1
|
Huang J, Yu H, Yuan X, Zhong Y, Li X, Chen Y. TCN1 as an inflammatory regulator in psoriasis: Activation of the NF-κB pathway and potential therapeutic target. Int Immunopharmacol 2025; 157:114784. [PMID: 40318273 DOI: 10.1016/j.intimp.2025.114784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVE This study investigates how TCN1 regulates inflammation and the cell cycle in psoriasis, focusing on the NF-κB pathway through in vitro experiments and bioinformatics analyses. METHODS DEGs were identified by analyzing transcriptome data from four datasets comparing psoriatic lesions and normal skin (GSE34248, GSE30999, GSE14905, and GSE13355). Validation of TCN1 expression following biologic treatment was conducted using GSE201827, GSE51440, and GSE117239. GO and GSEA were performed to explore biological pathways. The expression levels of TCN1 in psoriatic lesions and healthy skin were assessed by qPCR and immunohistochemistry (IHC). In vitro, HaCaT keratinocytes were stimulated with TNF-α and IL-17 A, and TCN1 expression was modulated through siRNA-mediated knockdown and plasmid-mediated overexpression. Subsequent changes in TCN1 and key inflammatory cytokines were evaluated by qPCR and Western blotting (WB). Furthermore, immunofluorescence assays were performed to visualize the subcellular localization of TCN1 and the nuclear translocation of phosphorylated p65 (p-p65) in HaCaT cells. Cell cycle progression was assessed using BrdU-PI flow cytometry. RESULTS TCN1 was upregulated in psoriatic lesions, and its expression levels were positively correlated with the PASI score. Following biologic treatment, TCN1 expression was reduced. TCN1 overexpression was associated with activation of the NF-κB signaling pathway, accompanied by increased synthesis of psoriasis-related inflammatory mediators, as well as an elevated proportion of cells in the S phase of the cell cycle. CONCLUSIONS TCN1 is essential in modulating inflammation and the cell cycle in psoriasis, implying its value as both a biomarker for diagnosis and a candidate for therapeutic intervention.
Collapse
Affiliation(s)
- Jian Huang
- Department of Dermatology, Guangdong College of Clinical Dermatology, Anhui Medical University, Hefei, Anhui Province, People's Republic of China; The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui Province, People's Republic of China; Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Huanhuan Yu
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xiuqing Yuan
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China; Shenzhen Children's Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Yuanqiu Zhong
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xinhui Li
- Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Yongfeng Chen
- Department of Dermatology, Guangdong College of Clinical Dermatology, Anhui Medical University, Hefei, Anhui Province, People's Republic of China; The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui Province, People's Republic of China; Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.
| |
Collapse
|
2
|
Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ballal S, Sharma R, Debnath S, Sinha A, Rekha A, Khan NH, Alrashoud MM, Kamal M, Imran M. Pathological interplay of NF-κB and M1 macrophages in chronic inflammatory lung diseases. Pathol Res Pract 2025; 269:155903. [PMID: 40081284 DOI: 10.1016/j.prp.2025.155903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/25/2024] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
Inflammatory lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), and pulmonary fibrosis depend on the pathology of the nuclear factor kappa B (NF-κB) signalling pathway and M1 macrophage polarization. This review discusses the intimate molecular interactions and processes that modulate NF-κB's promotion of M1 macrophages and chronic inflammation/tissue damage within the confines of this review. NF-κB activation in macrophages produces pro-inflammatory mediators (cytokines - TNFα, IL6, IL1β, and reactive oxygen species (ROS), further increasing airway remodeling and fibrosis. MAPK, JAK-STAT, and PI3K-Akt signalling systems cross-talked with the pathway, amplifying its effect on lung disease progression. Therapeutic strategies focused on inhibiting this axis, including inhibition of NF-κB and small molecule/modulation of macrophage polarization, represent potential ways to attenuate inflammation and promote tissue repair. The potential of precision medicine is illustrated by natural compounds such as curcumin and resveratrol and innovative RNA-based and nanoparticle delivery systems. Despite these challenges, specificity, minimizing systemic side effects, and optimized delivery methods remain difficult. To develop targeted therapies, more research must be conducted to refine targeted approaches, including immune profiling and single-cell analysis. This review aims to advance the management of hard-to-treat inflammatory lung diseases by addressing these complexities.
Collapse
Affiliation(s)
- Fadiyah Jadid Alanazi
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Al Jouf City 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rajesh Sharma
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Sourav Debnath
- Chandigarh pharmacy college, Chandigarh Group of colleges, Jhanjeri, Mohali 140307, Punjab, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - A Rekha
- Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | | | - Muhanad Mubarak Alrashoud
- Department of Inpatient Pharmacy, Dr. Sulaiman Alhabib Hospital, Alhamra Branch, Riyadh 13333, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohd Imran
- Center for Health Research, Northern Border University, Arar, Saudi Arabia; Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| |
Collapse
|
3
|
Guo Y, Yu H, Li J, Liu K, Han M, Tang Y, Su L, Li X, Wu H, Chen D. DNA-methylation eraser TET2 activates WTIP expression to suppress an AKT-dependent chemoresistance of gastric cancer. Neoplasia 2025; 65:101166. [PMID: 40279682 DOI: 10.1016/j.neo.2025.101166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/27/2025]
Abstract
Chemoresistance is one of the major causes of the failure in gastric cancer (GC) treatment and leads to poor clinical outcomes. Ten-eleven translocation (TET) 2 expression and activity are frequently reduced in solid tumors. However, whether TET2 participants in GC chemoresistance remains poorly understood. In this study, we demonstrated that the TET2 acts as a novel suppressor of GC chemoresistance. TET2 and transcription factor PATZ1 work together to promote the expression of WTIP. WTIP interacts with PP2A to inhibit the T308 phosphorylation and kinase activity of AKT, thereby suppressing stemness and chemoresistance of GC. Thus, the novel TET2-WTIP transcriptional axis, which is frequently silenced, suppresses an AKT-dependent chemoresistance of GC. TET2, combined with WTIP and AKT-pT308, can synergistically serve as a biomarker for predicting chemotherapy response in GC patients. Furthermore, we highlight that combining AKT inhibitor with chemotherapy is a promising therapeutic strategy for TET2-silenced GCs with chemoresistance in clinic.
Collapse
Affiliation(s)
- Yan Guo
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Hongyang Yu
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Jinyang Li
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Kewei Liu
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Mengyi Han
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Yuxin Tang
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Li Su
- Department of Oncology and Hematology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Xianfeng Li
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Haixia Wu
- School of Medical Technology, Chongqing Medical and Pharmaceutical College, Chongqing, China.
| | - Dongfeng Chen
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, China.
| |
Collapse
|
4
|
Onyiba CI, Kumar NK, Scarlett CJ, Weidenhofer J. Cell Progression and Survival Functions of Enzymes Secreted in Extracellular Vesicles Associated with Breast and Prostate Cancers. Cells 2025; 14:468. [PMID: 40214422 PMCID: PMC11988166 DOI: 10.3390/cells14070468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound cargoes secreted by normal and pathological cells. Through their protein, nucleic acid, and lipid cargoes, EVs mediate several cellular processes, such as cell-cell communication, cell development, immune response, and tissue repair. Most importantly, through their enzyme cargo, EVs mediate pathophysiological processes, including the pathogenesis of cancer. In this review, we enumerate several enzymes secreted in EVs (EV enzyme cargo) from cells and patient clinical samples of breast and prostate cancers and detail their contributions to the progression and survival of both cancers. Findings in this review reveal that the EV enzyme cargo could exert cell progression functions via adhesion, proliferation, migration, invasion, and metastasis. The EV enzyme cargo might also influence cell survival functions of chemoresistance, radioresistance, angiogenesis, cell death inhibition, cell colony formation, and immune evasion. While the current literature provides evidence of the possible contributions of the EV enzyme cargo to the progression and survival mechanisms of breast and prostate cancers, future studies are required to validate that these effects are modified by EVs and provide insights into the clinical applications of the EV enzyme cargo in breast and prostate cancer.
Collapse
Affiliation(s)
- Cosmos Ifeanyi Onyiba
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Niwasini Krishna Kumar
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Christopher J. Scarlett
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, University of Newcastle, Ourimbah, NSW 2258, Australia
| | - Judith Weidenhofer
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Ourimbah, NSW 2258, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
5
|
Sitthisuk P, Poorahong W, Innajak S, Krajarng A, Samosorn S, Watanapokasin R. Mammea siamensis Flower Extract-Induced Cell Death Apoptosis in HCT116 Colon Cancer Cells via Vacuolar-Type H +-ATPase Inhibition Associated with GSK-3β/β-Catenin, PI3K/Akt/NF-κB, and MAPK Signaling Pathway. Pharmaceuticals (Basel) 2025; 18:441. [PMID: 40283879 PMCID: PMC12030214 DOI: 10.3390/ph18040441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Background and Objective:Mammea siamensis (MS) is a Thai herb used in traditional medicine. Previous studies have reported the antiproliferative effects of its constituents in various cancer cell lines. However, the effects of MS extract on cytotoxicity and molecular mechanisms of apoptosis induction in HCT116 colon cancer cells have not been fully explored. Methods and Results: The cytotoxic effect of MS extract on HCT116 cells was assessed using the MTT assay. MS extract increased cytotoxicity in a concentration-dependent manner. It also induced nuclear morphological changes and disrupted the mitochondrial membrane potential (ΔΨm), as assessed by Hoechst 33342 and JC-1 staining, respectively. These findings indicated that MS extract induced apoptosis, which was further confirmed by flow cytometry showing an increase in the sub-G1 phase. To investigate the expression of signaling proteins, Western blot analysis was conducted. The results showed that MS extract activated caspase activity (caspase-8, -9, and -7) and inhibited PARP activity. Additionally, MS extract upregulated pro-apoptotic proteins (tBid, Bak, and cytochrome c) while downregulating anti-apoptotic proteins (Bcl-2 and Bcl-xL). Mechanistic studies revealed that MS extract activated MAPK pathways while inactivating the PI3K/Akt/NF-κB and GSK-3β/β-catenin pathways. Notably, MS extract also inhibited V-ATPases, as evaluated by acridine orange staining and Western blot analysis. Conclusions: Our findings suggest that MS extract induces apoptosis via the activation of both intrinsic and extrinsic pathways associated with the key signaling pathways. Therefore, MS extract shows potential as a therapeutic agent for colon cancer.
Collapse
Affiliation(s)
- Pornnapa Sitthisuk
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand; (P.S.); (S.I.)
| | - Watcharaporn Poorahong
- Department of Biochemistry, Faculty of Medicine, Bangkok Thonburi University, Bangkok 10170, Thailand;
| | - Sukanda Innajak
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand; (P.S.); (S.I.)
| | - Aungkana Krajarng
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand;
| | - Siritron Samosorn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Srinakharinwirot University, Bangkok 10110, Thailand;
| | - Ramida Watanapokasin
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand; (P.S.); (S.I.)
| |
Collapse
|
6
|
Li Y, Tian Y, Zhu L, Lin H, Zhao X, Liu C, Lv Y, Wang Z, Zuo Z, Wang J, Wang Z. Fuzi Lizhong Pill inhibited inflammatory response and promoted colon mucosal healing in dextran sulfate sodium-induced ulcerative colitis mice by down-regulating PI3K/AKT/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119483. [PMID: 39947366 DOI: 10.1016/j.jep.2025.119483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fuzi Lizhong Pill (FLP), a traditional Chinese herbal formula, has been historically used for treating gastrointestinal disorders characterized by cold deficiency patterns. Its application in ulcerative colitis (UC) stems from its warming and tonifying properties. AIM OF THE STUDY To evaluate the efficacy of FLP in the treatment of UC and investigate its mechanism of action. MATERIALS AND METHODS The chemical constituents of FLP were identified using UPLC-Q-Orbitrap HRMS. By establishing a preclinical UC mouse model with DSS and treating with FLP, we evaluated the effect of FLP on UC mice in terms of clinical symptoms, physiological indices, and histopathological examination. The anti-inflammatory and mucosal repair effects of FLP were examined at three levels: cellular, organoid, and animal, using immunohistochemistry, western blotting, RT-PCR, and other techniques. RESULTS We characterized the chemical composition of FLP and identified 99 compounds, including alkaloids, coumarins, and flavonoids. In UC mice, FLP alleviated clinical symptoms such as weight loss, blood in stools, and loose stools in UC mice; significantly reduced DAI scores in UC mice; significantly reversed splenomegaly and thymic atrophy caused by DSS; improved hemorrhage and inflammation-related hematological indices. In vitro and ex vivo studies showed that FLP inhibited the expression of TNF-α and IL-6, promoted the expression of the tight junction proteins ZO-1, Occludin, and Claudin 1, and promoted the proliferation of colonic epithelial cells in vivo. FLP also inhibited the transcription levels of PI3K, Akt, and NF-κB genes, as well as the expression or phosphorylation levels of related proteins in vitro and in vivo. CONCLUSION FLP may play a role in the treatment of UC by inhibiting the inflammatory response and repairing the colonic mucosal barrier by downregulating the PI3K/Akt/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yilin Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Yingying Tian
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, China
| | - Lei Zhu
- China National Accreditation Service for Conformity Assessment, Beijing, 100062, China
| | - Hongsai Lin
- China National Accreditation Service for Conformity Assessment, Beijing, 100062, China
| | - Xinyue Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Chuang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Yingnan Lv
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Zijian Wang
- Beijing Tongrentang Technology Co., LTD, Pharmaceutical Factory, Beijing, 100071, China
| | - Zeping Zuo
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, 264003, China.
| | - Jianfang Wang
- Department of Spleen, Stomach, Liver and Gallbladder, Dongfang Hospital, Beijing University of Chinese Medicine, 100078, China.
| | - Zhibin Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100105, China; Beijing Tongrentang Technology Co., LTD, Pharmaceutical Factory, Beijing, 100071, China.
| |
Collapse
|
7
|
Cordiano R, Caserta S, Minciullo PL, Allegra A, Gangemi S. Anthraquinones and Aloe Vera Extracts as Potential Modulators of Inflammaging Mechanisms: A Translational Approach from Autoimmune to Onco-Hematological Diseases. Molecules 2025; 30:1251. [PMID: 40142026 PMCID: PMC11944353 DOI: 10.3390/molecules30061251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Inflammaging is a chronic, low-grade inflammatory state that contributes to age-related diseases, including cardiovascular disorders, osteoporosis, neurodegeneration, and cancer. This process involves immunosenescence, oxidative stress, and immune aging, all of which contribute to the breakdown of immune tolerance and the onset of autoimmune disorders. Aloe vera (AV) has recently gained attention for its immunomodulatory, anti-inflammatory, and antioxidant properties. This review explores the effects of AV extracts and anthraquinones (e.g., aloe-emodin, emodin, aloin) on key inflammaging-driven mechanisms in autoimmunity. Our analysis highlights AV's ability to regulate hormone balance, autoantibody production, and cytokine/chemokine signaling (such as interleukin-1β, tumor necrosis factor-α, and interferon-γ). It modulates inflammatory pathways, including mitogen-activated protein kinases (MAPKs) and phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT), thereby inhibiting nuclear factor kappa-light-chain-enhancer of activated B-cell (NF-κB) activation. Additionally, AV enhances antioxidant defenses and restores immune balance by reducing Th1/Th17 subsets while promoting Th2-mediated regulation. Notably, AV also modulates inflammasome-mediated mechanisms and counteracts immunosenescence, which is driven by autophagy-related processes. These effects position AV as a potential integrative approach to mitigating inflammaging-driven autoimmunity. Furthermore, as inflammaging is increasingly recognized in onco-hematological diseases, AV-based strategies may offer novel therapeutic avenues. Future studies should focus on clinical validation, optimizing formulations, and expanding applications to broader age-related and immune-mediated disorders.
Collapse
Affiliation(s)
- Raffaele Cordiano
- Unit and School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.C.); (S.G.)
| | - Santino Caserta
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (S.C.); (A.A.)
| | - Paola Lucia Minciullo
- Unit and School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.C.); (S.G.)
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (S.C.); (A.A.)
| | - Sebastiano Gangemi
- Unit and School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.C.); (S.G.)
| |
Collapse
|
8
|
Son SK, Moon JS, Yang DW, Jung NR, Kang JH, Lee BN, Kim SH, Kim MS. Role of FOXO3a in LPS-induced inflammatory conditions in human dental pulp cells. J Oral Biosci 2025; 67:100614. [PMID: 39824385 DOI: 10.1016/j.job.2025.100614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
OBJECTIVES We investigated the involvement of FOXO3a in lipopolysaccharide (LPS)-induced inflammation in primary human dental pulp cells (HDPCs). METHODS HDPCs that were isolated from donors undergoing tooth extraction for orthodontic purposes were cultured with or without 1 μg/mL LPS at various intervals. The FOXO3a localization in the HDPCs was verified using immunofluorescence. Proinflammatory cytokines, such as interleukin (IL) 1β, IL6, and IL8, as well as their underlying mechanisms were assessed by observing gene and protein expressions through quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses. RESULTS LPS treatment enhanced the expressions of IL1β, IL6, and IL8 in HDPCs, concurrently activating nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB). Furthermore, FOXO3a expression was higher in the LPS-stimulated HDPCs, as confirmed by immunofluorescence localization. The results of loss-/gain-of-function approaches confirmed the regulatory role of FOXO3a in inflammatory HDPCs. FOXO3a knockdown attenuated proinflammatory cytokine expression; FOXO3a overexpression augmented their expression levels. FOXO3a inhibited retinoic acid receptor-related orphan receptor alpha (RORα) expression, thereby inactivating NFκB. CONCLUSION Our findings suggest that FOXO3a contributes to homeostasis in HDPCs through modulating the expression of proinflammatory cytokines under inflammatory conditions.
Collapse
Affiliation(s)
- Su-Kyung Son
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Jung-Sun Moon
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Dong-Wook Yang
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Na-Ri Jung
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Jee-Hae Kang
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Bin-Na Lee
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Sun-Hun Kim
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Min-Seok Kim
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea.
| |
Collapse
|
9
|
Ruggiero M, Motti ML, Meccariello R, Mazzeo F. Resveratrol and Physical Activity: A Successful Combination for the Maintenance of Health and Wellbeing? Nutrients 2025; 17:837. [PMID: 40077707 PMCID: PMC11902109 DOI: 10.3390/nu17050837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Physical exercise is an essential component of human health. In recent years, scientific research has focused on identifying natural compounds and formulating new supplements aimed at enhancing athletic performance, accelerating muscle recovery, and minimizing the damage caused by physical exertion. The use of antioxidants to counteract the formation of reactive oxygen species (ROS) following physical activity (PA) is already a widely adopted practice. Resveratrol (RES), a polyphenol belonging to the stilbene class, is well known for its potent antioxidant activity and anti-inflammatory effects primarily attributed to the activation of sirtuins. RES possesses multiple nutraceutical properties used for the prevention and treatment of inflammatory, cardiovascular, neoplastic, and infectious diseases, thus attracting attention to study its use in combination with physical exercise to promote well-being. Animal trials combining RES and PA have mainly reported improvements in muscle, energy, and cardiovascular functions. The data presented and discussed in this narrative review are from Pubmed, Scopus, and the Human Gene Database (search limited to 2011 to 2025 with the keywords RES, sirtuins, and physical activity altogether or in combination with each other). This review gathers several studies on RES focusing on its nutraceutical properties, epigenetic activities via sirtuins, and the potential benefits of combining RES with PA in maintaining health and well-being based on trials performed first in animals and later in humans. Human studies have been conducted on various populations, including active adults, sedentary individuals, patients with diseases, and elderly individuals. Some studies have confirmed the benefits of RES observed in animal experiments. However, in some cases, no substantial differences were found between RES supplementation and the control group. In conclusion, the benefits of RES on PA reported in the literature are still not fully evident, given the contrasting studies and the still limited number of trials, but both RES and PA are successful tools for the maintenance of health and wellbeing.
Collapse
Affiliation(s)
- Mario Ruggiero
- Department of Medical, Human Movement and Well-Being Sciences, University of Naples Parthenope, 80133 Naples, Italy; (M.R.); (M.L.M.); (R.M.)
| | - Maria Letizia Motti
- Department of Medical, Human Movement and Well-Being Sciences, University of Naples Parthenope, 80133 Naples, Italy; (M.R.); (M.L.M.); (R.M.)
| | - Rosaria Meccariello
- Department of Medical, Human Movement and Well-Being Sciences, University of Naples Parthenope, 80133 Naples, Italy; (M.R.); (M.L.M.); (R.M.)
| | - Filomena Mazzeo
- Department of Economics, Law, Cybersecurity and Sports Sciences, University of Naples Parthenope, 80035 Nola, Italy
| |
Collapse
|
10
|
Pouyan A, Ghorbanlo M, Eslami M, Jahanshahi M, Ziaei E, Salami A, Mokhtari K, Shahpasand K, Farahani N, Meybodi TE, Entezari M, Taheriazam A, Hushmandi K, Hashemi M. Glioblastoma multiforme: insights into pathogenesis, key signaling pathways, and therapeutic strategies. Mol Cancer 2025; 24:58. [PMID: 40011944 PMCID: PMC11863469 DOI: 10.1186/s12943-025-02267-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive primary brain tumor in adults, characterized by a poor prognosis and significant resistance to existing treatments. Despite progress in therapeutic strategies, the median overall survival remains approximately 15 months. A hallmark of GBM is its intricate molecular profile, driven by disruptions in multiple signaling pathways, including PI3K/AKT/mTOR, Wnt, NF-κB, and TGF-β, critical to tumor growth, invasion, and treatment resistance. This review examines the epidemiology, molecular mechanisms, and therapeutic prospects of targeting these pathways in GBM, highlighting recent insights into pathway interactions and discovering new therapeutic targets to improve patient outcomes.
Collapse
Affiliation(s)
- Ashkan Pouyan
- Department of Neurosurgery, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Masoud Ghorbanlo
- Department of Anesthesiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Eslami
- Department of Neurosurgery, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Jahanshahi
- Department of Neurosurgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Ziaei
- Department of Neurosurgery, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Salami
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khatere Mokhtari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Koorosh Shahpasand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Laboratory Medicine and Pathology, Institute for Translational Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Tohid Emami Meybodi
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Functional Neurosurgery Research Center, Shohada Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Epidemiology, University of Tehran, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
11
|
Fukuda R, Beppu S, Hinata D, Kamada Y, Okiyoneda T. Perturbation of EPHA2 and EFNA1 trans binding amplifies inflammatory response in airway epithelial cells. iScience 2025; 28:111872. [PMID: 39991543 PMCID: PMC11847143 DOI: 10.1016/j.isci.2025.111872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 06/07/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
The interactions between EPH receptors and ephrin (EFN) ligands play a crucial role in maintaining epithelial integrity and aiding in defense against infections. However, it remains unclear how the EPH-EFN trans-binding changes during infections and how this alteration affects inflammatory response. Here we report that pathogen-associated molecular patterns (PAMPs) disrupt the EPHA2-EFNA1 trans-binding in airway epithelial cells (AECs). Mechanistically, flagellin induces the TLR5-dependent EFNA1 cleavage through the metalloproteinase ADAM9 concomitant with the activation of ligand-independent EPHA2 signaling. We found that the ablation of EPHA2 reduced the responsiveness of respiratory inflammation induced by flagellin and Pseudomonas aeruginosa both in vitro and in vivo. Notably, even in the absence of PAMPs, the inflammatory response in AECs was stimulated by forcibly induced EFNA1 shedding. These findings illustrate that the perturbation of the EPHA2-EFNA1 trans-binding acts as a sensing mechanism for infections and amplifies the inflammatory response, providing a defense mechanism for respiratory epithelia.
Collapse
Affiliation(s)
- Ryosuke Fukuda
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1330, Japan
| | - Shiori Beppu
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1330, Japan
| | - Daichi Hinata
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1330, Japan
| | - Yuka Kamada
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1330, Japan
| | - Tsukasa Okiyoneda
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1330, Japan
| |
Collapse
|
12
|
Ranxhi B, Bangash ZR, Chbihi ZM, Todi SV, LeWitt PA, Tsou WL. The effect of AKT inhibition in α-synuclein-dependent neurodegeneration. Front Mol Neurosci 2025; 18:1524044. [PMID: 39974188 PMCID: PMC11835820 DOI: 10.3389/fnmol.2025.1524044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/08/2025] [Indexed: 02/21/2025] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder affecting millions of individuals worldwide. A hallmark of PD pathology is the accumulation of α-synuclein (α-Syn), a small protein known to support neuronal development and function. However, in PD, α-Syn cumulatively misfolds into toxic aggregates that disrupt cellular processes and contribute to neuronal damage and neurodegeneration. Previous studies implicated the AKT signaling pathway in α-Syn toxicity in cellular models of PD, suggesting AKT as a potential therapeutic target. Here, we investigated the effect of AKT inhibition in a Drosophila model of synucleinopathy. We observed that administration of the AKT inhibitor, A-443654 led to mild improvements in both survival and motor function in flies expressing human α-Syn. Genetic studies revealed that reduction of AKT levels decreased α-Syn protein levels, concomitant with improved physiological outcomes. The protective effects of AKT reduction appear to operate through the fly ortholog of NF-κB, Relish, suggesting a link between AKT and NF-κB in regulating α-Syn levels. These findings highlight the AKT cascade as a potential therapeutic target for synucleinopathies and provide insights into mechanisms that could be utilized to reduce α-Syn toxicity in PD and related disorders, such as multiple system atrophy.
Collapse
Affiliation(s)
- Bedri Ranxhi
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Zoya R. Bangash
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Zachary M. Chbihi
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Sokol V. Todi
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Peter A. LeWitt
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Neurology, Henry Ford Health Systems, Detroit, MI, United States
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
13
|
Jiang J, Wang Q, Wu Q, Deng B, Guo C, Chen J, Zeng J, Guo Y, Ma X. Angel or devil: the dual roles of 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucopyranoside in the development of liver injury based on integrating pharmacological techniques: a systematic review. Front Pharmacol 2025; 16:1523713. [PMID: 39963244 PMCID: PMC11830817 DOI: 10.3389/fphar.2025.1523713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025] Open
Abstract
Background and purpose 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucoside (TSG) exhibits a dualistic pharmacological profile, acting as both a hepatoprotective and hepatotoxic agent, which is intricately linked to its interaction with multiple signaling pathways and its stereoisomeric forms, namely, cis-SG and trans-SG. The purpose of this study is to evaluate both the hepatoprotective and hepatotoxic effects of TSG and give therapeutic guidance. Methods This study performed a systematic search of eight databases to identify preclinical literature up until March 2024. The CAMARADES system evaluated evidence quality and bias. STATA and Python were used for statistical analysis, including dose-effect maps, 3D maps and radar charts to show the dose-time-effect relationship of TSG on hepatoprotection and hepatotoxicity. Results After a rigorous screening process, a total of 24 studies encompassing 564 rodents were selected for inclusion in this study. The findings revealed that TSG exhibited bidirectional effects on the levels of ALT and AST, while also regulating the levels of ALT, AST, TNF-α, IL-6, serum TG, serum TC, SOD, MDA, IFN-γ, and apoptosis rate. The histological analysis of liver tissue confirmed the regulatory effects of TSG, and a comprehensive analysis revealed the optimal protective dosage range was 27.27-38.81 mg/kg/d and the optimal toxic dosage range was 51.93-76.07 mg/kg/d. TSG exerts the dual effects on liver injury (LI) through the network of Keap1/Nrf2/HO-1/NQO1, NF-κB, PPAR, PI3K/Akt, JAK/STAT and TGF-β pathways. Conclusion TSG could mediate the pathways of oxidation, inflammation, and metabolism to result in hepatoprotection (27.27-38.81 mg/kg/d) and hepatotoxicity (51.93-76.07 mg/kg/d).
Collapse
Affiliation(s)
- Jiajie Jiang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qixiu Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Qiang Wu
- Chengdu Shuangliu Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Bobin Deng
- School of Pharmacy, Xian Medical University, Xi’an, China
| | - Cui Guo
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Chen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yaoguang Guo
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
14
|
Chan H, Lin W, Kuo D, Chuang H. Beta-Caryophyllene Augments Radiotherapy Efficacy in GBM by Modulating Cell Apoptosis and DNA Damage Repair via PPARγ and NF-κB Pathways. Phytother Res 2025; 39:776-788. [PMID: 39668701 PMCID: PMC11832361 DOI: 10.1002/ptr.8413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/30/2024] [Accepted: 11/26/2024] [Indexed: 12/14/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain malignancy with limited treatment options. Radiotherapy (RT) is often used for treating unresectable GBM; however, the outcomes are often limited due to the radioresistance of GBM. Therefore, the discovery of potential radiosensitizers to enhance GBM responses to RT is crucial. Beta-caryophyllene (BCP), a natural cannabinoid, promotes cancer apoptosis by upregulating the PPARγ signaling pathway and can cross the blood-brain barrier due to its lipophilic nature. This study aimed to evaluate the radiosensitizing potential of BCP in GBM cells. U87MG and GL261 cells and a GL261 tumor-bearing model were treated with RT, BCP, or both. Treatment efficacy was assessed using the MTT assay and tumor growth tracking, and the underlying mechanisms were investigated using western blotting, immunofluorescence staining, and other analyses. BCP synergistically enhanced the efficacy of RT in cell culture, as evidenced by the combination index determined through the MTT assay. This enhancement was mediated by the BCP-induced deceleration of DNA damage repair, as demonstrated by sustained γH2AX signal, upregulated PPARγ levels, and reduced expression of pAKT, pERK, and NF-κB, indicating apoptosis induction and inhibition of survival pathways. BCP significantly inhibited tumor growth in GL261 tumor-bearing mice with no discernible side effects. These findings indicate that BCP may serve as a potential radiosensitizer for improving RT outcomes in GBM by inhibiting DNA repair, inducing apoptosis, and suppressing anti-apoptotic and survival pathways.
Collapse
Affiliation(s)
- Hui‐Wen Chan
- Department of Biomedical Imaging and Radiological SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Wei‐Chan Lin
- Department of RadiologyCathay General HospitalTaipei CityTaiwan
- School of MedicineFu‐Jen Catholic UniversityNew Taipei CityTaiwan
| | - Deng‐Yu Kuo
- Division of Radiation Oncology, Department of RadiologyFar Eastern Memorial HospitalNew Taipei CityTaiwan
| | - Hui‐Yen Chuang
- Department of Biomedical Imaging and Radiological SciencesNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
15
|
Wu XY, Dong QW, Zhang YB, Li JX, Zhang MQ, Zhang DQ, Cui YL. Cimicifuga heracleifolia kom. Attenuates ulcerative colitis through the PI3K/AKT/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118892. [PMID: 39395768 DOI: 10.1016/j.jep.2024.118892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/13/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cimicifuga heracleifolia Kom. (C. heracleifolia) has demonstrated efficacy in treating gastrointestinal disorders, including splenasthenic diarrhea. Ulcerative colitis (UC), a chronic inflammatory bowel disease, shares similarities with splenasthenic diarrhea. However, the pharmacological effects of C. heracleifolia on UC and the underlying mechanisms remain unexplored. AIM OF THE STUDY The present study investigates the therapeutic potential and mechanisms of C. heracleifolia in UC. METHODS Initially, network pharmacology analysis, encompassing ingredient screening, target prediction, protein-protein interaction (PPI) network analysis, and enrichment analysis, was employed to predict the mechanisms of C. heracleifolia. The findings were further validated using transcriptomics and functional assays in a dextran sulfate sodium (DSS)-induced UC model. Additionally, bioactive compounds were identified through surface plasmon resonance (SPR) analysis, molecular docking, and cell-based assays. RESULTS A total of 52 ingredients of C. heracleifolia were screened, and 32 key targets were identified within a PPI network comprising 285 potential therapeutic targets. Enrichment analysis indicated that the anti-UC effects of C. heracleifolia are mediated through immune response modulation and the inhibition of inflammatory signaling pathways. In vivo experiments showed that C. heracleifolia mitigated histological damage in the colon, reduced the expression of phosphorylated Akt1, nuclear factor-kappa B (NF-κB) p65, and inhibitor of Kappa B kinase α/β (IKKα/β), suppressed the content of interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), and enhanced the expression of tight junction proteins. Moreover, cimigenoside, caffeic acid, and methyl caffeate were identified as the bioactive constituents responsible for the UC treatment effects of C. heracleifolia. CONCLUSIONS In summary, this study is the first to demonstrate that C. heracleifolia exerts therapeutic effects on UC by enhancing the intestinal mucosal barrier and inhibiting the phosphatidylinositol 3-kinase (PI3K)/AKT/NF-κB signaling pathway. These findings offer valuable insights into the clinical application of C. heracleifolia for UC management.
Collapse
Affiliation(s)
- Xue-Yi Wu
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Qin-Wei Dong
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Yong-Bo Zhang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Jia-Xin Li
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Mei-Qing Zhang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - De-Qin Zhang
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China
| | - Yuan-Lu Cui
- State Key Laboratory of Component-based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, PR China.
| |
Collapse
|
16
|
Lu W, Feng W, Zhen H, Jiang S, Li Y, Liu S, Ru Q, Xiao W. Unlocking the therapeutic potential of WISP-1: A comprehensive exploration of its role in age-related musculoskeletal disorders. Int Immunopharmacol 2025; 145:113791. [PMID: 39667044 DOI: 10.1016/j.intimp.2024.113791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/03/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024]
Abstract
As the global population ages, the incidence of age-related musculoskeletal diseases continues to increase, driven by numerous complex and poorly understood factors. WNT-1 inducible secreted protein 1 (WISP-1), a secreted matrix protein, plays a critical role in the growth and development of the musculoskeletal system, including chondrogenesis, osteogenesis, and myogenesis. Numerous in vivo and in vitro studies have demonstrated that WISP-1 is significantly upregulated in age-related musculoskeletal conditions, such as osteoarthritis, osteoporosis, and sarcopenia, suggesting its involvement in the pathogenesis of these diseases. Regulating WISP-1 expression holds promise as a therapeutic strategy for improving musculoskeletal function, potentially offering new avenues for treating age-related musculoskeletal diseases in clinical practice. This review highlights the signaling pathways associated with WISP-1, its physiological roles within the musculoskeletal system, and its therapeutic potential in treating age-related musculoskeletal disorders.
Collapse
Affiliation(s)
- Wenhao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wenjie Feng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Haozu Zhen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Shide Jiang
- The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shuguang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710001, Shaanxi, China.
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
17
|
Florencio-Silva R, Sasso GRDS, Sasso-Cerri E, Cerri PS, Gil CD, de Jesus Simões M. Relationship between autophagy and NLRP3 inflammasome during articular cartilage degradation in oestrogen-deficient rats with streptozotocin-induced diabetes. Ann Anat 2025; 257:152318. [PMID: 39216675 DOI: 10.1016/j.aanat.2024.152318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Estrogen deficiency and Diabetes mellitus (DM) cause joint tissue deterioration, although the mechanisms are uncertain. This study evaluated the immunoexpression of autophagy and NLRP3-inflammasome markers, in rat articular cartilage with estrogen deficiency and DM. METHODS Twenty rats were sham-operated (SHAM) or ovariectomized (OVX) and equally allocated into four groups: SHAM and OVX groups administered with vehicle solution; SHAM and OVX groups treated with 60 mg/kg/body weight of streptozotocin, intraperitoneally, to induce DM (SHAM-DM and OVX-DM groups). After seven weeks, the rats were euthanized, and their joint knees were processed for paraffin embedding. Sections were stained with haematoxylin-eosin, toluidine blue, safranin-O/fast-green or subjected to picrosirius-red-polarisation method; immunohistochemistry to detect beclin-1 and microtubule-associated protein 1B-light chain 3 (autophagy markers), NLRP3 and interleukin-1β (IL-1β) (inflammasome activation markers), along with matrix metalloproteinase-9 (MMP-9), Nuclear factor-kappa B (NFκB), and Vascular endothelial growth factor A (VEGF-A) were performed. RESULTS Deterioration of articular cartilage and subchondral bone were greater in SHAM-DM and OVX-DM groups. Higher percentages of immunolabeled chondrocytes to NLRP3, IL-1β, MMP-9, NFκB, and VEGF-A, as well as lower percentages of chondrocytes immunolabeled to autophagy markers, were noticed in estrogen-deficient and diabetic groups. These differences were greater in the OVX-DM group. Percentages of immunolabeled chondrocytes showed negative correlation between autophagy markers v.s IL-1β, NLRP-3, MMP-9, NFκB, and VEGF-A, along with positive correlation between VEGF-A vs. MMP-9, NFκB, IL-1β, and NLRP3, and MMP-9 vs. NFκB. CONCLUSIONS In conclusion, autophagy reduction and NLRP3 inflammasome activation in chondrocytes may be implicated in articular cartilage degradation, under estrogen-deficient and DM conditions. Moreover, the combination of estrogen deficiency and DM may potentiate those effects.
Collapse
Affiliation(s)
- Rinaldo Florencio-Silva
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Ginecologia, São Paulo, SP, Brazil; Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil.
| | - Gisela Rodrigues da Silva Sasso
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| | - Estela Sasso-Cerri
- São Paulo State University (UNESP), School of Dentistry, Araraquara - Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry - Laboratory of Histology and Embryology, Araraquara, SP, Brazil
| | - Paulo Sérgio Cerri
- São Paulo State University (UNESP), School of Dentistry, Araraquara - Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry - Laboratory of Histology and Embryology, Araraquara, SP, Brazil
| | - Cristiane Damas Gil
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| | - Manuel de Jesus Simões
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Ginecologia, São Paulo, SP, Brazil; Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| |
Collapse
|
18
|
Hansman DS, Du J, Casson RJ, Peet DJ. Eye on the horizon: The metabolic landscape of the RPE in aging and disease. Prog Retin Eye Res 2025; 104:101306. [PMID: 39433211 PMCID: PMC11833275 DOI: 10.1016/j.preteyeres.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
To meet the prodigious bioenergetic demands of the photoreceptors, glucose and other nutrients must traverse the retinal pigment epithelium (RPE), a polarised monolayer of cells that lie at the interface between the outer retina and the choroid, the principal vascular layer of the eye. Recent investigations have revealed a metabolic ecosystem in the outer retina where the photoreceptors and RPE engage in a complex exchange of sugars, amino acids, and other metabolites. Perturbation of this delicate metabolic balance has been identified in the aging retina, as well as in age-related macular degeneration (AMD), the leading cause of blindness in the Western world. Also common in the aging and diseased retina are elevated levels of cytokines, oxidative stress, advanced glycation end-products, increased growth factor signalling, and biomechanical stress - all of which have been associated with metabolic dysregulation in non-retinal cell types and tissues. Herein, we outline the role of these factors in retinal homeostasis, aging, and disease. We discuss their effects on glucose, mitochondrial, lipid, and amino acid metabolism in tissues and cell types outside the retina, highlighting the signalling pathways through which they induce these changes. Lastly, we discuss promising avenues for future research investigating the roles of these pathological conditions on retinal metabolism, potentially offering novel therapeutic approaches to combat age-related retinal disease.
Collapse
Affiliation(s)
- David S Hansman
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia.
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Robert J Casson
- Discipline of Ophthalmology and Visual Science, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
19
|
Laganà A, Scalzulli E, Bisegna ML, Ielo C, Martelli M, Breccia M. Understanding and overcoming resistance to tyrosine kinase inhibitors (TKIs) in Chronic myeloid leukemia (CML). Expert Rev Hematol 2025; 18:65-79. [PMID: 39647915 DOI: 10.1080/17474086.2024.2440776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/01/2024] [Accepted: 12/07/2024] [Indexed: 12/10/2024]
Abstract
INTRODUCTION Chronic myeloid leukemia (CML) represents one of the first neoplasms whose molecular pathogenesis was successfully unraveled, with tyrosine kinase inhibitors (TKIs) representing one of the first-targeted therapies. TKIs have revolutionized long-term outcomes of CML patients and their life expectancy. Nonetheless, a minority of patients will develop TKI resistance due to a complex and multifactorial process that ultimately leads to the emergence of an unresponsive cancer clone. Overcoming TKI resistance is considered one of the major challenges in CML management. AREAS COVERED In this review, the main findings extrapolated from published research, guidelines, and clinical trials regarding TKI resistance (published before October 2024) are discussed. Data have been obtained through broad research on Medline, Embase, Pubmed, and archives from EHA and ASH congresses. EXPERT OPINION Nowadays, asciminib and ponatinib have expanded the therapeutic arsenal for resistant-CML management and allogenic transplant still represents an important alternative in the context of multiple TKI failures. Off-label use of TKIs combination therapies, although theoretically appealing, lacks robust clinical evidence and regulatory approval. Looking ahead, the introduction of novel technologies such as digital PCR (dPCR) and next generation sequencing (NGS) holds great potential to revolutionize the management of TKI-resistant CML cases.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Drug Resistance, Neoplasm/drug effects
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Tyrosine Kinase Inhibitors
- Imidazoles
- Pyridazines
Collapse
Affiliation(s)
- Alessandro Laganà
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Emilia Scalzulli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Laura Bisegna
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Claudia Ielo
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Maurizio Martelli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Massimo Breccia
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
20
|
Yan A, Li Z, Gao Y, Hu F, Han S, Liu F, Liu Z, Chen J, Yuan C, Zhou C. Isobicyclogermacrenal ameliorates hippocampal ferroptosis involvement in neurochemical disruptions and neuroinflammation induced by sleep deprivation in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156306. [PMID: 39647468 DOI: 10.1016/j.phymed.2024.156306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/12/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Sleep deprivation (SLD) is a widespread condition that disrupts physiological functions and may increase mortality risk. Valeriana officinalis, a traditional medicinal herb known for its sedative and hypnotic properties, contains isobicyclogermacrenal (IG), a newly isolated active compound. However, research on the therapeutic potential of IG for treating SLD remains limited. METHODS In this study, IG was extracted and characterized from Valeriana officinalis, and an SLD model was established in rats using p-chlorophenylalanine (PCPA). Behavioral tests and pathological studies were conducted to assess the effects of IG on SLD, and transcriptomic and metabolomic analyses were utilized to investigate its underlying mechanisms. RESULTS IG administration significantly improved the cognitive performance of SLD rats in behavioral tests and ameliorated histological injuries in the hippocampus and cerebral cortex. IG treatment increased the levels of brain-derived neurotrophic factor (BDNF) and neurotransmitters such as serotonin (5-HT) in SLD rats. Additionally, IG directly targets TFRC, thereby improving iron metabolism in the hippocampus. Comprehensive transcriptomic and metabolomic analyses revealed that the improvements from IG stemmed from the mitigation of abnormalities in iron metabolism, cholesterol metabolism, and glutathione metabolism, leading to reduced oxidative stress, ferroptosis, and neuroinflammation in the hippocampus caused by SLD. CONCLUSIONS Collectively, these findings suggest that IG has the potential to ameliorate neurological damage and cognitive impairment caused by SLD, offering a novel strategy for protection against the adverse effects of SLD.
Collapse
Affiliation(s)
- Ao Yan
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Zhejin Li
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Yuanwei Gao
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Fanglong Hu
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Shuo Han
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Fengjie Liu
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Zhongcheng Liu
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China
| | - Jinting Chen
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang 050017, China
| | - Chunmao Yuan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China.
| | - Chengyan Zhou
- College of Pharmaceutical Science, Hebei University, Baoding 071002, China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, China.
| |
Collapse
|
21
|
Stutz C, Gegout PY, Bloch C, Özçelik H, Anton N, Tabti R, Désaubry L, Huck O, Petit C. The prohibitin ligand IN44 decreases Porphyromonas gingivalis mediated inflammation. BMC Oral Health 2024; 24:1534. [PMID: 39709363 DOI: 10.1186/s12903-024-05209-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/14/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Periodontitis is an inflammatory disease causing destruction of periodontal tissues. Controlling inflammation is crucial for periodontitis treatment. Prohibitins (PHBs) are emerging targets in the treatment of inflammatory diseases. To identify compounds that would alleviate periodontitis, several small molecules that directly target PHBs and display various pharmacological activities were screened to decrease Porphyromonas gingivalis induced inflammation. Indeed, IN44, a new PHB ligand that has been shown to inhibit STAT3 and NF-kB signaling, suggesting that it may alleviate periodontitis. This study aimed to assess IN44's impact on inflammation elicited by P. gingivalis. METHODS In vitro, IN44 cytotoxicity was tested on periodontal cells with AlamarBlue and Live/Dead assays. Its effect on cytokines and mitochondrial ROS production were evaluated using ELISA and Mitosox assay. In mouse, systemic inflammation and experimental periodontitis were induced to assess IN44's therapeutic effects. RESULTS In vitro, IN44 (50 µM) showed no cytotoxicity on periodontal cells. It significantly reduced pro-inflammatory cytokine secretion and mitochondrial ROS in P. gingivalis-infected epithelial cells. Proteome analysis on infected epithelial cells revealed modulation of HSP60 and Akt expression by IN44. In vivo, IN44 demonstrated anti-inflammatory effects in a mouse model of systemic inflammation induced by P. gingivalis, and it improved periodontal healing. CONCLUSION These findings suggest that PHBs may warrant consideration as therapeutic targets for periodontitis and possibly other inflammatory disorders.
Collapse
Affiliation(s)
- Céline Stutz
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, Strasbourg, 67084, France
| | - Pierre-Yves Gegout
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, Strasbourg, 67084, France
- Faculté de Chirurgie-dentaire, Dental Faculty, Université de Strasbourg, 8 rue Sainte-Elisabeth, Strasbourg, 67000, France
- Pôle de médecine et chirurgie Bucco-dentaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, 67000, France
| | - Chloé Bloch
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, Strasbourg, 67084, France
| | - Hayriye Özçelik
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, Strasbourg, 67084, France
| | - Nicolas Anton
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, Strasbourg, 67084, France
| | - Redouane Tabti
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, Strasbourg, 67084, France
| | - Laurent Désaubry
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, Strasbourg, 67084, France
| | - Olivier Huck
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, Strasbourg, 67084, France.
- Faculté de Chirurgie-dentaire, Dental Faculty, Université de Strasbourg, 8 rue Sainte-Elisabeth, Strasbourg, 67000, France.
- Pôle de médecine et chirurgie Bucco-dentaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, 67000, France.
| | - Catherine Petit
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, Strasbourg, 67084, France
- Faculté de Chirurgie-dentaire, Dental Faculty, Université de Strasbourg, 8 rue Sainte-Elisabeth, Strasbourg, 67000, France
- Pôle de médecine et chirurgie Bucco-dentaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, 67000, France
| |
Collapse
|
22
|
Lee S, Lee H, Jang YJ, Lee K, Kim HJ, Lee JY, Kim JM, Park S, Song JS, Lee JH, Hyun TK, Park JI, Yi SJ, Kim K. Denatonium inhibits RANKL-induced osteoclast differentiation and rescues the osteoporotic phenotype by blocking p65 signaling pathway. Mol Med 2024; 30:248. [PMID: 39701944 DOI: 10.1186/s10020-024-01031-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Bone remodeling is a critical process that maintains skeletal integrity, orchestrated by the balanced activities of osteoclasts, which resorb bone, and osteoblasts, which form bone. Osteoclastogenesis, the formation of osteoclasts, is primarily driven by NFATc1, a process activated through c-Fos and NF-κB signaling pathways in response to receptor activator of nuclear factor κB ligand (RANKL). Dysregulation of RANKL signaling is a key contributor to pathological bone loss, as seen in conditions such as osteoporosis. METHODS We investigated the effects of denatonium, a known bitter compound, on RANKL-induced osteoclast differentiation. We used RNA sequencing (RNA-seq) to analyze gene expression profiles in osteoclast precursors treated with denatonium. Transcription factor prediction analysis was conducted to identify key targets of denatonium action. Additionally, we performed Western blotting to examine the phosphorylation status of AKT and p65, crucial components of the NF-κB pathway. Chromatin immunoprecipitation (ChIP) assays were employed to assess the binding of p65 to promoter regions of osteoclast-related genes. Finally, we tested the therapeutic potential of denatonium in a mouse model of osteoporosis. RESULTS Our findings demonstrated that denatonium significantly inhibited RANKL-induced osteoclastogenesis by targeting the p65 pathway. RNA-seq analysis revealed a downregulation of osteoclast-related genes following denatonium treatment, corroborated by transcription factor prediction analysis, which highlighted p65 as a key target. Denatonium effectively blocked the phosphorylation of AKT and p65, key steps in NF-κB activation. ChIP assays further confirmed that denatonium reduced the enrichment of p65 at promoter regions critical for osteoclast differentiation. In vivo, denatonium treatment in an osteoporosis animal model led to a significant restoration of bone health, demonstrating its potential as a therapeutic agent. CONCLUSIONS This study identifies denatonium as an inhibitor of RANKL-induced osteoclast differentiation, potentially acting through suppression of the p65 signaling pathway. The ability of denatonium to downregulate osteoclast-related genes and inhibit key signaling events highlights its potential as a candidate for further investigation in the context of bone loss and osteoporosis.
Collapse
Affiliation(s)
- Sheunghun Lee
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hyerim Lee
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - You-Jee Jang
- Department of Biomedical Laboratory Science, Honam University, Gwangju, Republic of Korea
| | - Kyubin Lee
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hye-Jung Kim
- New Drug Development Center, KBIO Osong Medical Innovation Foundation, Chungbuk, Republic of Korea
| | - Jung Yeol Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Jin-Man Kim
- Asan Medical Center, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Sunyou Park
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Jin Sook Song
- Data Convergence Drug Research Center, Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Ji Hoon Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Tae Kyung Hyun
- Department of Industrial Plant Science & Technology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jae-Il Park
- Korea Basic Science Institute, Gwangju Center at Chonnam National University, Gwangju, Republic of Korea.
| | - Sun-Ju Yi
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| | - Kyunghwan Kim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| |
Collapse
|
23
|
Soltani R, Tabibkhooei A, Hadizadeh M, Parvizpour S, Esferizi RM, Ghasemi S. Introduction of MYBL2 as a common regulator between AHR and RELA: Its relationship with lnc-UCC and lnc-HOTTIP in glioblastoma multiforme. GENE REPORTS 2024; 37:102046. [DOI: 10.1016/j.genrep.2024.102046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
24
|
Han NR, Park HJ, Ko SG, Moon PD. Tryptanthrin Down-Regulates Oncostatin M by Targeting GM-CSF-Mediated PI3K-AKT-NF-κB Axis. Nutrients 2024; 16:4109. [PMID: 39683503 DOI: 10.3390/nu16234109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Oncostatin M (OSM) is involved in several inflammatory responses. Tryptanthrin (TRYP), as a natural alkaloid, is a bioactive compound derived from indigo plants. Objectives/ Methods: The purpose of this study is to investigate the potential inhibitory activity of TRYP on OSM release from neutrophils using neutrophils-like differentiated (d)HL-60 cells and neutrophils from mouse bone marrow. RESULTS The results showed that TRYP reduced the production and mRNA expression levels of OSM in the granulocyte-macrophage colony-stimulating factor (GM-CSF)-stimulated neutrophils-like dHL-60 cells. In addition, TRYP decreased the OSM production levels in the GM-CSF-stimulated neutrophils from mouse bone marrow. TRYP inhibited the phosphorylation of phosphatidylinositol 3-kinase (PI3K), AKT, and nuclear factor (NF)-κB in the GM-CSF-stimulated neutrophils-like dHL-60 cells. CONCLUSIONS Therefore, these results reveal for the first time that TRYP inhibits OSM release via the down-regulation of PI3K-AKT-NF-κB axis from neutrophils, presenting its potential as a therapeutic agent for inflammatory responses.
Collapse
Affiliation(s)
- Na-Ra Han
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hi-Joon Park
- Department of Anatomy & Information Sciences, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seong-Gyu Ko
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Phil-Dong Moon
- Center for Converging Humanities, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
25
|
Schöpe PC, Torke S, Kobelt D, Kortüm B, Treese C, Dumbani M, Güllü N, Walther W, Stein U. MACC1 revisited - an in-depth review of a master of metastasis. Biomark Res 2024; 12:146. [PMID: 39580452 PMCID: PMC11585957 DOI: 10.1186/s40364-024-00689-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024] Open
Abstract
Cancer metastasis remains the most lethal characteristic of tumors mediating the majority of cancer-related deaths. Identifying key molecules responsible for metastasis, understanding their biological functions and therapeutically targeting these molecules is therefore of tremendous value. Metastasis Associated in Colon Cancer 1 (MACC1), a gene first described in 2009, is such a key driver of metastatic processes, initiating cellular proliferation, migration, invasion, and metastasis in vitro and in vivo. Since its discovery, the value of MACC1 as a prognostic biomarker has been confirmed in over 20 cancer entities. Additionally, several therapeutic strategies targeting MACC1 and its pro-metastatic functions have been developed. In this review, we will provide a comprehensive overview on MACC1, from its clinical relevance, towards its structure and role in signaling cascades as well as molecular networks. We will highlight specific biological consequences of MACC1 expression, such as an increase in stem cell properties, its immune-modulatory effects and induced therapy resistance. Lastly, we will explore various strategies interfering with MACC1 expression and/or its functions. Conclusively, this review underlines the importance of understanding the role of individual molecules in mediating metastasis.
Collapse
Affiliation(s)
- Paul Curtis Schöpe
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Sebastian Torke
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Dennis Kobelt
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Benedikt Kortüm
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Christoph Treese
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Malti Dumbani
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Nazli Güllü
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
- German Cancer Consortium (DKTK), Berlin and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
26
|
Liu X, Fan W, Lin S, Chen J, Zhang S, Li X, Jin M, He Q. Anti-Thrombotic Effect of Protoparaxotriol Saponins From Panax notoginseng Using Zebrafish Model. J Cardiovasc Pharmacol 2024; 84:528-538. [PMID: 39027983 DOI: 10.1097/fjc.0000000000001604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/01/2024] [Indexed: 07/20/2024]
Abstract
ABSTRACT Panax notoginseng has the effect of stimulating circulation to end stasis. Our study was designed to evaluate the anti-thrombotic effect of protoparaxotriol saponins (PTS) from P. notoginseng and the involved mechanisms. A thrombosis model was constructed, and the anti-thrombotic activity of PTS was determined by erythrocyte staining, heart rate, and blood flow velocity. In addition, quantitative real-time polymerase chain reaction was used to identify changes in the expression of genes related to coagulation, inflammation, and apoptosis. PTS alleviated arachidonic acid-induced caudal vein thrombosis, restored blood flow, and increased the area of cardiac erythrocyte staining, heart rate, and blood flow velocity. It reduced the ponatinib-induced cerebral thrombus area and decreased the intensity of erythrocyte staining. The quantitative polymerase chain reaction data showed that the anti-thrombotic effect of PTS was mediated by suppression of genes related to coagulation, inflammation, and apoptosis and also involved inhibition of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathways.
Collapse
Affiliation(s)
- Xin Liu
- Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Yang M, Chen T, Xu Y, Liu Q, Xu X. Study on the mechanism of Shenmai injection in the treatment of sepsis. J Cell Mol Med 2024; 28:e70201. [PMID: 39584444 PMCID: PMC11586680 DOI: 10.1111/jcmm.70201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024] Open
Abstract
Shenmai injection (SMI) is widely used in the clinical treatment of sepsis, but its mechanism is not yet clear. This study aimed to explore the molecular mechanism through network pharmacology, bioinformatics, and molecular docking technologies. The active ingredients and targets of SMI were screened through traditional Chinese medicine databases and the Swiss Target Prediction database, respectively. The disease genes were searched using GEO and GeneCards databases, and Venn mapping was used to screen potential therapeutic targets. The key targets were selected using Cytoscape 3.9.1 software. The BioGPS database was used to evaluate the expression of these targets in tissues/cells. The DAVID database is used for enrichment analysis. Molecular docking technology was used to evaluate the interaction between these targets and core active ingredients. 122 potential therapeutic targets and 28 key targets were identified. Forty-six potential therapeutic targets showed highly specific expression in 40 tissues/cells. The PI3K-AKT, RAP1, and MAPK signalling pathways are highly enriched. The molecular docking results showed good interactions. This study systematically analysed the mechanism of SMI in treating sepsis, involving multiple targets and pathways, possibly related to anti-inflammatory, anti-oxidative stress, and immune regulation, providing reference value for future basic research of sepsis.
Collapse
Affiliation(s)
- Mengxia Yang
- Beijing Hospital of Traditional Chinese MedicineCapital Medical UniversityBeijingChina
- Beijing institute of Traditional Chinese MedicineBeijingChina
- Graduate School of Beijing University of Chinese MedicineBeijingChina
| | - Tengfei Chen
- Beijing Hospital of Traditional Chinese MedicineCapital Medical UniversityBeijingChina
| | - Yue Xu
- China Science and Technology Development Center for Chinese MedicineBeijingChina
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese MedicineCapital Medical UniversityBeijingChina
- Beijing institute of Traditional Chinese MedicineBeijingChina
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese MedicineCapital Medical UniversityBeijingChina
- Beijing institute of Traditional Chinese MedicineBeijingChina
| |
Collapse
|
28
|
Couty N, Estienne A, Le Lay S, Rame C, Chevaleyre C, Allard-Vannier E, Péchoux C, Guerif F, Vasseur C, Aboulouard S, Salzet M, Dupont J, Froment P. Human ovarian extracellular vesicles proteome from polycystic ovary syndrome patients associate with follicular development alterations. FASEB J 2024; 38:e70113. [PMID: 39436214 DOI: 10.1096/fj.202400521rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
The development of the ovarian follicle requires the presence of several factors that come from the blood and follicular cells. Among these factors, extracellular vesicles (EVs) represent an original communication pathway inside the ovarian follicle. Recently, EVs have been shown to play potential roles in follicular development and reproduction-related disorders, including the polycystic ovary syndrome (PCOS). The proteomic analysis of sEVs isolated from FF in comparison to sEVs purified from plasma has shown a specific pattern of proteins secreted by ovarian steroidogenic cells such as granulosa cells. Thus, a human granulosa cell line exposed to sEVs from FF of normal patients increased their progesterone, estradiol, and testosterone secretion. However, if the sEVs were derived from FF of PCOS patients, the activity of stimulating progesterone production was lost. Stimulation of steroidogenesis by sEVs was associated with an increase in the expression of the StAR gene. In addition, sEVs from FF increased cell proliferation and migration of granulosa cells, and this phenomenon was amplified if sEVs were derived from PCOS patients. Interestingly, STAT3 is a protein overexpressed in sEVs from PCOS patients interacting with most of the cluster of proteins involved in the phenotype observed (cell proliferation, migration, and steroid production) in granulosa cells. In conclusion, this study has demonstrated that sEVs derived from FF could regulate directly the granulosa cell activity. The protein content in sEVs from FF is different in the case of PCOS syndrome and could perturb the granulosa cell functions, including inflammation, steroidogenesis, and cytoskeleton architecture.
Collapse
Affiliation(s)
- Noemie Couty
- INRAE, CNRS, Université de Tours, PRC, Nouzilly, France
| | | | - Soazig Le Lay
- L'Institut du Thorax, CNRS, INSERM, Nantes Université, Nantes, France
- Université Angers, SFR ICAT, Angers, France
| | | | | | | | - Christine Péchoux
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | | | - Claudine Vasseur
- Centre de fertilité, Pôle Santé Léonard de Vinci, Chambray-lès-Tours, France
| | - Soulaimane Aboulouard
- INSERM U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Université Lille 1, Villeneuve d'Ascq, France
| | - Michel Salzet
- INSERM U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse (PRISM), Université Lille 1, Villeneuve d'Ascq, France
| | - Joelle Dupont
- INRAE, CNRS, Université de Tours, PRC, Nouzilly, France
| | | |
Collapse
|
29
|
Chen Q, Wang W, Xu Q, Dai Y, Zhu X, Chen Z, Sun N, Leung C, Gao F, Wu K. The enhancing effects of selenomethionine on harmine in attenuating pathological cardiac hypertrophy via glycolysis metabolism. J Cell Mol Med 2024; 28:e70124. [PMID: 39351650 PMCID: PMC11443162 DOI: 10.1111/jcmm.70124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Pathological cardiac hypertrophy, a common feature in various cardiovascular diseases, can be more effectively managed through combination therapies using natural compounds. Harmine, a β-carboline alkaloid found in plants, possesses numerous pharmacological functions, including alleviating cardiac hypertrophy. Similarly, Selenomethionine (SE), a primary organic selenium source, has been shown to mitigate cardiac autophagy and alleviate injury. To explores the therapeutic potential of combining Harmine with SE to treat cardiac hypertrophy. The synergistic effects of SE and harmine against cardiac hypertrophy were assessed in vitro with angiotensin II (AngII)-induced hypertrophy and in vivo using a Myh6R404Q mouse model. Co-administration of SE and harmine significantly reduced hypertrophy-related markers, outperforming monotherapies. Transcriptomic and metabolic profiling revealed substantial alterations in key metabolic and signalling pathways, particularly those involved in energy metabolism. Notably, the combination therapy led to a marked reduction in the activity of key glycolytic enzymes. Importantly, the addition of the glycolysis inhibitor 2-deoxy-D-glucose (2-DG) did not further potentiate these effects, suggesting that the antihypertrophic action is predominantly mediated through glycolytic inhibition. These findings highlight the potential of SE and harmine as a promising combination therapy for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Qi Chen
- Wuxi School of MedicineJiangnan UniversityWuxiJiangsuP. R. China
| | - Wen‐Yan Wang
- Wuxi School of MedicineJiangnan UniversityWuxiJiangsuP. R. China
| | - Qing‐Yang Xu
- Wuxi School of MedicineJiangnan UniversityWuxiJiangsuP. R. China
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical NeurobiologySchool of Basic Medical Sciences, Fudan UniversityShanghaiP. R. China
| | - Yan‐Fa Dai
- Wuxi School of MedicineJiangnan UniversityWuxiJiangsuP. R. China
| | - Xing‐Yu Zhu
- Wuxi School of MedicineJiangnan UniversityWuxiJiangsuP. R. China
| | - Zhao‐Yang Chen
- Department of Cardiology, Heart Center of Fujian ProvinceFujian Medical University Union HospitalFuzhouFujianP. R. China
| | - Ning Sun
- Wuxi School of MedicineJiangnan UniversityWuxiJiangsuP. R. China
- Department of Physiology and Pathophysiology, State Key Laboratory of Medical NeurobiologySchool of Basic Medical Sciences, Fudan UniversityShanghaiP. R. China
| | - Chung‐Hang Leung
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences, University of MacauMacaoP. R. China
| | - Fei Gao
- Department of cardiology, Beijing An Zhen HospitalCapital Medical UniversityChaoyangBeijingP. R. China
| | - Ke‐Jia Wu
- Wuxi School of MedicineJiangnan UniversityWuxiJiangsuP. R. China
| |
Collapse
|
30
|
Mokhtari T, El-Meghawry El-Kenawy A. Molecular mechanisms of Schisandra chinensis in treating depression-neuropathic pain comorbidity by network pharmacology and molecular docking analysis. Neuroscience 2024; 555:92-105. [PMID: 39032805 DOI: 10.1016/j.neuroscience.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/10/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
This study utilized network pharmacology and docking analyses to explore a groundbreaking therapeutic approach for managing the neuropathic pain and depressive disorder (NP/DD) comorbidity. Schisandra chinensis (SC), a common Chinese medicine, has demonstrated numerous beneficial effects in treating neuropsychological disorders. The main objective of this study was to identify potential bioactive components of SC and investigate their interactions with relevant target genes associated with NP/DD. To gain insights into the underlying molecular mechanisms, GO and KEGG analyses were conducted. Furthermore, molecular docking analysis was employed to validate the therapeutic relevance of SC's active ingredients. Seven bioactive components of SC, namely Longikaurin A, Deoxyharringtonine, Angeloylgomisin O, Schisandrin B, Gomisin A, Gomisin G, and Gomisin R, exhibited effectiveness in the treatment of NP/DD. From this list, the first five components were selected for further analysis. The analyses revealed a complex network of interactions between the targets of SC and NP/DD, providing valuable information about the molecular mechanisms involved in the treatment of NP/DD with SC. SC components demonstrated the ability to regulate pathways involving tumor necrosis factor (TNF), vascular endothelial growth factor (VEGF), and other growth hormones (GH). Overall, this study contributes to our understanding of the molecular mechanisms underlying the effects of SC in treating NP/DD. Further investigation is necessary to explore the therapeutic potential of SC as a viable strategy for NP/DD comorbidity. These findings lay a solid foundation for future research endeavors in this field, holding potential implications for the development of novel therapeutic interventions targeting NP/DD.
Collapse
Affiliation(s)
- Tahmineh Mokhtari
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China; Department of Histology and Embryology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.
| | | |
Collapse
|
31
|
Méndez-Martínez M, Zamilpa A, Zavala-Sánchez MA, Almanza-Pérez JC, Jiménez-Ferrer JE, Herrera-Ruiz M, González-Cortázar M, Cervantes-Torres J, Fragoso G, Rosas-Salgado G. Anti-adipogenic effect of Malva parviflora on 3T3-L1 adipocytes. PLoS One 2024; 19:e0306903. [PMID: 39116155 PMCID: PMC11309439 DOI: 10.1371/journal.pone.0306903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/24/2024] [Indexed: 08/10/2024] Open
Abstract
Malva parviflora has shown anti-inflammatory, antihypertensive, antihyperlipidemic, and hypoglycemic effects. This study is aimed to evaluate the anti-adipogenic effect of M. parviflora on 3T3-L1 adipocytes. Fibroblast differentiation was induced either in the absence or presence of M. parviflora fractions (F3, F4, F7, F12, F13, F17, F18 and F19) for 4 days; F17 and 18 were the most effective fractions in reducing intracellular lipid accumulation (by 25.6% and 23.1%, respectively). EC50 of F17 and F18 (14 μg/mL and 17 μg/mL, respectively) were used to evaluate their anti adipogenic effect. After 10 days of inducing differentiation in the absence or presence of the extracts at the EC50 of F17 and F18, lipid accumulation, the concentration of interleukin 6 (IL-6) were measured in the culture medium; the presence of PPAR-γ, AKT, and p-AKT was also determined. In differentiated adipocytes (C2), F17 maintained intracellular lipid concentration at levels comparable to metformin, while decreasing PPAR-γ and increasing p-AKT presence; it also prevented IL-6 expression. F17 consists of alanine, valine, phenylalanine, and proline. On the other hand, F18 reduced intracellular lipid concentrations, prevented the increase of PPAR-γ and p-AKT, and maintained IL-6 expression at similar levels as metformin. F18 is mainly constituted by alanine, valine, proline, and sucrose. In conclusion, M. parviflora fractions (F17 and F18) control the process of adipogenesis, lipogenesis, and cellular dysfunction.
Collapse
Affiliation(s)
- Marisol Méndez-Martínez
- División de Ciencias Biológicas y de la Salud, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Mexico City, Mexico
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec, Morelos, Mexico
| | - Miguel A. Zavala-Sánchez
- División de Ciencias Biológicas y de la Salud, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Julio C. Almanza-Pérez
- Laboratorio de Farmacología, División de C.B.S., Departamento de Ciencias de la Salud, Unidad Iztapalapa, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - J. Enrique Jiménez-Ferrer
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec, Morelos, Mexico
| | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec, Morelos, Mexico
| | - Manasés González-Cortázar
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec, Morelos, Mexico
| | - Jaquelynne Cervantes-Torres
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gladis Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gabriela Rosas-Salgado
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
32
|
Martucci NJ, Stoops J, Bowen W, Orr A, Cotner MC, Michalopoulos GK, Bhushan B, Mars WM. A Novel Role for the Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Delta Isoform in Hepatocellular Proliferation. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1511-1527. [PMID: 38705383 PMCID: PMC11393825 DOI: 10.1016/j.ajpath.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/09/2024] [Accepted: 03/22/2024] [Indexed: 05/07/2024]
Abstract
The phosphatidylinositol-4,5-bisphosphate 3-kinase delta isoform (Pik3cd), usually considered immune-specific, was unexpectedly identified as a gene potentially related to either regeneration and/or differentiation in animals lacking hepatocellular Integrin Linked Kinase (ILK). Since a specific inhibitor (Idelalisib, or CAL101) for the catalytic subunit encoded by Pik3cd (p110δ) has reported hepatotoxicity when used for treating chronic lymphocytic leukemia and other lymphomas, the authors aimed to elucidate whether there is a role for p110δ in normal liver function. To determine the effect on normal liver regeneration, partial hepatectomy (PHx) was performed using mice in which p110δ was first inhibited using CAL101. Inhibition led to over a 50% decrease in proliferating hepatocytes in the first 2 days after PHx. This difference correlated with phosphorylation changes in the HGF and EGF receptors (MET and EGFR, respectively) and NF-κB signaling. Ingenuity Pathway Analyses implicated C/EBPβ, HGF, and the EGFR heterodimeric partner, ERBB2, as three of the top 20 regulators downstream of p110δ signaling because their pathways were suppressed in the presence of CAL101 at 1 day post-PHx. A regulatory role for p110δ signaling in mouse and rat hepatocytes through MET and EGFR was further verified using hepatocyte primary cultures, in the presence or absence of CAL101. Combined, these data support a role for p110δ as a downstream regulator of normal hepatocytes when stimulated to proliferate.
Collapse
Affiliation(s)
- Nicole J Martucci
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Stoops
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William Bowen
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anne Orr
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mary-Claire Cotner
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Bharat Bhushan
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wendy M Mars
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
33
|
Bartkowiak-Wieczorek J, Malesza M, Malesza I, Hadada T, Winkler-Galicki J, Grzelak T, Mądry E. Methylsulfinyl Hexyl Isothiocyanate (6-MSITC) from Wasabi Is a Promising Candidate for the Treatment of Cancer, Alzheimer's Disease, and Obesity. Nutrients 2024; 16:2509. [PMID: 39125389 PMCID: PMC11313713 DOI: 10.3390/nu16152509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Methylsulfinyl hexyl isothiocyanate (6-MSITC) isolated from Eutrema japonicum is a promising candidate for the treatment of breast cancer, colorectal and stomach cancer, metabolic syndrome, heart diseases, diabetes, and obesity due to its anti-inflammatory and antioxidant properties. Also, its neuroprotective properties, improving cognitive function and protecting dopaminergic neurons, make it an excellent candidate for treating neurodegenerative diseases like dementia, Alzheimer's, and Parkinson's disease. 6-MSITC acts on many signaling pathways, such as PPAR, AMPK, PI3K/AKT/mTOR, Nrf2/Keap1-ARE, ERK1/2-ELK1/CHOP/DR5, and MAPK. However, despite the very promising results of in vitro and in vivo animal studies and a few human studies, the molecule has not yet been thoroughly tested in the human population. Nonetheless, wasabi should be classified as a "superfood" for the primary and secondary prevention of human diseases. This article reviews the current state-of-the-art research on 6-MSITC and its potential clinical uses, discussing in detail the signaling pathways activated by the molecule and their interactions.
Collapse
Affiliation(s)
- Joanna Bartkowiak-Wieczorek
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| | - Michał Malesza
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| | - Ida Malesza
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Tomasz Hadada
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| | - Jakub Winkler-Galicki
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| | - Teresa Grzelak
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| | - Edyta Mądry
- Physiology Department, Poznan University of Medical Sciences, 6, Święcickiego Street, 60-781 Poznan, Poland; (M.M.); (T.H.); (J.W.-G.); (T.G.); (E.M.)
| |
Collapse
|
34
|
Yousof SM, Shehata SA, Ismail EA, Abd El-moneam SM, Mansour BS, Farag MA, Elshamy AI, El-Nasser G. El Gendy A, Serag A, Abd El-Fadeal NM, Abdel-Karim RI, Mostafa MM, El-Sheikh DH, Zayed MA. Acacia saligna extract alleviates quetiapine-induced sexual toxicity in male albino rats: Insights from UPLC-MS/MS metabolite profiling, structural and PI3K/NF-κB pathway assessments. Heliyon 2024; 10:e33993. [PMID: 39071580 PMCID: PMC11280294 DOI: 10.1016/j.heliyon.2024.e33993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Background Quetiapine (QET) abuse has increased due to its anxiolytic and hedonic effects, necessitating protective adjunct treatments. Acacia saligna (A. saligna) flowers, used in traditional medicine, have potential health benefits. Aim To investigate the protective role of A. saligna flower extract against QET-induced sexual toxicity, and to elucidate the possible underlying mechanisms through metabolomic and physiological studies. Methods A. saligna extract was subjected to metabolite profiling via High-Resolution Ultra-Performance Liquid Chromatography-Mass Spectrometry (UPLC-ESI-qTOF-MS). Forty-eight adult male albino rats were assigned into six groups for 30 days. The intracavernosal pressure (ICP), semen, biochemical, hormonal, histological, genetic and Western blot (WB) analyses were determined. Results A. saligna extract is rich in phenolic compounds, flavonoids, tannins, and unsaturated fatty acids. QET significantly decreased ICP and negatively affected semen parameters. A. saligna mitigated decreased sperm motility and ameliorated overexpressed proinflammatory genes in QET-55 group. A. saligna ameliorated the reduction of the antioxidant biomarkers, testosterone, luteinizing hormone (LH), and follicle-stimulating hormone (FSH), concurrent with downregulation of the nuclear factor kappa B (NF-κB) protein. A. saligna counteracted the disrupted testicular and prostatic structures revealed by histological examination. Conclusion The extract from A. saligna, which contains a high concentration of antioxidants and anti-inflammatory chemicals, effectively mitigates sexual toxicity caused by QET. This study provided the first known explanation of the hypothesized processes behind the protective properties of A. saligna through biological, biochemical, and histological parameters. The results emphasize the potential of A. saligna as a safeguarding agent against drug-induced sexual toxicity.
Collapse
Affiliation(s)
- Shimaa Mohammad Yousof
- Medical Physiology Department, Faculty of Medicine, King Abdulaziz University, Rabigh Branch, 21589, Saudi Arabia
- Medical Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Neuroscience and Geroscience Unit, King Fahad Research Centre, King Abdulaziz University, KSA
| | - Shaimaa A. Shehata
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine- Suez Canal University, Ismailia, 41522, Egypt
| | - Ezzat A. Ismail
- Urology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Samar M. Abd El-moneam
- Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Basma S.A. Mansour
- Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Mohamed A. Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr el Aini, Giza 12613, Egypt
| | - Abdelsamed I. Elshamy
- Department of Natural Compounds Chemistry, National Research Center, Dokki, Giza 12622, Egypt
| | - Abd El-Nasser G. El Gendy
- Medicinal and Aromatic Plants Research Department, National Research Centre, 33 El Bohouth St., Dokki, Giza, 12622, Egypt
| | - Ahmed Serag
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, 11751, Egypt
| | - Noha M. Abd El-Fadeal
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
- Biochemistry Department, Ibn Sina National College for Medical Studies, Jeddah, 22421, Saudi Arabia
| | - Rehab Ibrahim Abdel-Karim
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine- Suez Canal University, Ismailia, 41522, Egypt
| | - Mostafa M. Mostafa
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Rabigh Branch, 21589, Saudi Arabia
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Kasr Al Ainy, Cairo, 11562, Egypt
| | - Dina H. El-Sheikh
- Medical Physiology Department, Faculty of Medicine, Prince Sattam Bin Abdulaziz University, Al Kharj Branch, 16273, Saudi Arabia
- Medical Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr el Aini, Giza 12613, Egypt
| | - Mohamed A. Zayed
- Medical Physiology Department, Faculty of Medicine, King Abdulaziz University, Rabigh Branch, 21589, Saudi Arabia
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia, 13829, Egypt
- Neuroscience and Geroscience Unit, King Fahad Research Centre, King Abdulaziz University, KSA
| |
Collapse
|
35
|
Gamal RM, Hazem SH, Hamed MF, Abdelaziz RR. PI3K inhibitor "alpelisib" alleviates methotrexate induced liver injury in mice and potentiates its cytotoxic effect against MDA-MB-231 triple negative breast cancer cell line. Toxicol Appl Pharmacol 2024; 488:116979. [PMID: 38797265 DOI: 10.1016/j.taap.2024.116979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Hepatotoxicity is the main off-target effect of methotrexate (MTX) limiting its effective clinical use. Besides, MDA-MB231 breast cancer cells show chemoresistance, partly via PI3K/AKT pathway. Therefore, we investigated the ameliorative potentials of the PI3K inhibitor, alpelisib (ALP) on MTX-induced hepatotoxicity (in vivo) and the restraining potentials of ALP on MDA-MB231 chemoresistance to MTX (in vitro). Twenty-eight male BALB/c mice were divided into 4 groups. In treatment groups, mice were administered ALP (2.5 and 5 mg/kg) for 5 days and MTX (20 mg/kg) from day 2 till day 5. The results showed that ALP restored hepatic architecture, reduced immune cell infiltration (F4/80, Ly6G and MPO) and repressed the rise in liver enzymes (AST and ALT) induced by MTX. Additionally, ALP rectified the MTX-induced disruption of cellular oxidant status by boosting antioxidant defense systems (HO-1 and GSH) and repressing lipid peroxidation (MDA and 4-HNE). Finally, ALP curbed MTX-induced hepatocyte apoptosis (NF-κB and BAX) and shifted the cytokine milieu away from inflammation (IL-17, IL-22, IL-6 and IL- 10). The results of the in vitro experiments revealed that ALP alone and in combination with MTX, synergistically, reduced cancer cell viability (MTT assay), migration (wound healing assay) and their capacity to establish colonies (colony formation assay) as compared to MTX alone. RT-PCR revealed the antiproliferative (Bcl-2) and proapoptotic (BAX) potentials of ALP and ALP/MTX combination especially after 24 h. In conclusion, targeting PI3K/AKT pathway is a promising strategy in triple negative breast cancer patients by ameliorating hepatotoxicity and restraining chemoresistance to chemotherapy.
Collapse
Affiliation(s)
- Rana M Gamal
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Sara H Hazem
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Mohamed F Hamed
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Rania R Abdelaziz
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
36
|
Medeiros M, Guenka S, Bastos D, Oliveira KL, Brassesco MS. Amicis Omnia Sunt Communia: NF-κB Inhibition as an Alternative to Overcome Osteosarcoma Heterogeneity. Pharmaceuticals (Basel) 2024; 17:734. [PMID: 38931401 PMCID: PMC11206879 DOI: 10.3390/ph17060734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Tumor heterogeneity poses a significant challenge in osteosarcoma (OS) treatment. In this regard, the "omics" era has constantly expanded our understanding of biomarkers and altered signaling pathways (i.e., PI3K/AKT/mTOR, WNT/β-catenin, NOTCH, SHH/GLI, among others) involved in OS pathophysiology. Despite different players and complexities, many commonalities have been described, among which the nuclear factor kappa B (NF-κB) stands out. Its altered activation is pervasive in cancer, with pleiotropic action on many disease-relevant traits. Thus, in the scope of this article, we highlight the evidence of NF-κB dysregulation in OS and its integration with other cancer-related pathways while we summarize the repertoire of compounds that have been described to interfere with its action. In silico strategies were used to demonstrate that NF-κB is closely coordinated with other commonly dysregulated signaling pathways not only by functionally interacting with several of their members but also by actively participating in the regulation of their transcription. While existing inhibitors lack selectivity or act indirectly, the therapeutic potential of targeting NF-κB is indisputable, first for its multifunctionality on most cancer hallmarks, and secondly, because, as a common downstream effector of the many dysregulated pathways influencing OS aggressiveness, it turns complex regulatory networks into a simpler picture underneath molecular heterogeneity.
Collapse
Affiliation(s)
- Mariana Medeiros
- Cell Biology Department, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil;
| | - Sophia Guenka
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| | - David Bastos
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| | - Karla Laissa Oliveira
- Regional Blood Center, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14051-140, São Paulo, Brazil;
| | - María Sol Brassesco
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| |
Collapse
|
37
|
Rana H, Truong NR, Johnson B, Baharlou H, Herbert JJ, Kandasamy S, Goddard R, Cohen RC, Wines M, Nasr N, Harman AN, Bertram KM, Sandgren KJ, Cunningham AL. Herpes simplex virus spreads rapidly in human foreskin, partly driven by chemokine-induced redistribution of Nectin-1 on keratinocytes. PLoS Pathog 2024; 20:e1012267. [PMID: 38857290 PMCID: PMC11164381 DOI: 10.1371/journal.ppat.1012267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/17/2024] [Indexed: 06/12/2024] Open
Abstract
HSV infects keratinocytes in the epidermis of skin via nectin-1. We established a human foreskin explant infection model to investigate HSV entry and spread. HSV1 entry could only be achieved by the topical application of virus via high density microarray projections (HD-MAPs) to the epidermis, which penetrated beyond one third of its thickness, simulating in vivo microtrauma. Rapid lateral spread of HSV1 to a mean of 13 keratinocytes wide occurred after 24 hours and free virus particles were observed between keratinocytes, consistent with an intercellular route of spread. Nectin-1 staining was markedly decreased in foci of infection in the epidermis and in the human keratinocyte HaCaT cell line. Nectin-1 was redistributed, at the protein level, in adjacent uninfected cells surrounding infection, inducible by CCL3, IL-8 (or CXCL8), and possibly CXCL10 and IL-6, thus facilitating spread. These findings provide the first insights into HSV1 entry and spread in human inner foreskin in situ.
Collapse
Affiliation(s)
- Hafsa Rana
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Naomi R. Truong
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Blake Johnson
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Heeva Baharlou
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Jason J. Herbert
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Robert Goddard
- Research and Development, Vaxxas Pty Ltd., Brisbane, Queensland, Australia
| | - Ralph C. Cohen
- University of Sydney and Australian National University, Children’s Hospital at Westmead, New South Wales, Australia
| | - Michael Wines
- Urology, Sydney Adventist Hospital, Wahroonga, New South Wales, Australia
| | - Najla Nasr
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew N. Harman
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kirstie M. Bertram
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kerrie J. Sandgren
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anthony L. Cunningham
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
38
|
Chakraborty R, Tabassum H, Parvez S. Dantrolene alleviates mitochondrial dysfunction and neuroinflammation in traumatic brain injury by modulating the NF-ĸβ/Akt pathway. Biochem Pharmacol 2024; 224:116244. [PMID: 38685280 DOI: 10.1016/j.bcp.2024.116244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/04/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Traumatic brain injury (TBI) triggers a bevy of changes including mitochondrial dysfunction, apoptosis, oxidative stress, neurobehavioural impairment, and neuroinflammation, among others. Dantrolene (DNT), a muscle relaxant which inhibits intracellular Ca2+ signaling from the ER, has been repurposed as a potential neuroprotective agent in various neurological diseases. However, there have been limited studies on whether it can mitigate TBI-induced deficits and restore impaired mitochondrial dynamics. This study sought to evaluate whether Dantrolene can potentially provide neuroprotection in an in vivo model of TBI. Male wistar rats subjected to TBI were treated with DNT (10 mg/kg) 1 h and 12 h post surgery. Animals were assessed 24 h post-TBI to evaluate neurobehavioural deficits and cerebral edema. We evaluated the protein expressions of apoptotic, autophagic, and neuroinflammatory markers by immunoblotting, as well as Mitochondrial Membrane Potential (MMP) and Reactive Oxygen Species (ROS) via Flow Cytometry to ascertain the effects of DNT on TBI. We further analysed immunofluorescence staining with Glial Fibrillary Acidic Protein (GFAP) and immunohistochemistry with NF-κβ to investigate neuroinflammation. H&E staining was also performed post-TBI. Our findings revealed DNT administration inhibits mitochondria-mediated apoptotis and reduces heightened oxidative stress. DNT treatment was also found to reverse neurobehavioural impairments and offer neuroprotection by preserving neuronal architechture. We also demonstrated that DNT inhibits neuronal autophagy and alleviates neuroinflammation following TBI by modulating the NF-κβ/Akt signaling pathway. Thus, our results suggest a novel application of DNT in ameliorating the multitude of deficits induced by TBI, thereby conferring neuroprotection.
Collapse
Affiliation(s)
- Rohan Chakraborty
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Heena Tabassum
- Division of Basic Medical Sciences, Indian Council of Medical Research, Government of India, V. Ramalingaswamy Bhawan, New Delhi 110029, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India.
| |
Collapse
|
39
|
Göttig L, Schreiner S. E4orf1: The triple agent of adenovirus - Unraveling its roles in oncogenesis, infectious obesity and immune responses in virus replication and vector therapy. Tumour Virus Res 2024; 17:200277. [PMID: 38428735 PMCID: PMC10937242 DOI: 10.1016/j.tvr.2024.200277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
Human Adenoviruses (HAdV) are nearly ubiquitous pathogens comprising numerous sub-types that infect various tissues and organs. Among many encoded proteins that facilitate viral replication and subversion of host cellular processes, the viral E4orf1 protein has emerged as an intriguing yet under-investigated player in the complex interplay between the virus and its host. E4orf1 has gained attention as a metabolism activator and oncogenic agent, while recent research is showing that E4orf1 may play a more important role in modulating cellular pathways such as PI3K-Akt-mTOR, Ras, the immune response and further HAdV replication stages than previously anticipated. In this review, we aim to explore the structure, molecular mechanisms, and biological functions of E4orf1, shedding light on its potentially multifaceted roles during HAdV infection, including metabolic diseases and oncogenesis. Furthermore, we discuss the role of functional E4orf1 in biotechnological applications such as Adenovirus (AdV) vaccine vectors and oncolytic AdV. By dissecting the intricate relationships between HAdV types and E4orf1 proteins, this review provides valuable insights into viral pathogenesis and points to promising areas of future research.
Collapse
Affiliation(s)
- Lilian Göttig
- Institute of Virology, School of Medicine, Technical University of Munich, Germany
| | - Sabrina Schreiner
- Institute of Virology, School of Medicine, Technical University of Munich, Germany; Institute of Virology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (Resolving Infection Susceptibility; EXC 2155), Hannover, Germany; Institute of Virology, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
40
|
Xu T, Liu K, Zhang Y, Chen Y, Yin D. EGFR and Hippo signaling pathways are involved in organophosphate esters-induced proliferation and migration of triple-negative breast cancer cells. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:41939-41952. [PMID: 38856849 DOI: 10.1007/s11356-024-33872-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
The widespread application of organophosphate flame retardants has led to pervasive exposure to organophosphate esters (OPEs), prompting considerable concerns regarding their potential health risk to humans. Despite hints from previous research about OPEs' association with breast cancer, their specific effects and underlying mechanisms of triple-negative breast cancer (TNBC) remain unclear. In this study, we investigated the effects of four representative OPEs on cell proliferation, cell cycle regulation, migration, and the expression of genes and proteins associated with the epidermal growth factor receptor (EGFR) and Hippo signaling pathways in TNBC (MDA-MB-231) cells. Our findings revealed that treatment with 1-25 μM triphenyl phosphate (TPHP) and tris(1,3-dichloro-2-propyl) phosphate (TDCIPP) induced TNBC cell proliferation and accelerated cell cycle progression, with upregulation in MYC, CCND1, and BRCA1 mRNA. Moreover, exposure to 1-25 μM TPHP, 10-25 μM TDCIPP, and 1-10 μM tris (2-chloroethyl) phosphate (TCEP) induced MMP2/9 mRNA expression and enhanced migratory capacity, except for 2-ethylhexyl diphenyl phosphate (EHDPP). Mechanistically, four OPEs treatments activated the EGFR-ERK1/2 and EGFR-PI3K/AKT signaling pathways by increasing the transcript of EGFR, ERK1/2, PI3K, and AKT mRNA. OPEs treatment also suppressed the Hippo signaling pathway by inhibiting the expression of MST1 mRNA and phosphorylation of LATS1, leading to the overactivation of YAP1 protein, thereby promoting TNBC cell proliferation and migration. In summary, our study elucidated that activation of the EGFR signaling pathway and suppression of the Hippo signaling pathway contributed to the proliferation, cell cycle dysregulation, and migration of TNBC cells following exposure to OPEs.
Collapse
Affiliation(s)
- Ting Xu
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
- Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, China
| | - Kaiyue Liu
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Yajie Zhang
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Yawen Chen
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China.
- Post-doctoral Research Station of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China.
| | - Daqiang Yin
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, China
| |
Collapse
|
41
|
Zhao K, Wu C, Li X, Niu M, Wu D, Cui X, Zhao H. From mechanism to therapy: the journey of CD24 in cancer. Front Immunol 2024; 15:1401528. [PMID: 38881902 PMCID: PMC11176514 DOI: 10.3389/fimmu.2024.1401528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 06/18/2024] Open
Abstract
CD24 is a glycosylphosphatidylinositol-anchored protein that is expressed in a wide range of tissues and cell types. It is involved in a variety of physiological and pathological processes, including cell adhesion, migration, differentiation, and apoptosis. Additionally, CD24 has been studied extensively in the context of cancer, where it has been found to play a role in tumor growth, invasion, and metastasis. In recent years, there has been growing interest in CD24 as a potential therapeutic target for cancer treatment. This review summarizes the current knowledge of CD24, including its structure, function, and its role in cancer. Finally, we provide insights into potential clinical application of CD24 and discuss possible approaches for the development of targeted cancer therapies.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Caifeng Wu
- Department of Hand and Foot, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangjun Li
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mengchao Niu
- Department of Operation Room, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dan Wu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaofeng Cui
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
42
|
Kim MJ, Lim SG, Cho DH, Lee JY, Suk K, Lee WH. Regulation of inflammatory response by LINC00346 via miR-25-3p-mediated modulation of the PTEN/PI3K/AKT/NF-κB pathway. Biochem Biophys Res Commun 2024; 709:149828. [PMID: 38537596 DOI: 10.1016/j.bbrc.2024.149828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/16/2024] [Accepted: 03/24/2024] [Indexed: 04/13/2024]
Abstract
Long intergenic non-coding RNA 346 (LINC00346) has been reported to be involved in the development of atherosclerosis and specific cancers by affecting signaling pathways. However, its function in inflammation has not been thoroughly studied. Therefore, its expression pattern and function were determined in the human macrophage-like cell line THP-1. Lipopolysaccharide (LPS) treatment induced the expression of LINC00346. LPS-induced NF-κB activation and proinflammatory cytokine expression were suppressed or enhanced by the overexpression or knockdown of LINC00346, respectively. Analyses using dual luciferase assay and decoy RNAs that could block RNA-RNA interactions indicated that LINC00346 improves phosphatase and tensin homolog (PTEN) expression by sponging miR-25-3p. Subsequently, PTEN suppresses phosphoinositide-3 kinase (PI3K)-mediated conversion of phosphatidylinositol-4,5-bisphosphate (PIP2) into phosphatidylinositol-3,4,5-trisphosphate (PIP3) as well as consequent activation of protein kinase B (AKT) and NF-κB. Interestingly, database analysis revealed that the expression levels of LINC00346 and PTEN were simultaneously decreased in breast cancer tissues. Further analyses conducted using a breast cancer cell line, MDA-MB-231, confirmed the functional relationship among LINC00346, miR-25-3p, and PTEN in LPS-induced activation of NF-κB. These results indicate that miR-25-3p-sponging activity of LINC00346 affects the balance between PTEN and PI3K as well as the downstream activation of AKT/NF-κB pathway in inflammatory conditions.
Collapse
Affiliation(s)
- Min-Ji Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Su-Geun Lim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Dong-Hyung Cho
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jun-Yeong Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science & Engineering Institute, BK21 FOUR KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
43
|
Vitali E, Valente G, Panzardi A, Laffi A, Zerbi A, Uccella S, Mazziotti G, Lania A. Pancreatic neuroendocrine tumor progression and resistance to everolimus: the crucial role of NF-kB and STAT3 interplay. J Endocrinol Invest 2024; 47:1101-1117. [PMID: 37882947 DOI: 10.1007/s40618-023-02221-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/09/2023] [Indexed: 10/27/2023]
Abstract
PURPOSE The finding of mTOR overactivation in patients affected by pancreatic neuroendocrine tumors (Pa-NETs) led to their treatment with the mTOR inhibitor everolimus. Unfortunately, the efficacy of everolimus is restricted by the occurrence of resistance. The mechanisms leading to Pa-NETs' progression and resistance are not well understood. Notably, chronic inflammation is implicated in NET development. NF-kB is involved in inflammation and drug resistance mechanisms through the activation of several mediators, including STAT3. In this respect, NF-κB and STAT3 interaction is implicated in the crosstalk between inflammatory and tumor cells. METHODS We investigated the expression of NF-kB in different Pa-NETs by RT-qPCR and immunohistochemistry. Then, we studied the role of NF-κB and STAT3 interplay in QGP-1 cells. Subsequently, we assessed the impact of NF-κB and STAT3 inhibitors in QGP-1 cell proliferation and spheroids growth. Finally, we evaluated the implication of the NF-kB pathway in everolimus-resistant Pa-NET cells. RESULTS We found that the increased NF-kB expression correlates with a higher grade in Pa-NETs. The activation of the STAT3 pathway induced by TNFα is mediated by NF-kB p65. NF-kB p65 and STAT3 inhibitors decrease QGP-1 viability, spheroids growth, and Pa-NETs cell proliferation. These effects are maintained in everolimus-resistant QGP-1R cells. Interestingly, we found that NF-kB, STAT3, IL-8, and SOCS3 are overexpressed in QGP-1R compared to QGP-1. CONCLUSION Since the NF-kB pathway is implicated in Pa-NETs' progression and resistance to everolimus, these data could explain the potential use of NF-kB as a novel therapeutic target in Pa-NET patients.
Collapse
Affiliation(s)
- E Vitali
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Humanitas Research Hospital, Manzoni 56, 20089, Rozzano, Milan, Italy.
| | - G Valente
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Humanitas Research Hospital, Manzoni 56, 20089, Rozzano, Milan, Italy
| | - A Panzardi
- Laboratory of Cellular and Molecular Endocrinology, IRCCS Humanitas Research Hospital, Manzoni 56, 20089, Rozzano, Milan, Italy
| | - A Laffi
- Oncology Unit, IRCCS Humanitas Research Hospital, Manzoni 56, 20089, Rozzano, Milan, Italy
| | - A Zerbi
- Department of Biomedical Sciences, Humanitas University, Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- Surgery Unit, IRCCS Humanitas Research Hospital, Manzoni 56, 20089, Rozzano, Milan, Italy
| | - S Uccella
- Department of Biomedical Sciences, Humanitas University, Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- Pathology Unit, IRCCS Humanitas Research Hospital, Manzoni 56, 20089, Rozzano, ilan, Italy
| | - G Mazziotti
- Department of Biomedical Sciences, Humanitas University, Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- Endocrinology, Diabetology and Andrology Unit, IRCCS Humanitas Research Hospital, Manzoni 54, 20089, Rozzano, Milan, Italy
| | - A Lania
- Department of Biomedical Sciences, Humanitas University, Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- Endocrinology, Diabetology and Andrology Unit, IRCCS Humanitas Research Hospital, Manzoni 54, 20089, Rozzano, Milan, Italy
| |
Collapse
|
44
|
Yin Q, Huang Q, Zhang H, Zhang X, Fan C, Wang H. Anti-rheumatoid arthritis effects of traditional Chinese medicine Fufang Xiaohuoluo pill on collagen-induced arthritis rats and MH7A cells. Front Pharmacol 2024; 15:1374485. [PMID: 38741593 PMCID: PMC11089244 DOI: 10.3389/fphar.2024.1374485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/11/2024] [Indexed: 05/16/2024] Open
Abstract
Background Fufang Xiaohuoluo pill (FFXHL) is a commonly used prescription in clinical practice for treating rheumatoid arthritis in China, yet its specific mechanism remains unclear. This study aims to elucidate the pharmacological mechanisms of FFXHL using both in vivo and in vitro experiments. Methods The collagen-induced arthritis (CIA) rat model was established to evaluate FFXHL's therapeutic impact. Parameters that include paw swelling, arthritis scores, and inflammatory markers were examined to assess the anti-inflammatory and analgesic effects of FFXHL. Human fibroblast-like synoviocytes (MH7A cells) is activated by tumour necrosis factor-alpha (TNF-α) were used to explore the anti-inflammatory mechanism on FFXHL. Results Our findings indicate that FFXHL effectively reduced paw swelling, joint pain, arthritis scores, and synovial pannus hyperplasia. It also lowered serum levels of TNF-α, interleukin-1β (IL1β), and interleukin-6 (IL-6). Immunohistochemical analysis revealed decreased expression of nuclear factor-kappa B (NF-κB) p65 in FFXHL-treated CIA rat joints. In vitro experiments demonstrated FFXHL's ability to decrease protein secretion of IL-1β and IL-6, suppress mRNA expression of matrix metalloproteinases (MMP) -3, -9, and -13, reduce reactive oxygen species (ROS) levels, and inhibit NF-κB p65 translocation in TNF-α stimulated MH7A cells. FFXHL also suppressed protein levels of extracellular signal-regulated kinase (ERK), c-Jun Nterminal kinase (JNK), p38 MAP kinase (p38), protein kinase B (Akt), p65, inhibitor of kappa B kinase α/β (IKKα/β), Toll-like receptor 4 (TLR4), and myeloid differentiation primary response 88 (MyD88) induced by TNF-α in MH7A cells. Conclusion The findings imply that FFXHL exhibits significant anti-inflammatory and antiarthritic effects in both CIA rat models and TNF-α-induced MH7A cells. The potential mechanism involves the inactivation of TLR4/MyD88, mitogen-activated protein kinases (MAPKs), NF-κB, and Akt pathways by FFXHL.
Collapse
Affiliation(s)
- Qiong Yin
- Scientific Research Institute of Beijing Tongrentang Co., Ltd., Beijing, China
| | - Qian Huang
- Scientific Research Institute of Beijing Tongrentang Co., Ltd., Beijing, China
| | - Hantao Zhang
- Scientific Research Institute of Beijing Tongrentang Co., Ltd., Beijing, China
| | - Xiaodi Zhang
- Scientific Research Institute of Beijing Tongrentang Co., Ltd., Beijing, China
| | | | - Hongping Wang
- Scientific Research Institute of Beijing Tongrentang Co., Ltd., Beijing, China
| |
Collapse
|
45
|
Schwartz L, Salamon K, Simoni A, Eichler T, Jackson AR, Murtha M, Becknell B, Kauffman A, Linn-Peirano S, Holdsworth N, Tyagi V, Tang H, Rust S, Cortado H, Zabbarova I, Kanai A, Spencer JD. Insulin receptor signaling engages bladder urothelial defenses that limit urinary tract infection. Cell Rep 2024; 43:114007. [PMID: 38517889 PMCID: PMC11094371 DOI: 10.1016/j.celrep.2024.114007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/10/2024] [Accepted: 03/11/2024] [Indexed: 03/24/2024] Open
Abstract
Urinary tract infections (UTIs) commonly afflict people with diabetes. To better understand the mechanisms that predispose diabetics to UTIs, we employ diabetic mouse models and altered insulin signaling to show that insulin receptor (IR) shapes UTI defenses. Our findings are validated in human biosamples. We report that diabetic mice have suppressed IR expression and are more susceptible to UTIs caused by uropathogenic Escherichia coli (UPEC). Systemic IR inhibition increases UPEC susceptibility, while IR activation reduces UTIs. Localized IR deletion in bladder urothelium promotes UTI by increasing barrier permeability and suppressing antimicrobial peptides. Mechanistically, IR deletion reduces nuclear factor κB (NF-κB)-dependent programming that co-regulates urothelial tight junction integrity and antimicrobial peptides. Exfoliated urothelial cells or urine samples from diabetic youths show suppressed expression of IR, barrier genes, and antimicrobial peptides. These observations demonstrate that urothelial insulin signaling has a role in UTI prevention and link IR to urothelial barrier maintenance and antimicrobial peptide expression.
Collapse
Affiliation(s)
- Laura Schwartz
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH 43205, USA
| | - Kristin Salamon
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Aaron Simoni
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Tad Eichler
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Ashley R Jackson
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH 43205, USA
| | - Matthew Murtha
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Brian Becknell
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH 43205, USA
| | - Andrew Kauffman
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Tulane University, New Orleans, LA 70118, USA
| | - Sarah Linn-Peirano
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Department of Veterinary Biosciences, The Ohio State University College of Veterinary Medicine, Columbus, OH 43210, USA
| | - Natalie Holdsworth
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Ohio University Heritage College of Osteopathic Medicine, Athens, OH 45701, USA
| | - Vidhi Tyagi
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Hancong Tang
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Steve Rust
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Hanna Cortado
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Irina Zabbarova
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Anthony Kanai
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH 43205, USA.
| |
Collapse
|
46
|
Jia X, Gu M, Dai J, Wang J, Zhang Y, Pang Z. Quercetin attenuates Pseudomonas aeruginosa-induced acute lung inflammation by inhibiting PI3K/AKT/NF-κB signaling pathway. Inflammopharmacology 2024; 32:1059-1076. [PMID: 38310155 DOI: 10.1007/s10787-023-01416-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/16/2023] [Indexed: 02/05/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that commonly causes infections in immunocompromised individuals with significant morbidity and mortality. Quercetin is a natural flavonoid abundantly present in fruits and vegetables, exerting potent anti-inflammatory effects in treatment of various diseases. However, the molecular mechanisms of quercetin in treatment of P. aeruginosa-induced acute lung inflammation are unclear. In this study, we exploited network pharmacology- and molecular docking-based approach to explore the potential mechanisms of quercetin against P. aeruginosa pneumonia, which was further validated via in vivo and in vitro experiments. The in vivo experiments demonstrated that quercetin alleviated the P. aeruginosa-induced lung injury by diminishing neutrophil infiltration and production of proinflammatory cytokines (IL-1β, IL-6, and TNF), which was associated with decreased mortality. Moreover, the quercetin-treated mice displayed decreased phosphorylation levels of PI3K, AKT, IκBα, and NF-κB p65 in lung tissues compared to non-drug-treated mice. Similarly, the in vitro study showed that the phosphorylation of these regulatory proteins and production of the proinflammatory cytokines were impaired in the quercetin-pretreated macrophages upon P. aeruginosa infection. Altogether, this study suggested that quercetin reduced the P. aeruginosa-induced acute lung inflammation by suppressing PI3K/AKT/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiaolei Jia
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
| | - Mengdi Gu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
| | - Jiangqin Dai
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
| | - Jue Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China
| | - Yingying Zhang
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China.
| | - Zheng Pang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, 4655 University Road, Jinan, 250355, China.
| |
Collapse
|
47
|
Salmeri N, Viganò P, Cavoretto P, Marci R, Candiani M. The kisspeptin system in and beyond reproduction: exploring intricate pathways and potential links between endometriosis and polycystic ovary syndrome. Rev Endocr Metab Disord 2024; 25:239-257. [PMID: 37505370 DOI: 10.1007/s11154-023-09826-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Endometriosis and polycystic ovary syndrome (PCOS) are two common female reproductive disorders with a significant impact on the health and quality of life of women affected. A novel hypothesis by evolutionary biologists suggested that these two diseases are inversely related to one another, representing a pair of diametrical diseases in terms of opposite alterations in reproductive physiological processes but also contrasting phenotypic traits. However, to fully explain the phenotypic features observed in women with these conditions, we need to establish a potential nexus system between the reproductive system and general biological functions. The recent discovery of kisspeptin as pivotal mediator of internal and external inputs on the hypothalamic-pituitary-gonadal axis has led to a new understanding of the neuroendocrine upstream regulation of the human reproductive system. In this review, we summarize the current knowledge on the physiological roles of kisspeptin in human reproduction, as well as its involvement in complex biological functions such as metabolism, inflammation and pain sensitivity. Importantly, these functions are known to be dysregulated in both PCOS and endometriosis. Within the evolving scientific field of "kisspeptinology", we critically discuss the clinical relevance of these discoveries and their potential translational applications in endometriosis and PCOS. By exploring the possibilities of manipulating this complex signaling system, we aim to pave the way for novel targeted therapies in these reproductive diseases.
Collapse
Affiliation(s)
- Noemi Salmeri
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Paola Viganò
- Infertility Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via M. Fanti 6, 20122, Milan, Italy.
| | - Paolo Cavoretto
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Roberto Marci
- Gynecology & Obstetrics, University of Ferrara, 44121, Ferrara, Italy
| | - Massimo Candiani
- Gynecology and Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| |
Collapse
|
48
|
Jin Y, Eum DY, Lee C, Park SY, Shim JW, Choi YJ, Choi SH, Kim JG, Heo K, Park SJ. Breast cancer malignancy is governed by regulation of the macroH2A2/TM4SF1 axis, the AKT/NF-κB pathway, and elevated MMP13 expression. Mol Carcinog 2024; 63:714-727. [PMID: 38251858 DOI: 10.1002/mc.23683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/23/2024]
Abstract
The histone variant, macroH2A (mH2A) influences gene expression through epigenetic regulation. Tumor suppressive function of mH2A isoforms has been reported in various cancer types, but few studies have investigated the functional role of mH2A2 in breast cancer pathophysiology. This study aimed to determine the significance of mH2A2 in breast cancer development and progression by exploring its downstream regulatory mechanisms. Knockdown of mH2A2 facilitated the migration and invasion of breast cancer cells, whereas its overexpression exhibited the opposite effect. In vivo experiments revealed that augmenting mH2A2 expression reduced tumor growth and lung metastasis. Microarray analysis showed that TM4SF1 emerged as a likely target linked to mH2A2 owing to its significant suppression in breast cancer cell lines where mH2A2 was overexpressed among the genes that exhibited over twofold upregulation upon mH2A2 knockdown. Suppressing TM4SF1 reduced the migration, invasion, tumor growth, and metastasis of breast cancer cells in vitro and in vivo. TM4SF1 depletion reversed the increased aggressiveness triggered by mH2A2 knockdown, suggesting a close interplay between mH2A2 and TM4SF1. Our findings also highlight the role of the mH2A2/TM4SF1 axis in activating the AKT/NF-κB pathway. Consequently, activated NF-κB signaling leads to increased expression and secretion of MMP13, a potent promoter of metastasis. In summary, we propose that the orchestrated regulation of the mH2A2/TM4SF1 axis in conjunction with the AKT/NF-κB pathway and the subsequent elevation in MMP13 expression constitute pivotal factors governing the malignancy of breast cancer.
Collapse
Affiliation(s)
- Yunho Jin
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Da-Young Eum
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Chaeyoung Lee
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Soon Yong Park
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Jae Woong Shim
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Yoo Jin Choi
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Si Ho Choi
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Joong-Gook Kim
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Kyu Heo
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| | - Seong-Joon Park
- Research Center, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Busan, Republic of Korea
| |
Collapse
|
49
|
Su H, Peng C, Liu Y. Regulation of ferroptosis by PI3K/Akt signaling pathway: a promising therapeutic axis in cancer. Front Cell Dev Biol 2024; 12:1372330. [PMID: 38562143 PMCID: PMC10982379 DOI: 10.3389/fcell.2024.1372330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
The global challenge posed by cancer, marked by rising incidence and mortality rates, underscores the urgency for innovative therapeutic approaches. The PI3K/Akt signaling pathway, frequently amplified in various cancers, is central in regulating essential cellular processes. Its dysregulation, often stemming from genetic mutations, significantly contributes to cancer initiation, progression, and resistance to therapy. Concurrently, ferroptosis, a recently discovered form of regulated cell death characterized by iron-dependent processes and lipid reactive oxygen species buildup, holds implications for diseases, including cancer. Exploring the interplay between the dysregulated PI3K/Akt pathway and ferroptosis unveils potential insights into the molecular mechanisms driving or inhibiting ferroptotic processes in cancer cells. Evidence suggests that inhibiting the PI3K/Akt pathway may sensitize cancer cells to ferroptosis induction, offering a promising strategy to overcome drug resistance. This review aims to provide a comprehensive exploration of this interplay, shedding light on the potential for disrupting the PI3K/Akt pathway to enhance ferroptosis as an alternative route for inducing cell death and improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Hua Su
- Xingyi People’s Hospital, Xinyi, China
| | - Chao Peng
- Xingyi People’s Hospital, Xinyi, China
| | - Yang Liu
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
50
|
Chen X, Keller SJ, Hafner P, Alrawashdeh AY, Avery TY, Norona J, Zhou J, Ruess DA. Tyrosine phosphatase PTPN11/SHP2 in solid tumors - bull's eye for targeted therapy? Front Immunol 2024; 15:1340726. [PMID: 38504984 PMCID: PMC10948527 DOI: 10.3389/fimmu.2024.1340726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/19/2024] [Indexed: 03/21/2024] Open
Abstract
Encoded by PTPN11, the Src-homology 2 domain-containing phosphatase 2 (SHP2) integrates signals from various membrane-bound receptors such as receptor tyrosine kinases (RTKs), cytokine and integrin receptors and thereby promotes cell survival and proliferation. Activating mutations in the PTPN11 gene may trigger signaling pathways leading to the development of hematological malignancies, but are rarely found in solid tumors. Yet, aberrant SHP2 expression or activation has implications in the development, progression and metastasis of many solid tumor entities. SHP2 is involved in multiple signaling cascades, including the RAS-RAF-MEK-ERK-, PI3K-AKT-, JAK-STAT- and PD-L1/PD-1- pathways. Although not mutated, activation or functional requirement of SHP2 appears to play a relevant and context-dependent dichotomous role. This mostly tumor-promoting and infrequently tumor-suppressive role exists in many cancers such as gastrointestinal tumors, pancreatic, liver and lung cancer, gynecological entities, head and neck cancers, prostate cancer, glioblastoma and melanoma. Recent studies have identified SHP2 as a potential biomarker for the prognosis of some solid tumors. Based on promising preclinical work and the advent of orally available allosteric SHP2-inhibitors early clinical trials are currently investigating SHP2-directed approaches in various solid tumors, either as a single agent or in combination regimes. We here provide a brief overview of the molecular functions of SHP2 and collate current knowledge with regard to the significance of SHP2 expression and function in different solid tumor entities, including cells in their microenvironment, immune escape and therapy resistance. In the context of the present landscape of clinical trials with allosteric SHP2-inhibitors we discuss the multitude of opportunities but also limitations of a strategy targeting this non-receptor protein tyrosine phosphatase for treatment of solid tumors.
Collapse
Affiliation(s)
- Xun Chen
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Steffen Johannes Keller
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Philipp Hafner
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Asma Y. Alrawashdeh
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Thomas Yul Avery
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
| | - Johana Norona
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Dietrich Alexander Ruess
- Department of General and Visceral Surgery, Center for Surgery, Medical Center University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|