppressive effects of compound 12e on pro-inflammatory cytokines. Molecular docking studies of compound 12e into the COX-2 protein receptor (PDB ID: 5IKQ) active site was performed to rationalize their COX-2 inhibitory potency. The results were found to be in line with the biological findings as they exerted more favorable interactions compared to that of dexamethasone (DXM), explaining their remarkable COX-2 inhibitory activity. The findings revealed that these candidates could be identified as potent anti-inflammatory agents, compound 12e could be a promising drug for the treatment of inflammatory diseases.


Subject(s)
Cyclooxygenase 2 , Down-Regulation , Drug Design , Lipopolysaccharides , Macrophages , NF-kappa B , Nitric Oxide Synthase Type II , Pyrazoles , Animals , Mice , Lipopolysaccharides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , RAW 264.7 Cells , Cyclooxygenase 2/metabolism , NF-kappa B/metabolism , NF-kappa B/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Structure-Activity Relationship , Pyrazoles/pharmacology , Pyrazoles/chemistry , Pyrazoles/chemical synthesis , Macrophages/drug effects , Macrophages/metabolism , Down-Regulation/drug effects , Molecular Structure , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Models, Molecular , Dose-Response Relationship, Drug , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/chemistry , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Thiazoles/pharmacology , Thiazoles/chemistry , Thiazoles/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Steroids/pharmacology , Steroids/chemistry , Steroids/chemical synthesis , Molecular Docking Simulation
14.
Molecules ; 29(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731499

ABSTRACT

Carbon nanodots (CDs) are commonly found in food products and have attracted significant attention from food scientists. There is a high probability of CD exposure in humans, but its impacts on health are unclear. Therefore, health effects associated with CD consumption should be investigated. In this study, we attempted to create a model system of the Maillard reaction between cystine and glucose using a simple cooking approach. The CDs (CG-CDs) were isolated from cystine-glucose-based Maillard reaction products and characterized using fluorescence spectroscopy, X-ray diffractometer (XRD), and transmission electron microscope (TEM). Furthermore, human mesenchymal stem cells (hMCs) were used as a model to unravel the CDs' cytotoxic properties. The physiochemical assessment revealed that CG-CDs emit excitation-dependent fluorescence and possess a circular shape with sizes ranging from 2 to 13 nm. CG-CDs are predominantly composed of carbon, oxygen, and sulfur. The results of the cytotoxicity evaluation indicate good biocompatibility, where no severe toxicity was observed in hMCs up to 400 µg/mL. The DPPH assay demonstrated that CDs exert potent antioxidant abilities. The qPCR analysis revealed that CDs promote the downregulation of the key regulatory genes, PPARγ, C/EBPα, SREBP-1, and HMGCR, coupled with the upregulation of anti-inflammatory genes. Our findings suggested that, along with their excellent biocompatibility, CG-CDs may offer positive health outcomes by modulating critical genes involved in lipogenesis, homeostasis, and obesity pathogenesis.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha , Carbon , Maillard Reaction , Mesenchymal Stem Cells , PPAR gamma , Sterol Regulatory Element Binding Protein 1 , Humans , Carbon/chemistry , PPAR gamma/genetics , PPAR gamma/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , Quantum Dots/chemistry , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Antioxidants/pharmacology , Antioxidants/chemistry , Sulfur/chemistry
15.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731935

ABSTRACT

Cancer treatment is greatly challenged by drug resistance, highlighting the need for novel drug discoveries. Here, we investigated novel organoarsenic compounds regarding their resistance-breaking and apoptosis-inducing properties in leukemia and lymphoma. Notably, the compound (2,6-dimethylphenyl)arsonic acid (As2) demonstrated significant inhibition of cell proliferation and induction of apoptosis in leukemia and lymphoma cells while sparing healthy leukocytes. As2 reached half of its maximum activity (AC50) against leukemia cells at around 6.3 µM. Further experiments showed that As2 overcomes multidrug resistance and sensitizes drug-resistant leukemia and lymphoma cell lines to treatments with the common cytostatic drugs vincristine, daunorubicin, and cytarabine at low micromolar concentrations. Mechanistic investigations of As2-mediated apoptosis involving FADD (FAS-associated death domain)-deficient or Smac (second mitochondria-derived activator of caspases)/DIABLO (direct IAP binding protein with low pI)-overexpressing cell lines, western blot analysis of caspase-9 cleavage, and measurements of mitochondrial membrane integrity identified the mitochondrial apoptosis pathway as the main mode of action. Downregulation of XIAP (x-linked inhibitor of apoptosis protein) and apoptosis induction independent of Bcl-2 (B-cell lymphoma 2) and caspase-3 expression levels suggest the activation of additional apoptosis-promoting mechanisms. Due to the selective apoptosis induction, the synergistic effects with common anti-cancer drugs, and the ability to overcome multidrug resistance in vitro, As2 represents a promising candidate for further preclinical investigations with respect to refractory malignancies.


Subject(s)
Apoptosis , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Leukemia , Lymphoma , Mitochondria , X-Linked Inhibitor of Apoptosis Protein , X-Linked Inhibitor of Apoptosis Protein/metabolism , Humans , Apoptosis/drug effects , Drug Resistance, Neoplasm/drug effects , Lymphoma/drug therapy , Lymphoma/metabolism , Lymphoma/pathology , Leukemia/metabolism , Leukemia/drug therapy , Leukemia/pathology , Drug Resistance, Multiple/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Cell Proliferation/drug effects , Cytostatic Agents/pharmacology , Antineoplastic Agents/pharmacology
16.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791386

ABSTRACT

Metastasis in breast cancer is the major cause of death in females (about 30%). Based on our earlier observation that Vitamin D3 downregulates mTOR, we hypothesized that Vitamin D3 conjugated to gold nanoparticles (VD3-GNPs) reduces breast cancer aggressiveness by downregulating the key cancer controller PI3K/AKT/mTOR. Western blots, migration/invasion assays, and other cell-based, biophysical, and bioinformatics studies are used to study breast cancer cell aggressiveness and nanoparticle characterization. Our VD3-GNP treatment of breast cancer cells (MCF-7 and MDA-MB-231) significantly reduces the aggressiveness (cancer cell migration and invasion rates > 45%) via the simultaneous downregulation of ETV7 and the Hippo pathway. Consistent with our hypothesis, we, indeed, found a downregulation of the PI3K/AKT/mTOR pathway. It is surprising that the extremely low dose of VD3 in the nano formulation (three orders of magnitude lower than in earlier studies) is quite effective in the alteration of cancer invasiveness and cell signaling pathways. Clearly, VD3-GNPs are a viable candidate for non-toxic, low-cost treatment for reducing breast cancer aggressiveness.


Subject(s)
Breast Neoplasms , Cholecalciferol , Gold , Metal Nanoparticles , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases , Signal Transduction , TOR Serine-Threonine Kinases , Humans , TOR Serine-Threonine Kinases/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Female , Phosphatidylinositol 3-Kinases/metabolism , Metal Nanoparticles/chemistry , Signal Transduction/drug effects , Gold/chemistry , Cholecalciferol/pharmacology , Cell Movement/drug effects , Hippo Signaling Pathway , Protein Serine-Threonine Kinases/metabolism , Cell Line, Tumor , MCF-7 Cells , Transcription Factors/metabolism , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Proto-Oncogene Proteins c-akt/metabolism
17.
Int J Mol Sci ; 25(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38791190

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) is more prevalent in post- compared to pre-menopausal women. The underlying mechanisms are not fully understood. Data in humans is confounded by age and co-morbidities. We investigated the effects of ovariectomy and estrogen replacement on the left ventricular (LV) gene expression of pro-inflammatory and pro-fibrotic factors involved in HFpEF and putative regulating miRNAs. Nine-week-old C57BL/6 female mice were subjected to ovariectomy (OVX) or SHAM operation. OVX and SHAM groups were sacrificed 1-, 6-, and 12-weeks post-surgery (T1/SHAM; T1/OVX; T6/SHAM; T6/OVX, T12/SHAM). 17ß-estradiol (E2) or vehicle (VEH) was then administered to the OVX groups for 6 weeks (T12/OVX/E2; T12/OVX/VEH). Another SHAM group was sacrificed 12-weeks post-surgery. RNA and miRNAs were extracted from the LV apex. An early 3-fold increase in the gene expression of IL-1α, IL-6, Mmp9, Mmp12, Col1α1, and Col3α1 was observed one-week post-surgery in T1/OVX vs. T1/SHAM, but not at later time points. miRNA-26a was lower in T1/OVX vs. T1/SHAM and was inversely correlated with Col1α1 and Col3α1 expression 1-week post-surgery (r = -0.79 p < 0.001; r = -0.6 p = 0.007). miRNAs-26a, 29b, and 133a were significantly higher, while Col1α1, Col3α1, IL-1α, IL-6, Tnfα, Mmp12, and FasL gene expression was significantly lower in E2- compared to vehicle-treated OVX mice. miRNA-26a was inversely correlated with Col3α1 in T12/OVX/ E2 (r = -0.56 p = 0.02). OVX triggered an early increase in the gene expression of pro-inflammatory and pro-fibrotic factors, highlighting the importance of the early phase post-cessation of ovarian function. E2 replacement therapy, even if it was not immediately initiated after OVX, reversed these unfavorable changes and upregulated cardiac miRNA-26a, previously unknown to be affected by menopausal status.


Subject(s)
Collagen Type I , Estradiol , Heart Ventricles , Mice, Inbred C57BL , MicroRNAs , Ovariectomy , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Female , Estradiol/pharmacology , Mice , Collagen Type I/genetics , Collagen Type I/metabolism , Heart Ventricles/metabolism , Heart Ventricles/drug effects , Collagen Type III/genetics , Collagen Type III/metabolism , Gene Expression Regulation/drug effects , Down-Regulation/drug effects , Heart Failure/genetics , Heart Failure/metabolism , Collagen Type I, alpha 1 Chain/metabolism , Up-Regulation/drug effects , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Estrogen Replacement Therapy
18.
Int J Mol Sci ; 25(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38791186

ABSTRACT

Malignant melanoma represents a form of skin cancer characterized by a bleak prognosis and heightened resistance to traditional therapies. Quercetin has demonstrated notable anti-carcinogenic, anti-inflammatory, anti-oxidant, and pharmacological effects across various cancer types. However, the intricate relationship between quercetin's anti-cancer properties and ganglioside expression in melanoma remains incompletely understood. In this study, quercetin manifests specific anti-proliferative, anti-migratory, and cell-cycle arrest effects, inducing mitochondrial dysfunction and apoptosis in two melanoma cancer cell lines. This positions quercetin as a promising candidate for treating malignant melanoma. Moreover, our investigation indicates that quercetin significantly reduces the expression levels of ganglioside GD3 and its synthetic enzyme. Notably, this reduction is achieved through the inhibition of the FAK/paxillin/Akt signaling pathway, which plays a crucial role in cancer development. Taken together, our findings suggest that quercetin may be a potent anti-cancer drug candidate for the treatment of malignant melanoma.


Subject(s)
Apoptosis , Gangliosides , Melanoma , Mitochondria , Quercetin , Quercetin/pharmacology , Humans , Melanoma/metabolism , Melanoma/drug therapy , Melanoma/pathology , Apoptosis/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Cell Line, Tumor , Gangliosides/metabolism , Cell Proliferation/drug effects , Signal Transduction/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Cell Movement/drug effects , Skin Neoplasms/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism
19.
Mol Med Rep ; 30(1)2024 07.
Article in English | MEDLINE | ID: mdl-38757304

ABSTRACT

Gut microbiota dysfunction is a key factor affecting chronic kidney disease (CKD) susceptibility. Puerariae lobatae Radix (PLR), a traditional Chinese medicine and food homologous herb, is known to promote the gut microbiota homeostasis; however, its role in renoprotection remains unknown. The present study aimed to investigate the efficacy and potential mechanism of PLR to alleviate CKD. An 8­week 2% NaCl­feeding murine model was applied to induce CKD and evaluate the therapeutic effect of PLR supplementary. After gavage for 8 weeks, The medium and high doses of PLR significantly alleviated CKD­associated creatinine, urine protein increasement and nephritic histopathological injury. Moreover, PLR protected kidney from fibrosis by reducing inflammatory response and downregulating the canonical Wnt/ß­catenin pathway. Furthermore, PLR rescued the gut microbiota dysbiosis and protected against high salt­induced gut barrier dysfunction. Enrichment of Akkermansia and Bifidobacterium was found after PLR intervention, the relative abundances of which were in positive correlation with normal maintenance of renal histology and function. Next, fecal microbiota transplantation experiment verified that the positive effect of PLR on CKD was, at least partially, exerted through gut microbiota reestablishment and downregulation of the Wnt/ß­catenin pathway. The present study provided evidence for a new function of PLR on kidney protection and put forward a potential therapeutic strategy target for CKD.


Subject(s)
Drugs, Chinese Herbal , Gastrointestinal Microbiome , Pueraria , Renal Insufficiency, Chronic , Wnt Signaling Pathway , Gastrointestinal Microbiome/drug effects , Animals , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Wnt Signaling Pathway/drug effects , Mice , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Male , Pueraria/chemistry , Disease Models, Animal , Dysbiosis/drug therapy , Down-Regulation/drug effects , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Mice, Inbred C57BL , Fecal Microbiota Transplantation
20.
PLoS One ; 19(5): e0301966, 2024.
Article in English | MEDLINE | ID: mdl-38776280

ABSTRACT

The purpose of this study is to assess the bioactive peptides derived from the defatted lemon basil seeds hydrolysate (DLSH) for their ability to inhibit pancreatic lipase, decrease intracellular lipid accumulation, and reduce adipogenesis. Response surface methodology (RSM) was employed to optimize trypsin hydrolysis conditions for maximizing lipase inhibitory activity (LI). A hydrolysis time of 387.06 min, a temperature of 49.03°C, and an enzyme concentration of 1.61% w/v, resulted in the highest LI with an IC50 of 368.07 µg/mL. The ultrafiltration of the protein hydrolysate revealed that the fraction below 0.65kDa exhibited the greatest LI potential. Further purification via RP-HPLC identified the Gly-Arg-Ser-Pro-Asp-Thr-His-Ser-Gly (GRSPDTHSG) peptide in the HPLC fraction F1 using mass spectrometry. The peptide was synthesized and demonstrated LI with an IC50 of 0.255 mM through a non-competitive mechanism, with a constant (Ki) of 0.61 mM. Docking studies revealed its binding site with the pancreatic lipase-colipase complex. Additionally, GRSPDTHSG inhibited lipid accumulation in 3T3-L1 cells in a dose-dependent manner without cytotoxic effects. Western blot analysis indicated downregulation of PPAR-γ and SREBP-1c levels under GRSPDTHSG treatment, while an increase in AMPK-α phosphorylation was observed, suggesting a role in regulating cellular lipid metabolism. Overall, GRSPDTHSG demonstrates potential in attenuating lipid absorption and adipogenesis, suggesting a prospective application in functional foods and nutraceuticals.


Subject(s)
3T3-L1 Cells , Adipocytes , Adipogenesis , Lipase , Ocimum basilicum , PPAR gamma , Peptides , Seeds , Sterol Regulatory Element Binding Protein 1 , Mice , Animals , Adipogenesis/drug effects , Seeds/chemistry , PPAR gamma/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Hydrolysis , Lipase/antagonists & inhibitors , Lipase/metabolism , Peptides/pharmacology , Peptides/chemistry , Peptides/isolation & purification , Sterol Regulatory Element Binding Protein 1/metabolism , Ocimum basilicum/chemistry , Down-Regulation/drug effects , Molecular Docking Simulation
SELECTION OF CITATIONS
SEARCH DETAIL
...