1
|
Rani P, Koulmane Laxminarayana SL, Swaminathan SM, Nagaraju SP, Bhojaraja MV, Shetty S, Kanakalakshmi ST. TGF-β: elusive target in diabetic kidney disease. Ren Fail 2025; 47:2483990. [PMID: 40180324 PMCID: PMC11980245 DOI: 10.1080/0886022x.2025.2483990] [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: 08/21/2024] [Revised: 02/17/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
Transforming growth factor-beta (TGF-β), a cytokine with near omnipresence, is an integral part of many vital cellular processes across the human body. The family includes three isoforms: Transforming growth factor-beta 1, 2, and 3. These cytokines play a significant role in the fibrosis cascade. Diabetic kidney disease (DKD), a major complication of diabetes, is increasing in prevalence daily, and the classical diagnosis of diabetes is based on the presence of albuminuria. The occurrence of nonalbuminuric DKD has provided new insight into the pathogenesis of this disease. The emphasis on multifactorial pathways involved in developing DKD has highlighted some markers associated with tissue fibrosis. In diabetic nephropathy, TGF-β is significantly involved in its pathology. Its presence in serum and urine means that it could be a diagnostic tool while its regulation provides potential therapeutic targets. Completely blocking TGF-β signaling could reach untargeted regions and cause unanticipated effects. This paper reviews the basic details of TGF-β as a cytokine, its role in DKD, and updates on research carried out to validate its candidacy.
Collapse
Affiliation(s)
- Priya Rani
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | | | - Shilna Muttickal Swaminathan
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Shankar Prasad Nagaraju
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | | | - Sahana Shetty
- Department of Endocrinology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | | |
Collapse
|
2
|
Zhou S, Jiang D, Liu Y, Wang Q, Hu M, Dai K, Chen L, Zhang T, Cai C, Wang J. The role of Sine Oculis Homeobox Homolog 2 in colon Cancer: Insights into prognosis, immune regulation, and therapeutic implications. Biochem Biophys Res Commun 2025; 772:152038. [PMID: 40414014 DOI: 10.1016/j.bbrc.2025.152038] [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: 02/22/2025] [Revised: 04/19/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Colon cancer (CC) remains a significant global health burden, and the search for novel prognostic biomarkers and therapeutic targets is crucial. This study comprehensively analyzed the role of SIX2 (Sine Oculis Homeobox Homolog 2) in CC. Utilizing data from TCGA, GTEx, and CCLE databases, differential expression of SIX2 was observed in multiple cancers, with significant upregulation in many tumors compared to normal tissues. In CC, SIX2's differential expression was notable. Cox regression analysis revealed its prognostic significance, with overexpression associated with poor survival outcomes. SIX2 was strongly associated with gene alterations and correlated with key signaling pathways like WNT and TGF-β. In the tumor microenvironment, SIX2 was related to immune cell infiltration and immune-related molecules. Notably, in CC, it was associated with immunosuppressive cells and checkpoint molecules. Additionally, ABT737 was found to sensitize tumor immunotherapy in the context of SIX2. Animal experiments demonstrated that ABT737 effectively restricted the growth of CC in mice, and its combination with antiPD-1 immunotherapy was more effective. It could reduce the infiltration of CD163+ tumor-associated macrophages but without significantly increasing the infiltration of CD8+ T cells. Our findings suggest that SIX2 is a potential key player in CC, offering insights into future research and the development of targeted therapies.
Collapse
Affiliation(s)
- Shicheng Zhou
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Dan Jiang
- Department of Surgery, Wucheng District People's Hospital, Jinhua, Zhejiang, 321000, China
| | - Yu Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qin Wang
- Department of Endocrinology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Manyi Hu
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Kangfu Dai
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Lin Chen
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, China
| | - Tianming Zhang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Cheng Cai
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China.
| | - Jianping Wang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China.
| |
Collapse
|
3
|
Jin P, Cai J, Chen N, Liu Y, Zhao H, Wang Y, Chen J, Li M, Xiao T, Shan C, Yu M, Zhang JV. TGF-β/snail-mediated epithelial-to-mesenchymal transition disrupts estradiol metabolism through suppressing the HSD17B2 expression in endometriotic epithelial cells. Biochem Biophys Res Commun 2025; 771:151964. [PMID: 40393157 DOI: 10.1016/j.bbrc.2025.151964] [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: 01/15/2025] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/22/2025]
Abstract
Endometriosis affects nearly 10 % of reproductive-age women and is characterized by the growth of endometrial-like tissues outside the uterus. This disease poses significant diagnostic and therapeutic challenges due to its unknown origins and complex pathophysiology. Our study investigates how epithelial-mesenchymal transition (EMT) contributes to the dysregulation of estradiol metabolism by suppressing hydroxysteroid 17β dehydrogenase 2 (HSD17B2) expression in endometriotic epithelial cells. We used Gene Set Variation Analysis (GSVA) on public microarray data to correlate EMT scores with HSD17B2 levels. This approach revealed a significant correlation, showing that EMT is linked to reduced HSD17B2 expression in endometriotic tissues. Furthermore, our qPCR and immunoblotting results showed that TGF-β-induced EMT significantly reduced HSD17B2 expression in human endometriotic 12Z epithelial cells. Additionally, our data showed that Snail, an EMT-related transcription factor, acts on the E-box motif in the HSD17B2 promoter to suppress transcription. Our findings show that EMT is associated with decreased HSD17B2 expression in endometriotic tissues. This downregulation disrupts estradiol metabolism, possibly contributing to endometriosis pathogenesis. Our study offers critical insights into the molecular mechanisms of endometriosis and suggests that targeting EMT, especially the TGF-β/Snail axis, could provide a new therapeutic approach.
Collapse
Affiliation(s)
- Ping Jin
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, 518055, China; The First School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, 518055, China
| | - Jinxuan Cai
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Na Chen
- Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, 518055, China
| | - Yang Liu
- Health Sciences Institute of China Medical University, Shenyang, Liaoning, 110000, China
| | - Hao Zhao
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China; College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Yichun Wang
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Jie Chen
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Mengxia Li
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Tianxia Xiao
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Chunhua Shan
- College of Life Science, Northeast Forestry University, Harbin, 150040, China.
| | - Ming Yu
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.
| | - Jian V Zhang
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China; Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong, 518000, China; Sino-European Center of Biomedicine and Health, Shenzhen, Guangdong, 518000, China.
| |
Collapse
|
4
|
Robby AI, Jiang S, Jin EJ, Park SY. Semiconducting polymer dot-based wireless electrochemical aptasensor for detection of aging-related TGF-β1 and IL-6. Anal Chim Acta 2025; 1360:344139. [PMID: 40409909 DOI: 10.1016/j.aca.2025.344139] [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: 01/31/2025] [Revised: 04/01/2025] [Accepted: 04/30/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND The senescence-associated secretory phenotype (SASP) is closely linked to aging by promoting inflammation and tissue degradation. Sensing SASP is crucial for early detection of and intervention in age-related diseases to enhance therapeutic outcomes. Herein, SASP-selective sensors (transforming growth factor [TGF]-β1 and interleukin [IL]-6 probes) were designed by utilizing TGF-β1/IL-6 aptamers-functionalized copper-immobilized polymer dots that promoted specific binding between TGF-β1/IL-6 aptamers on the probe surface with aging factors (TGF-β1 and IL-6). RESULTS The selective binding was reflected by changes in the conductivity of the probes. The TGF-β1 and IL-6 probes showed high sensitivity towards TGF-β1 and IL-6, with limits of detection of 193.09 pg/mL for the TGF-β1 (R2 = 0.9989) and 16.49 pg/mL (R2 = 0.9998) for IL-6 probes. In vitro study using senescent cells confirmed that the probes could selectively detect TGF-β1 and IL-6, indicated by increased resistance with longer incubation times (TGF-β172h = 2.775 MΩ, IL-672h = 2.401 MΩ). Furthermore, the TGF-β1 and IL-6 probes exhibited excellent detection performance in in vivo samples from aging mouse models when monitoring the levels of TGF-β1 and IL-6 at different times after lenti soup injection and at different mouse ages (6-20 months). Additionally, the electrical signals generated during sensing can be displayed on a smartphone via a wireless sensing system. SIGNIFICANCE TGF-β1 and IL-6 probes provide a sensitive, specific and accessible diagnostic platform for senescence aging factors monitoring, which are expected to be an essential tool that transforms the analysis of aging and age-related diseases.
Collapse
Affiliation(s)
- Akhmad Irhas Robby
- Chemical Industry Institute, Korea National University of Transportation, Chungju, 27469, Republic of Korea; Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Songling Jiang
- Integrated Omics Institute, Wonkwang University, Iksan, Chungbuk, 54538, Republic of Korea
| | - Eun-Jung Jin
- Integrated Omics Institute, Wonkwang University, Iksan, Chungbuk, 54538, Republic of Korea; Department of Biomedical Materials Science, Graduate School of JABA, Wonkwang University, Iksan, Jeonbuk State, 54538, Republic of Korea.
| | - Sung Young Park
- Chemical Industry Institute, Korea National University of Transportation, Chungju, 27469, Republic of Korea; Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea; Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, 27469, Republic of Korea.
| |
Collapse
|
5
|
Shin JW, Jang DH, Kim SY, Lee JJ, Gil TH, Shim E, Kim JY, Kim HS, Conboy MJ, Conboy IM, Wiley CD, Shin JS, Jeon OH. Propagation of senescent phenotypes by extracellular HMGB1 is dependent on its redox state. Metabolism 2025; 168:156259. [PMID: 40189139 DOI: 10.1016/j.metabol.2025.156259] [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: 02/19/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND & PURPOSE Cellular senescence spreads systemically through blood circulation, but its mechanisms remain unclear. High mobility group box 1 (HMGB1), a multifunctional senescence-associated secretory phenotype (SASP) factor, exists in various redox states. Here, we investigate the role of redox-sensitive HMGB1 (ReHMGB1) in driving paracrine and systemic senescence. METHODS We applied the paracrine senescence cultured model to evaluate the effect of ReHMGB1 on cellular senescence. Each redox state of HMGB1 was treated extracellularly to assess systemic senescence both in vitro and in vivo. Senescence was determined by SA-β-gal & EdU staining, p16INK4a and p21 expression, RT-qPCR, and Western blot methods. Bulk RNA sequencing was performed to investigate ReHMGB1-driven transcriptional changes and underlying pathways. Cytokine arrays characterized SASP profiles from ReHMGB1-treated cells. In vivo, young mice were administered ReHMGB1 systemically to induce senescence across multiple tissues. A muscle injury model in middle-aged mice was used to assess the therapeutic efficacy of HMGB1 blockade. RESULTS Extracellular ReHMGB1, but not its oxidized form, robustly induced senescence-like phenotypes across multiple cell types and tissues. Transcriptomic analysis revealed activation of RAGE-mediated JAK/STAT and NF-κB pathways, driving SASP expression and cell cycle arrest. Cytokine profiling confirmed paracrine senescence features induced by ReHMGB1. ReHMGB1 administration elevated senescence markers in vivo, while HMGB1 inhibition reduced senescence, attenuated systemic inflammation, and enhanced muscle regeneration. CONCLUSION ReHMGB1 is a redox-dependent pro-geronic factor driving systemic senescence. Targeting extracellular HMGB1 may offer therapeutic potential for preventing aging-related pathologies.
Collapse
Affiliation(s)
- Ji-Won Shin
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Dong-Hyun Jang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - So Young Kim
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Je-Jung Lee
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Tae-Hwan Gil
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Eunha Shim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Ji Yeon Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hyeon Soo Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA 94720, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA 94720, USA
| | - Christopher D Wiley
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ok Hee Jeon
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea.
| |
Collapse
|
6
|
Lin TE, Hsu KC, Chou CH, Tsai EY, Wu YW, Sung TY, Hsu JY, Hsieh JH, Yen SC, Chang YW, Pan SL, Huang WJ, Yang CR. Identification of pyrazole scaffold inhibitors targeting cyclin-dependent kinase 8 for potential use in pulmonary fibrosis. Biochem Pharmacol 2025; 237:116959. [PMID: 40280247 DOI: 10.1016/j.bcp.2025.116959] [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: 01/13/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a disease that includes inflammation and scarring of the lung tissues. Cyclin-dependent kinase 8 (CDK8) is a target of interest due to its role in inflammatory pathways. CDK8 can also modulate the TGF-β/Smad signaling associated with IPF. Herein, a structure-based virtual screening (SBVS) campaign led to the identification of three CDK8 inhibitors. Testing of candidate inhibitors in protein and cellular assays confirmed CDK8 inhibition, with the most potent inhibitor producing an IC50 value of 398.8 nM. Computational analysis identified pharmacological interactions that lead to CDK8 inhibition. No significant cytotoxicity was observed when the inhibitor was treated in vitro. Further results showed that the inhibitor can disrupt proteins associated with the epithelial-mesenchymal transition (EMT) and reduce cell migration. Additionally, the inhibitor can disrupt the TGF- β1/Smad signaling axis in the nucleus, potentially impacting the transcription of IPF related protein expression, when treated in cells at 5 µM. Comparisons to structures of known CDK8 inhibitors showed the identified inhibitor to be structurally novel. When tested against a panel of kinases at 1 µM, the most potent inhibitor demonstrated a favorable CDK8 selectivity profile. The identification of the CDK8 inhibitors in this study can be used in future drug design studies and as CDK8 probes to explore alternative therapeutics for IPF.
Collapse
Affiliation(s)
- Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ching-Hsuan Chou
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - En-Yun Tsai
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Wen Wu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Ying Sung
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jui-Yi Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jui-Hua Hsieh
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, USA
| | - Shih-Chung Yen
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong, China
| | - Yu-Wei Chang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung Medical Center, Keelung, Taiwan
| | - Shiow-Lin Pan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Wei-Jan Huang
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Chia-Ron Yang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
7
|
Wang Z, Cheng S, Wei J, Hu J, Li F, Yang W. Evolving role of deubiquitinating enzymes in oral cancer (Review). Oncol Lett 2025; 30:354. [PMID: 40438866 PMCID: PMC12117358 DOI: 10.3892/ol.2025.15100] [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: 12/03/2024] [Accepted: 04/25/2025] [Indexed: 06/01/2025] Open
Abstract
Oral cancer affects the mucosal epithelium located within the oral cavity. The prevalence of oral cancer is projected to increase by ~40% by 2040, leading to a subsequent rise in mortality rates. Oral carcinogenesis is complex and multifactorial and numerous signaling pathways are involved in disease development. Deubiquitination is commonly involved in the post-translational process of proteins, and serves a key role in tumorigenesis and cancer development. The present review aims to discuss the function of deubiquitinating enzymes (DUBs) in oral cancer, with a particular focus on oral squamous cell carcinoma (OSCC). The present review also aims to investigate the functional mechanisms, tumorigenic regulation and therapeutic targets of DUBs in OSCC, which may potentially provide a novel theoretical basis for the utilization of DUBs as molecular targets in the treatment of OSCC in the future.
Collapse
Affiliation(s)
- Zidi Wang
- Department of Dentistry, Tianjin Beichen Hospital, Tianjin 300400, P.R. China
| | - Siyuan Cheng
- Department of Dentistry, Tianjin Beichen Hospital, Tianjin 300400, P.R. China
| | - Jianhui Wei
- Department of Oncology, Tianjin Beichen Hospital, Tianjin 300400, P.R. China
| | - Jiandong Hu
- Core Laboratory, Tianjin Beichen Hospital, Tianjin 300400, P.R. China
| | - Fenge Li
- Department of Oncology, Tianjin Beichen Hospital, Tianjin 300400, P.R. China
- Core Laboratory, Tianjin Beichen Hospital, Tianjin 300400, P.R. China
- Cancer Diagnosis and Treatment Center, Tianjin Union Medical Cancer (The First Affiliated Hospital of Nankai University), Tianjin 300121, China
| | - Wenhua Yang
- Department of Dentistry, Tianjin Beichen Hospital, Tianjin 300400, P.R. China
| |
Collapse
|
8
|
Liu W, Chi X, Wu MP, Zhai Y, Jia R. Induction of immune priming against white spot syndrome in Procambarus clarkii through oral administration of transgenic Synechococcus sp. PCC7942: Insights from transcriptome analysis. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110352. [PMID: 40268071 DOI: 10.1016/j.fsi.2025.110352] [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: 01/18/2025] [Revised: 03/24/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
White Spot Syndrome Virus (WSSV) poses a significant threat to aquaculture, particularly affecting the red swamp crayfish (Procambarus clarkii). This study explores the efficacy of oral administration of transgenic Synechococcus sp. PCC7942, engineered to express WSSV envelop protein VP19 and VP (19 + 28), in inducing immune priming in red swamp crayfish. Our results demonstrate that the transgenic cyanobacteria significantly enhance the immune responses of crayfish, as evidenced by the upregulation of immune-related genes and increased survival rates post-WSSV challenge. Furthermore, the immune-stimulating activity of these proteins is maintained even after fragmentation into polypeptides during digestion. These findings highlight the potential of using genetically modified algae as a sustainable and effective strategy for disease management in aquaculture. Additionally, the molecular mechanism of immune priming effect of crayfish was explained, which provided theoretical support for long-term protection of aquatic economic species against virus.
Collapse
Affiliation(s)
- Wei Liu
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai, 201306, China; School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Xiaoping Chi
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Max Peiyuan Wu
- Shanghai High School International Division, Shanghai, 200231, China
| | - Yufeng Zhai
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Rui Jia
- College of Oceanography and Ecological Science, Shanghai Ocean University, Shanghai, 201306, China; Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, 201306, China.
| |
Collapse
|
9
|
Pandey S, Anshu T, Maharana KC, Sinha S. Molecular insights into diabetic wound healing: Focus on Wnt/β-catenin and MAPK/ERK signaling pathways. Cytokine 2025; 191:156957. [PMID: 40367830 DOI: 10.1016/j.cyto.2025.156957] [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: 12/23/2024] [Revised: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
Diabetic wounds manifest significant clinical challenge with approximately 50-70 % reporting non-traumatic lower limb amputations annually. This review examines the intricate relationship between impaired wound healing in diabetes mellitus and two crucial signaling pathways: Wnt/β-catenin and MAPK/ERK. Chronic hyperglycemia in diabetes mellitus leads to peripheral neuropathy, vascular dysfunction, and compromised immune responses, resulting in delayed wound healing. The Wnt/β-catenin pathway, which is essential for cellular proliferation, differentiation, and tissue homeostasis, shows altered activity in diabetic wounds, particularly through decreased R-spondin 3 protein expression. Similarly, the MAPK/ERK pathway, which regulates cellular proliferation and differentiation through hierarchical kinase cascades, exhibits dysregulation under diabetic conditions. This review describes the current understanding of normal wound healing processes, diabetic wound pathophysiology, and the molecular mechanisms of both signaling pathways. Evidence suggests that targeting these pathways, either individually or synergistically offer promising therapeutic approaches for diabetic wound management. Future directions include, developing targeted delivery systems, exploring pathway cross-talk, and investigating dual-pathway modulators to enhance wound healing outcomes in diabetic patients. This comprehensive analysis provides insights into potential therapeutic strategies and emphasizes the necessity of research in this crucial area of diabetes treatment. (Graphical Abstract).
Collapse
Affiliation(s)
- Shricharan Pandey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| | - Tushar Anshu
- Department of Pharmaceutical sciences and drug research, Punjabi University Patiala, India
| | - Krushna Ch Maharana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| | - Suhani Sinha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India.
| |
Collapse
|
10
|
Yu Y, Yu T, Liu K, Li Y, Luan Y, Yang T, Li W, Cong H, Wu X. Perimenopausal depression: Targeting inflammation and oxidative stress (Review). Mol Med Rep 2025; 31:161. [PMID: 40211702 PMCID: PMC12015406 DOI: 10.3892/mmr.2025.13526] [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: 01/15/2025] [Accepted: 03/20/2025] [Indexed: 04/25/2025] Open
Abstract
Depressive disorder is a highly disabling condition that affects more than 300 million individuals worldwide, with women affected at a higher rate than men. With the aging of the population, the incidence of perimenopausal depression has risen markedly, seriously jeopardizing women's physical and mental health. Symptoms of perimenopausal depression include feelings of depression, stress, anxiety and endocrine dysfunctions, particularly hypogonadism and senescence. During perimenopause, estrogen and progesterone levels fluctuate erratically, adding to the risk of developing depression associated with perimenopause. As a result of these hormonal changes, proinflammatory mediators are produced and oxidative stress is induced, which finally leads to progressive neuronal damage. The present study mainly reviewed roles of neuroinflammation in perimenopausal depression and explained potential anti‑inflammatory and anti‑oxidative stress mechanisms for clinically effective therapeutic treatment.
Collapse
Affiliation(s)
- Yang Yu
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- Department of Gynecology, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Tianyang Yu
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- Department of Acupuncture, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Kaili Liu
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Yushuai Li
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Yifeng Luan
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- Department of Gynecology, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Tianyi Yang
- Department of Respiratory Medicine, Heilongjiang Academy of Sciences of Traditional Chinese Medicine, Harbin, Heilongjiang 150036, P.R. China
| | - Wenzhong Li
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Huifang Cong
- Department of Second Clinical Medical School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
- Department of Gynecology, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150001, P.R. China
| | - Xiuhong Wu
- Department of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
11
|
Sivalingam AM, Sureshkumar DD. Exosomes in Regulating miRNAs for Biomarkers of Neurodegenerative Disorders. Mol Neurobiol 2025; 62:7576-7596. [PMID: 39918711 DOI: 10.1007/s12035-025-04733-8] [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: 09/26/2024] [Accepted: 01/29/2025] [Indexed: 05/15/2025]
Abstract
Exosomal proteins and miRNAs, including α-synuclein, Aβ, tau, CXCL12, miR-24, and miR-23b-3p, are emerging as valuable biomarkers for Parkinson's disease and prenatal diagnostics, with significant potential for personalized therapies. Advances in MRI and chitosan-based drug delivery systems are creating new opportunities for diagnosing and treating neurodegenerative disorders. Exosomes regulate miRNAs and proteins, presenting theranostic potential for Alzheimer's and Huntington's diseases, yet facing delivery and targeting challenges. Exosomal miRNAs, such as miR-1234, miR-5678, and miR-29a, are crucial for the early detection and monitoring of the progression of neurodegenerative diseases. Additionally, novel biomarkers such as SCA27B and FGF14 gene mutations and serum miR-455-3p offer promising noninvasive diagnostic methods for Alzheimer's disease. The expanding role of exosome-derived miRNAs in targeting oncogenes and regulating the cell cycle enhances therapeutic strategies for neurological disorders, opening doors to more personalized and effective disease management.
Collapse
Affiliation(s)
- Azhagu Madhavan Sivalingam
- Natural Products & Nanobiotechnology Research Lab, Department of Community Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Thandalam, Chennai, 602 105, Tamil Nadu, India.
| | - Darshitha D Sureshkumar
- Department of Forensic Science, NIMS Institute of Allied Medical Science and Technology, (NIMS University), Jaipur, 303121, Rajasthan, India
| |
Collapse
|
12
|
Manich G, Pérez B, Penas C, Dantas AP, Coutinho J, Sánchez-Bernadó P, García-Aranda J, Fraile-Ramos J, Benseny-Cases N, Martín-Mur B, Esteve-Codina A, Rodríguez-Rovira I, Giménez-Llort L, Egea G, Jiménez-Altayó F. Sex- and age-dependent neurovascular abnormalities linked to neuroinflammation lead to exacerbated post-ischemic brain injury in Marfan syndrome mice. Redox Biol 2025; 83:103662. [PMID: 40349485 DOI: 10.1016/j.redox.2025.103662] [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: 03/26/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/14/2025] Open
Abstract
Fibrillin 1 gene (Fbn1) mutations cause Marfan syndrome (MFS), triggering life-threatening aortic complications and multi-organ effects. MFS is increasingly linked to neurovascular complications, amplified by aortic surgery risks. However, the impact of MFS on the brain remains unclear, including the roles of sex, aging, and their contribution to cerebral injury. This study examines brain alterations and their role in cerebral ischemic injury in an MFS mouse model. RNA-seq analysis of young (3-month-old) and aged (13-month-old) male and female wild-type and MFS (Fbn1C1041G/+) mice revealed disruptions in TGF-β and extracellular matrix (ECM) pathways in MFS brains, most pronounced in young males and aged females with reduced estrogen levels. Inflammatory pathways were upregulated across all MFS mice. Consequently, changes in TGF-β signaling, ECM turnover, redox stress and inflammatory pathways were assessed through RT-qPCR, immunostaining, Western blot, lucigenin chemiluminescence, spectrophotometry, HPLC, and synchrotron radiation-based microspectroscopy, while cerebrovascular properties were assessed by pressure myography and confocal microscopy in the basilar artery. Aged MFS mice showed decreased brain TGF-β1 levels, while dysregulated collagen turnover was only observed in female MFS mice. Despite increased NADPH oxidase activity and redox damage in the corpus callosum of male MFS mice, brain redox stress levels remain largely unchanged. Young female MFS mice exhibited hypertrophic remodeling of the basilar artery. Remarkably, neuroinflammation driven by reactive gliosis increased in MFS mice, regardless of sex and age. To determine the impact on ischemic vulnerability, young mice underwent bilateral common carotid artery occlusion (5 min)/reperfusion (3 days). MFS mice showed greater post-ischemic brain damage, evidenced by worsened behavioral impairments, hippocampal neurodegeneration, and neuroinflammation. This study identifies sex- and age-dependent disruptions in TGF-β1, ECM, and cerebrovascular integrity in MFS mice. Persistent neuroinflammation and increased vulnerability to post-ischemic brain injury suggests that MFS patients, alongside well-documented aortic complications, have an intrinsic predisposition to cerebral damage.
Collapse
Affiliation(s)
- Gemma Manich
- Department of Morphological Sciences, School of Medicine, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain
| | - Belén Pérez
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Department of Pharmacology, Therapeutics, and Toxicology, School of Medicine, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain
| | - Clara Penas
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Cerdanyola Del Vallès, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Red Española de Terapias Avanzadas (RED-TERAV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Paula Dantas
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, Institut D'Investigacions Biomediques August Pi I Sunyer, Hospital Clinic Cardiovascular Institute, Barcelona, Spain
| | - Joana Coutinho
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Department of Pharmacology, Therapeutics, and Toxicology, School of Medicine, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain
| | - Paula Sánchez-Bernadó
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Department of Pharmacology, Therapeutics, and Toxicology, School of Medicine, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain
| | - Julián García-Aranda
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Department of Pharmacology, Therapeutics, and Toxicology, School of Medicine, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Juan Fraile-Ramos
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Núria Benseny-Cases
- Unitat de Biofísica. Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Beatriz Martín-Mur
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain; Universitat de Barcelona, Barcelona, Spain
| | - Anna Esteve-Codina
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain; Universitat de Barcelona, Barcelona, Spain
| | - Isaac Rodríguez-Rovira
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona-IDIBAPS, Barcelona, Spain
| | - Lydia Giménez-Llort
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Department of Psychiatry and Forensic Medicine, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gustavo Egea
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona-IDIBAPS, Barcelona, Spain; Department of Medical Genetics, University of Antwerpen, Antwerpen, Belgium
| | - Francesc Jiménez-Altayó
- Institute of Neurosciences, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Department of Pharmacology, Therapeutics, and Toxicology, School of Medicine, Universitat Autònoma de Barcelona, Cerdanyola Del Vallès, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
13
|
Tran HH, Yamaguchi A, Manning HC. Radiotheranostic landscape: A review of clinical and preclinical development. Eur J Nucl Med Mol Imaging 2025; 52:2685-2709. [PMID: 39891713 DOI: 10.1007/s00259-025-07103-7] [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: 11/04/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Radiotheranostics combines diagnostic imaging with targeted radionuclide therapy, representing a transformative approach in precision oncology. Landmark approvals of Lutathera® and Pluvicto® have catalyzed significant advancements in this field, driving research into novel radionuclides, targeting strategies, and clinical applications. This review evaluates the evolving clinical and preclinical landscape of radiotheranostics, highlighting advancements, emerging trends, and persistent challenges in radionuclide therapy. METHODS A comprehensive analysis was performed, encompassing active clinical trials as of December 2024, sourced from ClinicalTrials.gov and TheranosticTrials.org. Preclinical developments were evaluated through a review of recent literature, focusing on innovations in radionuclide production, targeting molecules, and radiochemistry. RESULTS In reviewing the clinical landscape, agents targeting somatostatin receptors (SSTR) and prostate-specific membrane antigen (PSMA) still dominate the field, but new targets such as fibroblast activation protein (FAP), integrins, and gastrin-releasing peptide receptors (GRPR) are gaining traction in both clinical and preclinical development. While small molecules and peptides remain the most common radionuclide carriers, antibody-based carriers including bispecific antibodies, immunoglobin-derived antigen-binding fragments, and antibody-mimetic proteins are on the rise due to their specificity and adaptability. Innovations in radioligand design are driving a shift from agonists to antagonists, accompanied by the development of modified peptides with enhanced pharmacokinetics and tumor-targeting properties. Next-generation therapeutic radionuclides, such as the beta-emitter terbium-161 and alpha-emitters actinium-225 and lead-212, are under investigation to complement or replace lutetium-177, addressing the need for improved efficacy and reduced toxicity. Paired isotopic radionuclides are gaining popularity for their ability to optimize imaging and therapeutic dosimetry as they offer near-identical specificity, biodistribution, and metabolism. Additionally, radiohybrid systems represent an innovative approach to chelating chemically distinct radionuclide pairs within a single molecule, further enhancing flexibility in radiotheranostic design. CONCLUSION Radiotheranostics has transformed cancer care through its precision and adaptability, but challenges in radionuclide production, regulatory frameworks, and workforce training hinder broader adoption. Advances in isotopic pairing, next-generation radionuclides, and radiohybrid systems in preclinical and clinical settings hold promise to overcome these barriers. Collaborative efforts among academia, industry, and regulatory bodies are critical to accelerating innovation and optimizing clinical outcomes.
Collapse
Affiliation(s)
- Ha H Tran
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aiko Yamaguchi
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - H Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
14
|
Hong Y, Feng Z, Ge Y, Xi Y, Zhang B, Wu J, Xia T, Tang B, Wang W, Chen J, Wang H, Xiao H. miR-145-enriched BMSCs-derived exosomes ameliorate neurogenic erectile dysfunction in aged rats via TGFBR2 inhibition. Regen Ther 2025; 29:455-465. [PMID: 40308644 PMCID: PMC12041780 DOI: 10.1016/j.reth.2025.04.004] [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: 02/14/2025] [Revised: 03/13/2025] [Accepted: 04/06/2025] [Indexed: 05/02/2025] Open
Abstract
Background Neurogenic erectile dysfunction (ED) is a prevalent complication following radical prostatectomy in elderly patients, primarily resulting from the apoptosis of corpus cavernosum smooth muscle cells (CCSMCs) and the subsequent excessive fibrosis of the corpus cavernosum. Aim This study aimed to compare the therapeutic effects of exosomes derived from lentivirus-transfected miR-145 bone marrow mesenchymal stem cells (Exo-145) and unmodified BMSCs-derived exosomes (Exo) in aged rats with bilateral cavernous nerve injury (BCNI) and investigate the underlying mechanisms. Methods Twenty-four-month-old male rats were assigned to four groups, namely Sham, BCNI, Exo, and Exo-145. Three weeks after treatment, erectile function was assessed by measuring the maximal intracavernosal pressure to mean arterial pressure (ICP/MAP) ratio. Apoptosis and fibrosis were semi-quantitatively analyzed using TUNEL and Masson's trichrome staining, respectively. In vitro, CCSMCs were subjected to H2O2-induced oxidative stress, and the protective effects of Exo-145 were evaluated through flow cytometry and Western blot. Lastly, the targets and mechanisms of miR-145 were further validated using dual-luciferase reporter assays and rescue experiments. Results Exo-145 significantly outperformed Exo in restoring erectile function in aged BCNI rats, as evidenced by the significantly higher maximal ICP/MAP ratio, a marked reduction in TUNEL-positive cell count, and marked suppression of fibrosis in cavernous tissue. Moreover, Masson's trichrome staining displayed a substantial decrease in collagen deposition. In vitro, Exo-145 alleviated H2O2-induced apoptosis in CCSMCs by downregulating Cleaved Caspase-3 expression and Bax while concurrently upregulating Bcl-2 expression. TGFBR2 was identified as a direct target of miR-145 through dual-luciferase reporter assays, with its overexpression partially reversing the protective effects of Exo-145. Conclusion Exo-145 demonstrates superior efficacy compared to Exo in treating aged neurogenic ED by targeting TGFBR2 to alleviate apoptosis and fibrosis. It may represent a promising cell-free therapeutic option for neurogenic erectile dysfunction in elderly patients and could offer new perspectives for improving their prognosis.
Collapse
Affiliation(s)
- Yude Hong
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Urology, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Zejia Feng
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yunlong Ge
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuhang Xi
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bowen Zhang
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jianjie Wu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Tian Xia
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bowen Tang
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Wang
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Chen
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hua Wang
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hengjun Xiao
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
15
|
Govindasamy N, Barman M, Salleh N, Giribabu N, Shahzad H. Effects of 17β estradiol on blood pressure elevation in ovariectomized rats with collagen-induced arthritis via modulation of oxidative stress, inflammation, fibrosis, and apoptosis in the aorta involving TLR4/NOX4/NF-kβ and TGFβ1/fibronectin/α-SMA pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6941-6962. [PMID: 39702602 DOI: 10.1007/s00210-024-03700-9] [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: 06/24/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Rheumatoid arthritis (RA) can cause blood pressure (BP) elevation in estrogen-deficient, post-menopausal women; however, the underlying mechanisms are not well understood. In this study, the aortic involvement and its underlying mechanisms that contribute to the BP elevation in estrogen-deficient, RA condition were identified. Ovariectomy was performed to create a state of estrogen deficiency and RA was then induced in ovariectomized rats by using incomplete Freund's adjuvant and immune-mediated collagen type-II. Ovariectomized, RA-induced rats (Ovx + RA) were given either 17β-estradiol, baricitinib, or losartan. Direct blood pressure (BP) monitoring was made via cannulation of the carotid artery. Rats were then sacrificed and the aorta was harvested followed by H&E and Picrosirius staining to evaluate histological changes and collagen deposition. Oxidative stress, inflammation, apoptosis, growth, and fibrosis levels in the aorta were assessed by using molecular biological techniques. Mean arterial pressure (MAP) was significantly elevated in Ovx + RA rats when compared to sham and Ovx rats (p < 0.05). 17β-estradiol and losartan treatment significantly reduced the MAP and heart rate in Ovx + RA rats when compared to untreated Ovx + RA rats. Expression of iNOS, Nox2 and Nox4, TLR4, NF-ĸB, TNF-α, VEGF, FGF-2, αSMA, eNOS, and caspase-3 were elevated in the aorta of Ovx + RA rats and were reduced upon 17β-estradiol treatment. However, expression of TGFβ1, Bax-2, fibronectin, and Smad2 in the aorta of Ovx + RA rats was increased following 17β-estradiol treatment (p < 0.05 compared to without treatment). The presence of RA with estrogen deficiency enhanced the BP elevation due to changes in the aorta which could be ameliorated by estrogen.
Collapse
Affiliation(s)
- Navishaa Govindasamy
- School of Postgraduate Studies (SOPG), International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Madhumanti Barman
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Lembah Pantai, 59100, Kuala Lumpur, Malaysia
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Lembah Pantai, 59100, Kuala Lumpur, Malaysia.
- Human Reproduction Research Group, Faculty of Medicine, Universiti Malaya, Lembah Pantai, 59100, Kuala Lumpur, Malaysia.
| | - Nelli Giribabu
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Lembah Pantai, 59100, Kuala Lumpur, Malaysia.
- Human Reproduction Research Group, Faculty of Medicine, Universiti Malaya, Lembah Pantai, 59100, Kuala Lumpur, Malaysia.
| | - Huma Shahzad
- Department of Human Biology to the Physiology, School of Medicine, International Medical University, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
16
|
Conlon T, Schaaf M, Mateos-Maroto A, Picciotto S, Morsbach S, Adamo G, Si S, Lieberwirth I, Rosenauer C, Landfester K, Bongiovanni A, Touzet N. Comparative effects of extracellular vesicles and liposomal nanocarriers on bleomycin-induced stress in A549 human adenocarcinoma cells. Biomed Pharmacother 2025; 187:118081. [PMID: 40273689 DOI: 10.1016/j.biopha.2025.118081] [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: 02/04/2025] [Revised: 03/27/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025] Open
Abstract
Lung cancer and chronic respiratory diseases are among the leading causes of death worldwide. Key factors in their pathogenesis include reactive oxygen species (ROS), transforming growth factor-β1 (TGF-β1) and epithelial-mesenchymal transition (EMT). Exogenous antioxidants can mitigate the oxidative stress that drives TGF-β1-mediated respiratory pathologies. Given their role in cellular communication and natural biocompatibility, extracellular vesicles (EVs) are emerging as promising candidates for the delivery of therapeutic cargo to pathological cells. Notably, microalgal-derived EVs (i.e., nanoalgosomes) have been shown to exhibit antioxidant and anti-inflammatory activity. In this study, the bioactivity of EVs derived from Tetraselmis chuii (CCAP 66/21B) was investigated in a bleomycin-stressed (8 µg mL-1) human adenocarcinoma alveolar epithelial cell model (A549). Moreover, the effects of these EVs were compared to liposomes loaded with established therapeutics (pirfenidone and quercetin), synthesised using the lipid film hydration method. In vitro assessments included cell viability (MTS), intracellular ROS, morphological changes, cell migration, EMT-related mRNA expression (qPCR), and TGF-β1 release (ELISA). Both the EVs (nanoalgosomes) and pirfenidone- and quercetin-loaded liposomal nanocarriers (1-4 µg mL-1) effectively attenuated bleomycin-induced EMT, inhibited cell migration, suppressed profibrotic TGF-β1, lowered intracellular ROS and upregulated glutathione peroxidase 4 (GPX4). Importantly, the innate bioactive cargo of the naturally derived nanoalgosomes exhibited comparable effects to the liposome therapeutic formulations in mitigating bleomycin-induced stress in A549 cells.
Collapse
Affiliation(s)
- Thomas Conlon
- Centre for Environmental Research Innovation and Sustainability (CERIS), Atlantic Technological University Sligo, Sligo, Ireland.
| | - Maximilian Schaaf
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Ana Mateos-Maroto
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Sabrina Picciotto
- Cell-Tech HUB at Institute of Biophysics (IBF) - National Research Council of Italy (CNR), Palermo 90146, Italy
| | - Svenja Morsbach
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Giorgia Adamo
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo 90146, Italy
| | - Shutian Si
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Ingo Lieberwirth
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Christine Rosenauer
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Antonella Bongiovanni
- Cell-Tech HUB at Institute for Research and Biomedical Innovation, National Research Council of Italy (CNR), Palermo 90146, Italy
| | - Nicolas Touzet
- Centre for Environmental Research Innovation and Sustainability (CERIS), Atlantic Technological University Sligo, Sligo, Ireland
| |
Collapse
|
17
|
Xu Y, Gu X, Li W, Lin B, Xu Y, Wei Q, Liu Q, Zhao Y, Long R, Jiang H, Wu Z, Liu Y, Qiang L. Autophagic degradation of SQSTM1 enables fibroblast activation to accelerate wound healing. Autophagy 2025:1-21. [PMID: 40400126 DOI: 10.1080/15548627.2025.2508546] [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: 09/03/2024] [Revised: 05/07/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025] Open
Abstract
Wound healing is a meticulously coordinated and intricate progression that necessitates precise regulation of fibroblast behavior. Macroautophagy/autophagy is a degradation system for clearing damaged cellular components. SQSTM1/p62 (sequestosome 1), a well-established autophagy receptor, also functions as a signaling hub beyond autophagy. Here, we observed a significant upregulation of autophagy in fibroblasts after wounding. Using mice with fibroblast-specific deletion of Atg7 (autophagy related 7), we found that fibroblast autophagy governed wound healing. Fibroblast autophagy deficiency delayed proper dermal repair that was mired in insufficient fibroblast proliferation, migration, and myofibroblast transition. In vitro experiments further revealed that autophagy deficiency disrupted TGFB1 (transforming growth factor beta 1)-induced fibroblast proliferation, migration, and myofibroblast differentiation. Mechanistically, autophagy deficiency led to SMAD2 (SMAD family member 2) and SMAD3 sequestration within SQSTM1 bodies and attenuated TGFB1-induced receptor-regulated SMAD (R-SMAD) phosphorylation in an SQSTM1-dependent manner. Furthermore, sqstm1 deletion rescued the delayed skin wound healing caused by autophagy deficiency, and autophagy inducers promoted wound healing in an SQSTM1-dependent manner. Our findings highlight the critical role of fibroblast autophagy in wound healing and elucidate the underlying mechanisms by which autophagy regulates fibroblast behavior.Abbreviation: 3-MA: 3-methyladenine; ACTA2/α-SMA: actin alpha 2, smooth muscle; ACTB: actin beta; AMPK: AMP-activated protein kinase; ATG: autophagy related; BiFC: bimolecular fluorescence complementation; COL1A2: collagen type I alpha 2 chain; ECM: extracellular matrix; FGF: fibroblast growth factor; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; HDF: human dermal fibroblast; HVGs: highly variable genes; KO: knockout; LMNB1: lamin B1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MKI67/Ki-67: marker of proliferation Ki-67; MTOR/mTOR: mechanistic target of rapamycin kinase; NFE2L2/NRF2: NFE2 like bZIP transcription factor 2; NFKB: nuclear factor kappa B; NLRP3: NLR family pyrin domain containing 3; PCA: principal component analysis; PI3K: phosphoinositide 3-kinase; R-SMAD: receptor-regulated SMAD; SBE: SMAD binding element; shCON: small hairpin negative control; siNC: negative control; siRNA: small interfering RNA; SMAD: SMAD family member; SQSTM1/p62: sequestosome 1; ssGSEA: single-sample gene set enrichment analysis; TGFB/TGF-β: transforming growth factor beta; TGFBR1: transforming growth factor beta receptor 1; TGFBR2: transforming growth factor beta receptor 2; VIM: vimentin; WT: wild-type; ZFYVE9/SARA: zinc finger FYVE-type containing 9.
Collapse
Affiliation(s)
- Yujiao Xu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xin Gu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Wenshu Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Boyang Lin
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yiting Xu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qingcheng Wei
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qingyuan Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yamin Zhao
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Rongzhuo Long
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Hulin Jiang
- State Key Laboratory of Natural Medicines, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Zhaoqiu Wu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yunyao Liu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu 210042, China
| | - Lei Qiang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| |
Collapse
|
18
|
Lee GG, Peterson AJ, Kim MJ, Shimell M, O’Connor MB, Park JH. Linking expression and function of Drosophila type-I TGF-β receptor baboon isoforms: Multiple roles of BaboA isoform in shaping of the adult central nervous system. PLoS One 2025; 20:e0318406. [PMID: 40445987 PMCID: PMC12124520 DOI: 10.1371/journal.pone.0318406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 05/05/2025] [Indexed: 06/02/2025] Open
Abstract
Evolutionarily conserved transforming growth factor β (TGF-β) signaling is used in both vertebrates and invertebrates to regulate a variety of developmental and cellular processes. The baboon (babo) gene encoding a Drosophila type-I TGF-β receptor produces three isoforms via alternative splicing: BaboA, BaboB, and BaboC. In this study, we generated three fly lines, each carrying an isoform-specific GFP tag, and another line with a GFP conjugated at the C-terminus common to all isoforms. Using these lines, we assessed (1) whether the tagged proteins function properly in rescue assays and (2) how the isoform expression is regulated in various tissues including the central nervous system (CNS). A Gal4 knock-in line in the babo locus was also characterized for reporter expression, mutant phenotypes, and isoform-specific knockdown phenotypes. We found that the C-terminal tag does not interrupt the subcellular targeting and functions of the tagged isoforms, but the internal isoform tags do so in a cell- and isoform-specific fashion. Nevertheless, our results demonstrated that these tags faithfully reflect endogenous expression of individual isoforms. Certain cell types express single or multiple isoforms at different levels, suggesting that alternative splicing could determine the isoform types and their levels depending on cell (or tissue) type. The larval CNS displays distinct patterns of two isoforms, BaboA and BaboC. BaboC is mostly expressed in neural cells originating during embryogenesis, while BaboA is broadly expressed in neural cells produced from both embryonic and postembryonic stages. Assays of both isoform-specific mutants and cell-specific knockdown of individual isoforms revealed broad roles played by BaboA in postembryonic neurogenesis and differentiation of precursor neurons, remodeling processes of persisting larval neurons, and metamorphic CNS reorganization, which are essential for establishing of the adult CNS. Taken together, this study demonstrates that the GFP-tagged lines permit visualization of endogenous expression of individual isoforms, which further provides clues about cell- and stage-specific functions played by each isoform.
Collapse
Affiliation(s)
- Gyunghee G. Lee
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Aidan J. Peterson
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - MaryJane Shimell
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Michael B. O’Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jae H. Park
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, United States of America
| |
Collapse
|
19
|
Cheng W, Zheng Y, Tang Q, Qi L, Shi Z, Yu Q, Li M, Wei X, Zhou Y, Jiang X. Discovery of Novel Cyclic Peptides as SMAD2-SMAD4 Interaction Inhibitors for the Treatment of Hepatic Fibrosis. J Med Chem 2025; 68:9958-9972. [PMID: 40320643 DOI: 10.1021/acs.jmedchem.4c02938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Hepatic fibrosis, characterized by the excessive deposition of the extracellular matrix, represents a common consequence of various chronic liver disorders. However, no specific drugs are available for antifibrotic therapy to date. SMAD2 is phosphorylated by transforming growth factor-β and subsequently binds to SMAD4 to generate a heteromeric complex, which then translocates into the nucleus and aggravates liver fibrosis. Herein, based on molecular docking simulation and structure-activity relationship study, we report the discovery of a novel cyclic peptide CMF9 that targets SMAD2 and potently interferes with the SMAD2-SMAD4 interaction. The subsequent in vivo and in vitro pharmacological studies demonstrated that CMF9 dramatically suppressed hepatic stellate cells activation and collagen synthesis, alleviating CCl4-induced hepatic inflammation and fibrosis. Overall, we first demonstrated that the novel cyclic peptide CMF9 could efficiently block the SMAD2-SMAD4 interaction via selectively inhibiting SMAD2 phosphorylation, providing a promising therapeutic strategy for targeting SMAD2 and an alternative candidate for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Wei Cheng
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yawen Zheng
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qinglin Tang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Liang Qi
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zihan Shi
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qihong Yu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Mingmin Li
- College of Life Science, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Xianzhi Wei
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yifeng Zhou
- College of Life Science, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Xianxing Jiang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
20
|
Jamal MH, Khan MN. Developments in pancreatic cancer emerging therapies, diagnostic methods, and epidemiology. Pathol Res Pract 2025; 271:156012. [PMID: 40414076 DOI: 10.1016/j.prp.2025.156012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/27/2025]
Abstract
Pancreatic cancer is still one of the deadliest malignancies, characterised by late-stage diagnosis, aggressive biology, and considerable resistance to conventional treatments. Despite improvements in understanding the molecular mechanisms and innovations in treatment, the overall survival remains abysmal: fewer than 9 % of patients survive beyond 5 years. By 2030, PC is predicted to become the second leading cause of cancer-related deaths in the U.S. owing to chemoresistance, rapid metastatic spread, and limited effective immunotherapeutic choices. This review highlights current progress in this field, including epidemiology, risk factors, diagnostic tools, and emerging biomarkers. Recent progress in genetic and molecular profiling has provided important information about pancreatic cancer. It has identified key mutations in genes like KRAS, TP53, CDKN2A, and SMAD4 that play a major role in driving the disease. Such revelations have provided the impetus to explore novel targeted therapies against these mutations. Furthermore, the advances in liquid biopsies incorporating circulating tumour cells, circulating tumour DNA, and exosomes hold substantial promise for early diagnosis, treatment response monitoring, and detection of minimal residual disease-any of which could radically transform PC management. While very limited options for the treatment of advanced-stage PC remain, the only potential curative treatment is surgery, yet only 10-15 % of patients are diagnosed with potentially resectable disease. Researchers are looking into new methods to help more patients qualify for surgery. This involves using chemotherapy and radiotherapy to reduce the size of the tumor before the operation. New chemotherapy treatments like FOLFIRINOX (which includes 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin) have improved results for some patients, but they can still cause significant side effects. Immunotherapy, though revolutionary in other cancers, has had limited success in PC due to the tumour's immunosuppressive microenvironment. Researchers are looking into using immune checkpoint inhibitors together with chemotherapy, radiation, and drugs that target the surrounding tissue to improve the body's immune response. There is also considerable excitement surrounding personalised approaches with adoptive cell therapies such as CAR-T cells and TILs, which are trialled with early evidence of potential efficacy. Attempts are also being made to address the dense desmoplastic stroma of the tumour that characterises PC. Drugs that can fight resistance or new medicines that might affect the tumor environment, stop changes in surrounding tissues, and improve how drugs are delivered have shown some potential in laboratory tests so far. Nanoparticle-based drug delivery systems are also being developed to improve the bioavailability and targeted delivery of chemotherapy.
Collapse
Affiliation(s)
- Mohd Haris Jamal
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, India
| | - Md Nasiruddin Khan
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
21
|
Jee YM, Lee JY, Ryu T. Chronic Inflammation and Immune Dysregulation in Metabolic-Dysfunction-Associated Steatotic Liver Disease Progression: From Steatosis to Hepatocellular Carcinoma. Biomedicines 2025; 13:1260. [PMID: 40427086 PMCID: PMC12109540 DOI: 10.3390/biomedicines13051260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Metabolic-dysfunction-associated steatotic liver disease (MASLD) progresses from hepatic steatosis to hepatocellular carcinoma (HCC) as a result of systemic immunometabolic dysfunction. This review summarizes the key roles of the innate and adaptive immune mechanisms driving hepatic injury, fibrogenesis, and carcinogenesis in MASLD. Methods: A comprehensive literature review was performed using PubMed to identify relevant published studies. Eligible articles included original research and clinical studies addressing immunological and metabolic mechanisms in MASLD, as well as emerging therapeutic strategies. Results: We highlight the roles of cytokine networks, the gut-liver axis, and immune cell reprogramming. Emerging therapeutic strategies, including cytokine inhibitors, anti-fibrotic agents, metabolic modulators, and nutraceuticals, offer several indications for attenuating MASLD progression and reducing the prevalence of extrahepatic manifestations. Conclusions: Given the heterogeneity of MASLD, personalized combination-based approaches targeting both inflammation and metabolic stress are essential for effective disease management and the prevention of systemic complications.
Collapse
Affiliation(s)
- Young-Min Jee
- Department of Family Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea;
- Department of Family Medicine, Graduate School of Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jeong-Yoon Lee
- Department of Neurology, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea;
- Department of Translational Medicine, Graduate School of Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Tom Ryu
- Department of Internal Medicine, Institute for Digestive Research, Digestive Disease Center, Soonchunhyang University College of Medicine, Seoul 04401, Republic of Korea
| |
Collapse
|
22
|
Zhang Q, Liu G, Jing L, Aghayants S, Xu F, Fan Y. The landscape of N 6-methyladenosine RNA methylation in skin diseases. Br J Dermatol 2025; 192:983-994. [PMID: 40059697 DOI: 10.1093/bjd/ljaf087] [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: 12/03/2024] [Revised: 02/15/2025] [Accepted: 03/05/2025] [Indexed: 05/20/2025]
Abstract
Skin diseases encompass a diverse range of conditions with significant psychological and physiological impacts. N6-methyladenosine (m6A) RNA methylation is a key epitranscriptomic modification that regulates gene expression by influencing RNA stability, splicing, translation, export and degradation. Recent studies have highlighted the crucial role of m6A modification in the pathogenesis and progression of various skin diseases. m6A modification affects critical biologic processes of the skin, such as inflammation, immune response and cellular ageing. This review systematically explores the landscape of m6A modification in nontumour skin diseases, elucidating its regulatory roles and therapeutic implications, including wound healing, scar and keloid, skin ageing, psoriasis, systemic lupus erythematosus, acne vulgaris, rosacea, chronic actinic dermatitis and scleroderma. The intricate mechanisms of m6A modification can lead to the development of novel diagnostic biomarkers and therapeutic strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guozhen Liu
- Department of Spinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Li Jing
- School of Basic Medical Sciences, Ningxia Key Laboratory of Vascular Injury and Repair, Ningxia Medical University, Yinchuan, China
| | - Sis Aghayants
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangjing Xu
- Department of Critical Care Medicine, Yinchuan Hospital of Traditional Chinese Medicine, Affiliated to Ningxia Medical University, Yinchuan, China
| | - Yucheng Fan
- Department of Pathology, The First People's Hospital of Shizuishan, Affiliated to Ningxia Medical University, Shizuishan, China
| |
Collapse
|
23
|
Műzes G, Sipos F. PANoptosis as a Two-Edged Sword in Colorectal Cancer: A Pathogenic Mechanism and Therapeutic Opportunity. Cells 2025; 14:730. [PMID: 40422233 DOI: 10.3390/cells14100730] [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: 04/06/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025] Open
Abstract
The examination of PANoptosis in colorectal cancer is particularly important, as many tumor cells can evade apoptotic cell death while continuing to proliferate through inflammatory mediators and creating an immunosuppressive environment. The PANoptosome functions as a regulatory complex that unites proteins governing pyroptotic, apoptotic, and necroptotic pathways, rather than allowing distinct death pathways to compete. The expression and functional status of key molecules within the PANoptosome, such as ZBP1, RIPK1, RIPK3, CASP8, and ASC, may influence tumor viability and immune detection. The tumorigenic impact of PANoptosis is complex and predominantly manifests through chronic inflammation, immune response modulation, and changes in the tumor microenvironment. PANoptosis also aids in the defense against colon cancer by directly eradicating tumor cells and modifying the cellular environment. The expression profile of PANoptosis components may possess prognostic and predictive significance. The therapeutic ramifications of PANoptosis in colorectal cancer are now being investigated through many avenues. It provides an opportunity to develop targeted therapeutic techniques. In contrast, it may also be pertinent in conjunction with immunotherapy, as PANoptosis signifies an immunogenic type of cell death and may consequently enhance the anti-tumor immune response. A thorough comprehension of how these parameters influence PANoptosis is crucial for practical implementation.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
24
|
Ahn D, Lee HK, Bae SH, Na H, Choi KC. Downregulation of transforming growth factor-β2 enhances the chemosensitivity to gemcitabine with diminished metastasis in pancreatic cancers. Biomed Pharmacother 2025; 188:118151. [PMID: 40378770 DOI: 10.1016/j.biopha.2025.118151] [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: 01/09/2025] [Revised: 04/27/2025] [Accepted: 05/07/2025] [Indexed: 05/19/2025] Open
Abstract
Pancreatic cancer is characterized by high rates of metastasis, recurrence, and chemoresistance, contributing to its poor prognosis. Transforming growth factor-β2 (TGF-β2), a member of the TGF-β family, plays a pivotal role in promoting cancer cell metastasis and mediating chemoresistance, particularly in advanced stages of tumor progression. However, the precise role of TGF-β in chemoresistance and metastasis in pancreatic cancer has not been studied yet. In the current study, we investigated the potential of human TGF-β2 antisense oligonucleotides (TGF-β2i) to enhance the chemosensitivity to gemcitabine in pancreatic cancer, using human pancreatic cancer cell lines (hPCCs; PANC-1, MIA PaCa-2, and AsPC-1), a co-culture model with human pancreatic stellate cells (hPSCs), a cancer-associated fibroblast-integrated pancreatic cancer organoid model (CIPCO), and an orthotopic xenograft mouse model. TGF-β2i decreased cell proliferation, migration, and viability in hPCCs, and its combination with gemcitabine exhibited a synergistic effect in PANC-1 and MIA PaCa-2 cells. Flow cytometry demonstrated a decrease in CD44 +CD24 +EpCAMHigh cancer stem-like cell populations following TGF-β2i treatment. In co-culture models, hPSCs-induced enhancement of hPCCs migration was attenuated by TGF-β2i. In the CIPCOs, TGF-β2i suppressed the gemcitabine-induced expression of extracellular matrix components such as COL1A1 and VIM. Furthermore, in an orthotopic mouse model generated by co-inoculating hPCCs and hPSCs into the pancreatic wall, co-treatment of TGF-β2i with gemcitabine significantly delayed tumor growth and metastasis to the liver compared to vehicle control. These findings suggest that TGF-β2i enhances chemosensitivity and suppresses metastatic properties by regulating both tumor-intrinsic and -extrinsic factors, indicating that targeting TGF-β2 could be a promising strategy for managing pancreatic cancer.
Collapse
Affiliation(s)
- Dohee Ahn
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Hong Kyu Lee
- Department of Companion Animal Health, College of Biomedical Science & Health, Inje University, Gimhae, Gyeongsangnam-do 50834, Republic of Korea
| | - Sang Hyeok Bae
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Hwayoung Na
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea.
| |
Collapse
|
25
|
Wen Y, Nie JH, Qin XM, Li ZY. Functional metabolomics revealed pyroglutamic acid may play a key role in idiopathic pulmonary fibrosis. J Pharm Biomed Anal 2025; 264:116967. [PMID: 40398246 DOI: 10.1016/j.jpba.2025.116967] [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: 02/17/2025] [Revised: 05/12/2025] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and irreversible respiratory disease with poor survival rates. Despite significant research efforts, IPF still lacks a curative treatment. Excessive epithelial-mesenchymal transition (EMT) contributes to approximately one-third of fibroblasts in pulmonary fibrosis and plays a critical role in IPF pathogenesis. Identifying factors that regulate EMT is essential for developing effective therapeutic strategies for IPF. In this study, functional metabolomics revealed significant alterations in multiple metabolites in transforming growth factor-beta 1 (TGF-β1)-induced A549 cells, with pyroglutamic acid and 5-oxoprolinase (OPLAH) being identified as the most critical factors. Cellular experiments demonstrated that pyroglutamic acid effectively inhibited TGF-β1-induced EMT in A549 cells. Mechanistically, pyroglutamic acid inhibited IPF by suppressing EMT through the inhibition of Smad2/3 expression in TGF-β1-induced A549 cells. Bioinformatics analysis further elucidated the pyroglutamate is a potential metabolite that inhibits EMT. In addition, this study is the first to highlight the pivotal role of pyroglutamic acid and OPLAH in regulating EMT in IPF, offering novel insights into the metabolic mechanisms involved in IPF inhibition and providing a foundation for developing innovative therapeutic approaches for IPF.
Collapse
Affiliation(s)
- Yang Wen
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan 030006, China
| | - Jia-Hui Nie
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan 030006, China.
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan 030006, China
| | - Zhen-Yu Li
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
26
|
Babiker NA, Nadeem S, Abu Kariem H, Abdul Hameed A, Negmeldin AT, El-labbad EM. Medicinal Chemistry Strategies in Targeting TGF-βR1 Kinase Domain: Unveiling Insights into Inhibitor Structure-Activity Relationship (SAR). Pharmaceuticals (Basel) 2025; 18:716. [PMID: 40430535 PMCID: PMC12114876 DOI: 10.3390/ph18050716] [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: 04/15/2025] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
The transforming growth factor-β (TGF-β) signaling pathway is involved in various cellular functions, including immunological response, extracellular matrix formation, differentiation, growth and development, and cell cycle regulation. The TGF β receptor type 1 (TGF-βR1) has emerged as a key component of this pathway, exhibiting significant overexpression in diverse malignancies, including hepatocellular carcinoma, gastric cancer, breast cancer, and colon cancer. Multiple therapeutic targets have been identified for the TGF-β signaling pathway, encompassing antibodies, ligand traps, vaccines, antisense oligonucleotides, and small-molecule TGF-βR1 kinase inhibitors. This review delineates the structural and functional characteristics of the small-molecule TGF-βR1 kinase inhibitors. The inhibitors discussed herein are categorized based on shared pharmacophoric features, notably a five-membered heterocyclic ring linked to three distinct features (R1, R2, and R3). These features interact with amino acids within the selectivity pocket, hinge region, or solvent-exposed area, respectively. These insights contribute to a clearer understanding of the structural requirements for selective TGF-βR1 inhibition. The presented findings in this review article offer a valuable foundation for future drug discovery efforts targeting the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Nusaiba A. Babiker
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Soam Nadeem
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Hasan Abu Kariem
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Afra Abdul Hameed
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Ahmed T. Negmeldin
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Eman M. El-labbad
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| |
Collapse
|
27
|
Toft K, Mardahl M, Hedberg-Alm Y, Anlén K, Tydén E, Nielsen MK, Honoré ML, Fromm B, Nielsen LN, Nejsum P, Thamsborg SM, Cirera S, Pihl TH. Evaluation of circulating microRNAs in plasma from horses with non-strangulating intestinal infarction and idiopathic peritonitis. Vet J 2025; 313:106378. [PMID: 40374098 DOI: 10.1016/j.tvjl.2025.106378] [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: 01/07/2025] [Revised: 05/12/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Non-strangulating intestinal infarctions (NSII) associated with Strongylus vulgaris infection and idiopathic peritonitis (IP) share similar clinical presentation but require different treatment approaches. Horses with NSII need surgical intervention, while idiopathic peritonitis cases can be successfully treated with antimicrobials. A correct diagnosis is thus crucial, but because the two diseases overlap in clinicopathological features, differentiation is difficult in clinical practice. MicroRNAs (miRNAs) are non-coding RNAs that exhibit measurable changes in abundance in tissues and circulation during disease. This study aimed to explore differences in plasma miRNA abundance between patients with NSII and IP. Plasma samples were collected from 43 horses, consisting of 21 with NSII and 22 with IP. A subset (n = 12) was submitted for deep small RNA sequencing to identify miRNAs differing between the groups. Next, a panel of nine miRNAs (two were potential normalizers) were selected for evaluation and confirmation by reverse transcription quantitative real-time PCR (RT-qPCR). Small RNA sequencing detected 628 miRNAs in the blood samples, but no miRNAs were differentially abundant between the disease groups. This finding was confirmed by qPCR. In agreement with previous studies, the top abundant miRNAs in both groups included Eca-Mir-122-5p and Eca-Mir-486-5p, as well as Eca-Mir-223-3p, which has previously been associated with inflammation. Target prediction for the most abundant miRNAs additionally predicted targets in inflammatory pathways. Evaluation of clinicopathological parameters revealed differences between the groups in two measures (white blood cell count and blood neutrophil count), which aligns with findings from previous studies. The results demonstrate that NSII and IP elicit similar miRNA profiles in plasma and are characterized by systemic inflammation.
Collapse
Affiliation(s)
- Katrine Toft
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | - Ylva Hedberg-Alm
- Department of Biomedical Science and Veterinary Public Health, Parasitology Unit, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Karin Anlén
- Evidensia Equine Hospital Helsingborg, Helsingborg, Sweden
| | - Eva Tydén
- Department of Biomedical Science and Veterinary Public Health, Parasitology Unit, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Martin K Nielsen
- M.H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Marie Louise Honoré
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bastian Fromm
- The Arctic University Museum of Norway, UiT The Arctic University of Norway, Tromsø, Norway
| | - Lise N Nielsen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Stig Milan Thamsborg
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susanna Cirera
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Holberg Pihl
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Xiao Q, Liu Y, Li T, Wang C, He S, Zhai L, Yang Z, Zhang X, Wu Y, Liu Y. Viral oncogenesis in cancer: from mechanisms to therapeutics. Signal Transduct Target Ther 2025; 10:151. [PMID: 40350456 PMCID: PMC12066790 DOI: 10.1038/s41392-025-02197-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 05/14/2025] Open
Abstract
The year 2024 marks the 60th anniversary of the discovery of the Epstein-Barr virus (EBV), the first virus confirmed to cause human cancer. Viral infections significantly contribute to the global cancer burden, with seven known Group 1 oncogenic viruses, including hepatitis B virus (HBV), human papillomavirus (HPV), EBV, Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis C virus (HCV), human T-cell leukemia virus type 1 (HTLV-1), and human immunodeficiency virus (HIV). These oncogenic viruses induce cellular transformation and cancer development by altering various biological processes within host cells, particularly under immunosuppression or co-carcinogenic exposures. These viruses are primarily associated with hepatocellular carcinoma, gastric cancer, cervical cancer, nasopharyngeal carcinoma, Kaposi sarcoma, lymphoma, and adult T-cell leukemia/lymphoma. Understanding the mechanisms of viral oncogenesis is crucial for identifying and characterizing the early biological processes of virus-related cancers, providing new targets and strategies for treatment or prevention. This review first outlines the global epidemiology of virus-related tumors, milestone events in research, and the process by which oncogenic viruses infect target cells. It then focuses on the molecular mechanisms by which these viruses induce tumors directly or indirectly, including the regulation of oncogenes or tumor suppressor genes, induction of genomic instability, disruption of regular life cycle of cells, immune suppression, chronic inflammation, and inducing angiogenesis. Finally, current therapeutic strategies for virus-related tumors and recent advances in preclinical and clinical research are discussed.
Collapse
Affiliation(s)
- Qing Xiao
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yi Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Tingting Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Chaoyu Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Sanxiu He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Liuyue Zhai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Zailin Yang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaomei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yongzhong Wu
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yao Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
29
|
Ding B, Liu X, Li Z, Xie X, Li J, Wang J, Li S, Wang P, Xie Y, Ma X, Wang H, Xie C, Qiao X, Wang Y, Xu J, Feng Y, Hao J. A novel platinum(IV) prodrug, gramine-Pt(IV) enhances chemoimmunotherapy by activating cGAS-STING and modulating TGF-β-MHC-I axis. Drug Resist Updat 2025; 81:101252. [PMID: 40382984 DOI: 10.1016/j.drup.2025.101252] [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: 02/24/2025] [Revised: 04/30/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025]
Abstract
Platinum(II) (Pt(II)) drugs, such as cisplatin and oxaliplatin, played critical roles in cancer therapy; however, their efficacy is often limited by significant toxicity and the development of drug resistance. Recently, multi-target platinum(IV) (Pt(IV)) complexes, particularly those optimized with axial ligands, have emerged as promising alternatives enhancing tumor selectivity and drug stability. In this study, we synthesized a series of novel platinum(IV) prodrugs, gramine-platinum(IV), by incorporating gramine-a natural indole alkaloid that antagonizes TGF-β receptors I and II to inhibit the TGF-β signaling pathway-as an axial ligand. Among them, compound 8 (referred to as GP) was screened out to have the best antitumor activity. GP not only enhances the therapeutic efficacy of platinum(II) drugs but also targets TGF-β signaling. Our findings demonstrate that GP rapidly enters cells and preferentially accumulates in critical subcellular compartments, such as the nucleus and mitochondria, significantly amplifying its therapeutic impact. Notably, GP exhibits great tumor accumulation compared to cisplatin and oxaliplatin, with minimal uptake in normal tissues, highlighting its superior tumor specificity with reduced systemic toxicity. This unique characteristic enables GP to enhance therapeutic efficiency through multiple modalities, including strengthening DNA damage, reducing mitochondrial membrane potential, promoting apoptosis, and arresting cell cycle in the S phase. Moreover, GP activates the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling (cGAS-STING) pathway, enhancing antigen presentation and fostering robust anti-tumor immune responses. In mouse models of pancreatic and breast cancer, GP significantly inhibits tumor growth and triggers strong innate immune activation. By combining GP with anti-PD-1 therapy, immunotherapy-resistant tumors are rendered responsive, leading to a pronounced suppression of tumor growth. Overall, GP not only amplifies the DNA-damaging effects of platinum(II) drugs but also elicits durable immune responses, establishing itself as a promising chemo-immune-combined strategy for treating pancreatic and breast cancers.
Collapse
Affiliation(s)
- Bowen Ding
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Department of Breast Oncoplastic and Reconstructive Surgery, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Xiaomeng Liu
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| | - Zhe Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| | - Xinru Xie
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jiaqi Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jiaqian Wang
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Shouyi Li
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Pengyu Wang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yongjie Xie
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiaoqing Ma
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Hongwei Wang
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Chengzhi Xie
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin 300070, China
| | - Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China.
| | - Jingyuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University, Tianjin 300070, China.
| | - Yukuan Feng
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Jihui Hao
- Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| |
Collapse
|
30
|
Rocans RP, Zarins J, Bine E, Mahauri I, Deksnis R, Citovica M, Donina S, Gravelsina S, Vilmane A, Rasa-Dzelzkaleja S, Sabelnikovs O, Mamaja B. Early Postoperative Increase in Transforming Growth Factor Beta-1 Predicts Microvascular Flap Loss in Reconstructive Surgery: A Prospective Cohort Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:863. [PMID: 40428821 PMCID: PMC12113236 DOI: 10.3390/medicina61050863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/21/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Microvascular flap surgery is a widely used reconstructive technique for the repair of various defects. Biomarkers have become an essential tool for monitoring flap viability, early detection of complications, and prediction of surgical outcomes. Studies focusing on immunomodulatory cytokines in the early prediction of microvascular flap complications are lacking. We aimed to investigate the predictive value of postoperative changes in transforming growth factor beta-1 (TGF-β1) for microvascular flap complications. Materials and Methods: This prospective observational study comprised 44 adults scheduled for elective microvascular flap surgery. Preoperative blood samples for analysis were obtained before surgery, prior to the administration of intravenous fluids. Postoperative blood draws were collected after surgery, before leaving the operating room. Preoperative and postoperative serum concentrations of TGF-β1, as well as preoperative plasma albumin, total protein, total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, full blood count, albumin, interleukin-6, C-reactive protein, and fibrinogen, were determined. Results: Postoperative changes in TGF-β1 were higher in cases with flap loss compared to patients with healthy recovery or patients with minor flap complications (0.403 log10 of ng/mL [0.024-0.782] vs. 0.157 [0.029-0.285] vs. -0.089 [-0.233-0.056], p = 0.002). Increased postoperative TGF-β1 was positively linked to preoperative C-reactive protein (p = 0.021), fibrinogen (p = 0.020), hematocrit (p = 0.039), and hemoglobin (p = 0.009). Conclusions: The postoperative increase in circulating TGF-β1 was associated with microvascular flap complications. Assessment of the postoperative changes in circulating TGF-β1 may be valuable for the early postoperative prediction of true flap loss.
Collapse
Affiliation(s)
- Rihards Peteris Rocans
- Intensive Care Clinic, Riga East Clinical University Hospital, Hipokrata Street 2, LV-1079 Riga, Latvia;
- Department of Anaesthesia and Intensive Care, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia; (I.M.); (O.S.); (B.M.)
| | - Janis Zarins
- Department of Hand and Plastic Surgery, Microsurgery Centre of Latvia, Brivibas Street 410, LV-1024 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Riga Technical University, Pulka Street 3, LV-1007 Riga, Latvia
| | - Evita Bine
- Intensive Care Clinic, Riga East Clinical University Hospital, Hipokrata Street 2, LV-1079 Riga, Latvia;
| | - Insana Mahauri
- Department of Anaesthesia and Intensive Care, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia; (I.M.); (O.S.); (B.M.)
| | - Renars Deksnis
- Surgical Oncology Clinic, Riga East Clinical University Hospital, Hipokrata Street 4, LV-1079 Riga, Latvia;
| | - Margarita Citovica
- Laboratory Department, Riga East Clinical University Hospital, Hipokrata Street 2, LV-1079 Riga, Latvia;
| | - Simona Donina
- Institute of Microbiology and Virology, Rīga Stradiņš University, Ratsupites Street 5, LV-1067 Riga, Latvia; (S.D.); (S.G.); (S.R.-D.)
- Outpatient Department, Riga East Clinical University Hospital, Hipokrata Street 4, LV-1079 Riga, Latvia
| | - Sabine Gravelsina
- Institute of Microbiology and Virology, Rīga Stradiņš University, Ratsupites Street 5, LV-1067 Riga, Latvia; (S.D.); (S.G.); (S.R.-D.)
| | - Anda Vilmane
- Institute of Microbiology and Virology, Rīga Stradiņš University, Ratsupites Street 5, LV-1067 Riga, Latvia; (S.D.); (S.G.); (S.R.-D.)
| | - Santa Rasa-Dzelzkaleja
- Institute of Microbiology and Virology, Rīga Stradiņš University, Ratsupites Street 5, LV-1067 Riga, Latvia; (S.D.); (S.G.); (S.R.-D.)
| | - Olegs Sabelnikovs
- Department of Anaesthesia and Intensive Care, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia; (I.M.); (O.S.); (B.M.)
| | - Biruta Mamaja
- Department of Anaesthesia and Intensive Care, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia; (I.M.); (O.S.); (B.M.)
| |
Collapse
|
31
|
Micale L, Di Muro E, De Cegli R, Tumaini B, Capuozzo A, Bernardi P, Morlino S, Fusco C, Nardella G, Mormone E, Vaccaro L, Del Prete E, Giachino D, Giuliani M, Leoni C, Mercadante F, Moroni A, Piscopo C, Zollino M, Cacchiarelli D, Sbarbati A, Medina DL, Di Bernardo D, Castori M. Multi-OMICs analysis on tridimensional fibroblast spheroids to model vascular Ehlers-Danlos syndrome pathogenesis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167896. [PMID: 40345454 DOI: 10.1016/j.bbadis.2025.167896] [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: 01/09/2025] [Revised: 04/04/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Three-dimensional (3D) spheroids are an innovative cellular model mimicking tissue-like properties for a more effective replication of physiological cellular environment. Vascular Ehlers-Danlos syndrome (vEDS) is a rare hereditary connective tissue disorder caused by heterozygous deleterious variants in COL3A1. Affected individuals are at increased risk of early death due to ruptures of arteries, large intestine, and gravid uterus. vEDS cellular pathogenesis is only partially understood and the disease remains without effective treatment. We integrated transcriptomic and proteomic data generated from 2D fibroblast cultures and 3D spheroids from ten patients and four controls. Transcriptomic analysis revealed upregulation of genes related to mitochondrial function, organellar ribosomal subunits, and biosynthesis processes, to indicate an augmented adaptive metabolic response, while downregulation of genes involved in cell migration, differentiation, and stress response highlighted abnormalities in cellular signaling and extracellular matrix maintenance. Proteomic analysis found that induced proteins were significantly enriched for the mitochondrial matrix and minichromosome maintenance complex as well as in biological processes involving low-density lipoprotein particles, and cellular response to catabolic processes and DNA damage stimuli. Ultrastructural analysis and high-content imaging documented an endoplasmic reticulum dilation, increased autophagosomes and lipofuscin deposits. Our findings expand current knowledge on the multi-OMIC profile of vEDS by highlighting potential convergent mechanisms and novel features acting as master regulators of the emerging phenotype. This study supports, for the first time, 3D fibroblast spheroids as a suitable experimental tool to dissect vEDS pathogenesis and a crucial model for identifying new therapeutic targets.
Collapse
Affiliation(s)
- Lucia Micale
- UOC Genetica Medica, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Ester Di Muro
- UOC Genetica Medica, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Barbara Tumaini
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | | | - Paolo Bernardi
- Department of Neurosciences, Biomedicine and Movement Sciences, Unit of Human Anatomy, University of Verona, Verona, Italy
| | - Silvia Morlino
- UOC Genetica Medica, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Carmela Fusco
- UOC Genetica Medica, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Grazia Nardella
- UOC Genetica Medica, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Elisabetta Mormone
- Institute of Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Lorenzo Vaccaro
- Armenise/Harvard Laboratory of Integrative Genomics, Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy; Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| | - Eugenio Del Prete
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Daniela Giachino
- Medical Genetic Unit, San Luigi Gonzaga University Hospital, Torino, Italy; Dept. of Clinical and Biological Sciences, University of Torino, Italy
| | - Michele Giuliani
- Unit of Dermatology, Division of Internal Medicine, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Chiara Leoni
- Center for Rare Disease and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione IRCCS Policlinico Universitario Agostino Gemelli, Rome, Italy
| | - Francesca Mercadante
- Division of Medical Genetics, AOOR Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Alice Moroni
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Carmelo Piscopo
- Medical and Laboratory Genetics Unit, AORN Antonio Cardarelli Hospital, Naples, Italy
| | - Marcella Zollino
- Institute of Genomic Medicine, Department of Life Sciences and Public Health, 'Sacro Cuore' Catholic University of Rome, Rome, Italy
| | - Davide Cacchiarelli
- Armenise/Harvard Laboratory of Integrative Genomics, Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy; Department of Translational Medicine, University of Naples Federico II, Naples, Italy; Genomics and Experimental Medicine Program, Scuola Superiore Meridionale, Naples, Italy
| | - Andrea Sbarbati
- Department of Neurosciences, Biomedicine and Movement Sciences, Unit of Human Anatomy, University of Verona, Verona, Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy; Department of Translational Medicine, University of Naples Federico II, Naples, Italy
| | - Diego Di Bernardo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy; Department of Chemical Materials and Industrial Engineering, University of Naples Federico II, Naples, Italy
| | - Marco Castori
- UOC Genetica Medica, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| |
Collapse
|
32
|
Milić MS, Dželetović B, Radičević BA, Milosavljević N, Jovanović ND, Dožić I, Krunić J, Đukić L. Transforming growth factor-β1 and its soluble receptor type 2 in saliva of young adults: Sex-related differences and predictive modeling of salivary concentrations. Arch Oral Biol 2025; 175:106279. [PMID: 40347849 DOI: 10.1016/j.archoralbio.2025.106279] [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: 01/04/2025] [Revised: 03/24/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
OBJECTIVE To investigate and compare salivary presence and levels of Transforming Growth Factor-β1 (TGF-β1) and its soluble receptor type 2 (TGFBR2), along with the biochemical profile of the unstimulated whole saliva (UWS) of healthy young males and females; and to assess the potential of the predictive modeling for estimating the active TGF-β1 and TGFBR2 levels based on sex and individual salivary biochemical profile. DESIGN Study sample included 20 participants, both sexes, with the median value of age being 20.00 (1.00) years. Total and active TGF-β1 and TGFBR2 levels were tested with ELISA. Biochemical analysis of saliva, including lactat dehydrogenase (LDH) activity and uric acid levels, was conducted via spectrophotometry. Salivary pH and buffer capacity were determined with potentiometry. To model the relationship of active TGF-β1 and TGFBR2 levels with sex and individual salivary biochemical profiles, multivariate regression analysis was employed. RESULTS The median/mean values of active TGF-β1 (33.13 (27.26) vs. 14.24 (9.89) pg/ml, p = 0.013), uric acid (260.00 (136.00) vs. 199.00 (74.50) μmol/L, p = 0.031), and LDH activity (82.00 ± 45.23 vs. 42.40 ± 30.99 U/L, p = 0.035) were significantly higher in males vs. females, respectively. Multivariate regression method demonstrated 71.9 % accuracy in predicting the levels of active TGF-β1, while for TGFBR2 the accuracy was 86.01 %. CONCLUSIONS Salivary levels of active TGF-β1 are significantly higher in young healthy males compared to females. The multivariate regression model demonstrates promising predictive potential for estimating the levels of active TGF-β1 and TGFBR2 in young healthy individuals, based on sex and individual salivary biochemical profiles.
Collapse
Affiliation(s)
- Marija S Milić
- University of Belgrade, School of Dental Medicine, Department of General and Oral Physiology, Belgrade, Serbia
| | - Bojan Dželetović
- University of Belgrade, School of Dental Medicine, Department of Restorative Odontology and Endodontics, Belgrade, Serbia
| | - Biljana Anđelski Radičević
- University of Belgrade, School of Dental Medicine, Department of General and Oral Biochemistry, Belgrade, Serbia
| | - Nataša Milosavljević
- University of Belgrade, Faculty of Agriculture, Department of Mathematics and Informatics, Belgrade, Serbia
| | - Nina Dimitrijević Jovanović
- University of Belgrade, School of Dental Medicine, Department of General and Oral Biochemistry, Belgrade, Serbia
| | - Ivan Dožić
- University of Belgrade, School of Dental Medicine, Department of General and Oral Biochemistry, Belgrade, Serbia
| | - Jelena Krunić
- University of East Sarajevo, Faculty of Medicine Foca, Department of Dental Pathology, Foca, Republic of Srpska, Bosnia and Herzegovina; University of Montenegro, Faculty of Medicine, Department of Dentistry, Podgorica, Montenegro
| | - Ljiljana Đukić
- University of Belgrade, School of Dental Medicine, Department of Pharmacology in Dentistry, Belgrade, Serbia.
| |
Collapse
|
33
|
Mimmi S, Parrotta EI, Tolomeo AM, Maisano D, Crapella V, Pingitore E, Fatima K, Zimbo AM, Talarico M, Cristiani CM, Scaramuzzino L, Valente D, Zannino C, Cuda G, Quattrone A, Iaccino E, Quattrone A. Neuronally-Derived Extracellular Vesicles Transforming Growth Factor Beta-1 Levels in Progressive Supranuclear Palsy. Mov Disord 2025. [PMID: 40317792 DOI: 10.1002/mds.30222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/26/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Differentiating progressive supranuclear palsy (PSP) from other parkinsonian disorders may be challenging. OBJECTIVES To investigate the role of transforming growth factor beta-1 (TGFβ1) in PSP. METHODS A total of 33 PSP, 39 Parkinson's disease (PD), 8 multiple system atrophy (MSA) patients, and 50 healthy controls (HC) were enrolled. TGFβ1 levels, including both active and inactive forms (latency-associated peptide [LAP]-TGFβ1), were measured in serum, total extracellular vesicles (EVs), and neuronally-derived EVs (NDEVs) using microfluidic assays and ELISA. RESULTS PSP patients exhibited a marked increase in TGFβ1 and LAP-TGFβ1 levels in NDEVs, while no differences were observed across groups in serum or total EVs. Receiver operating characteristic (ROC) analysis demonstrated outstanding performance in differentiating PSP from non-PSP patients (TGFβ1, area under the curve [AUC]: 0.97; LAP-TGFβ1, AUC: 1.00), HC, AUC: 1.00). CONCLUSIONS This study highlights TGFβ1 and LAP-TGFβ1 in NDEVs as promising blood-based non-invasive biomarkers for PSP diagnosis, paving the way for further research on these proteins in PSP. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Selena Mimmi
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | | | - Anna Maria Tolomeo
- Department of Cardiac, Thoracic and Vascular Science and Public Health, University of Padova, Padua, Italy
- Institute of Pediatric Research 'Città della Speranza', Padua, Italy
| | | | - Valentina Crapella
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Elisabetta Pingitore
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Khushboo Fatima
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Anna Maria Zimbo
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Mariagrazia Talarico
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Costanza Maria Cristiani
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Luana Scaramuzzino
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Desirèe Valente
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Clara Zannino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Gianni Cuda
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Aldo Quattrone
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Enrico Iaccino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Andrea Quattrone
- Neuroscience Research Center, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
- Institute of Neurology, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
34
|
Davis J, Maranto M, Kennedy J, Wang X, Azhar M, Jain A, Evans CE. Transforming Growth Factors in Venous Thrombus Formation and Resolution. Arterioscler Thromb Vasc Biol 2025; 45:643-653. [PMID: 40109257 PMCID: PMC12018122 DOI: 10.1161/atvbaha.124.322395] [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] [Indexed: 03/22/2025]
Abstract
Deep vein thrombosis (DVT) and pulmonary embolism are vascular occlusive disorders categorized under the term venous thromboembolism. Venous thromboembolism affects ≈900 000 people per year in the United States alone. Understanding of the multifaceted process of DVT has improved in recent years, and current DVT treatments reduce thrombus propagation, but they also increase bleeding risk and fail to accelerate natural venous thrombus resolution. Multiple inflammatory cytokines regulate the development and subsequent resolution of DVT. One family of cytokines involved in DVT and venous thrombus resolution is the TGF-β (transforming growth factor-β) family. A comprehensive understanding of the control of venous thrombus formation and resolution by the TGF-β family could lead to the development of novel treatments for DVT that target ≥1 of the TGF-β isoforms. The aim of this review is to describe studies of the roles of the TGF-β isoforms in venous thrombus formation and resolution and to highlight opportunities for future research. TGF-β isoforms include TGF-β1, TGF-β2, and TGF-β3. TGF-β1 has a well-characterized role in the positive regulation of venous thrombus formation and the negative regulation of venous thrombus resolution. Further research is necessary, however, to understand the potential roles of TGF-β2 and TGF-β3 in venous thrombus formation and resolution. Given that TGF-β1 expression increases during venous thrombosis and that inhibition or knockdown of TGF-β1 reduces thrombus burden, TGF-β1 represents a potential diagnostic marker for DVT and a putative target for therapies that aim to prevent or treat DVT.
Collapse
Affiliation(s)
- Jonathan Davis
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Molly Maranto
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Jonathan Kennedy
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Xiaoqin Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Mohamad Azhar
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Colin E. Evans
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, South Carolina, USA
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
- Department of Biomedical Engineering, University of South Carolina, Columbia, South Carolina, USA
- Institute on Cardiovascular Disease Research, University of South Carolina, Columbia, USA
| |
Collapse
|
35
|
Wang C, Shu Y, Shan J, Li K, Wan S, Chen S, Li X, Niu J, Yang L. Discovery and Validation of a New Biomarker Integrating Ferroptosis and Glycolysis-Related Genes in Bladder Cancer. IUBMB Life 2025; 77:e70028. [PMID: 40401561 DOI: 10.1002/iub.70028] [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: 03/12/2025] [Revised: 04/29/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
Bladder cancer (BCa) is a highly invasive tumor with few successful therapies, and its unfavorable prognosis mainly stems from late diagnosis and resistance to treatment. Ferroptosis is a type of non-apoptotic cell death characterized by iron-dependent regulated necrosis due to extensive lipid peroxidation. Glycolysis is fundamental to cancer cell metabolism, with cancer cells developing various strategies to enhance this process. In this study, we combined ferroptosis and glycolysis gene sets, two biological processes closely related to tumorigenesis and development, and obtained ferroptosis and glycolysis-related gene sets (FGRGs). By leveraging both single-cell and bulk transcriptome data from BCa, we have investigated the presence and role of FGRGs in the onset and progression of BCa through various approaches. Using machine learning algorithms, we identified a feature gene set consisting of 13 genes in the TCGA data set to predict the prognosis of BCa and verified it in the GEO data set. After that, we explored FGRGs in depth using a variety of bioinformatics analyses, such as mutational landscape analysis, functional enrichment analysis, immune infiltration analysis, FGRGs-associated risk and clinical characterization, and drug susceptibility analysis. Finally, we validated the function of the core gene chondroitin polymerizing factor 2 (CHPF2) using CCK-8, clone formation, transwell, and wound healing assays. Our research innovatively combines ferroptosis with glycolytic genes and applies it as an independent prognostic factor in the study of BCa. It reveals new characteristic genes and therapeutic targets that can predict the prognosis of BCa patients and lays a foundation for the study of the occurrence and development mechanism of BCa and targeted data strategies.
Collapse
Affiliation(s)
- Chenyang Wang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yuncong Shu
- School of Life Science, Lanzhou University, Lanzhou, China
| | - Jiaqi Shan
- School of Medicine, Hubei Minzu University, Enshi, China
| | - Kunpeng Li
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Shun Wan
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Siyu Chen
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaoran Li
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jiping Niu
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Li Yang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
36
|
Höpfl S, Özverin M, Nowack H, Tamas R, Clark AG, Radde N, Olayioye MA. Integrated mathematical and experimental modeling uncovers enhanced EMT plasticity upon loss of the DLC1 tumor suppressor. PLoS Comput Biol 2025; 21:e1013076. [PMID: 40354489 PMCID: PMC12121911 DOI: 10.1371/journal.pcbi.1013076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 05/29/2025] [Accepted: 04/21/2025] [Indexed: 05/14/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays an essential role in embryonic development, wound healing, and tumor progression. Partial EMT states have been linked to metastatic dissemination and drug resistance. Several interconnected feedback loops at the RNA and protein levels control the transition between different cellular states. Using a combination of mathematical modeling and experimental analyses in the TGFβ-responsive breast epithelial MCF10A cell model, we identify a central role for the tumor suppressor protein Deleted in Liver Cancer 1 (DLC1) during EMT. By extending a previous model of EMT comprising key transcription factors and microRNAs, our work shows that DLC1 acts as a positive regulator of TGFβ-driven EMT, mainly by promoting SNAIL1 expression. Our model predictions indicate that DLC1 loss impairs EMT progression. Experimental analyses confirm this prediction and reveal the acquisition of a partial EMT phenotype in DLC1-depleted cells. Furthermore, our model results indicate a possible EMT reversion to partial or epithelial states upon DLC1 loss in MCF10A cells induced toward mesenchymal phenotypes. The increased EMT plasticity of cells lacking DLC1 may explain its importance as a tumor suppressor.
Collapse
Affiliation(s)
- Sebastian Höpfl
- Institute for Stochastics and Applications, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Merih Özverin
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Helena Nowack
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Raluca Tamas
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Andrew G. Clark
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- University of Tübingen, Center for Personalized Medicine, Tübingen, Stuttgart, Germany
| | - Nicole Radde
- Institute for Stochastics and Applications, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Monilola A. Olayioye
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
37
|
Kuchay MS, Choudhary NS, Ramos-Molina B. Pathophysiological underpinnings of metabolic dysfunction-associated steatotic liver disease. Am J Physiol Cell Physiol 2025; 328:C1637-C1666. [PMID: 40244183 DOI: 10.1152/ajpcell.00951.2024] [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: 01/22/2025] [Revised: 01/31/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is emerging as the leading cause of chronic liver disease worldwide, reflecting the global epidemics of obesity, metabolic syndrome, and type 2 diabetes. Beyond its strong association with excess adiposity, MASLD encompasses a heterogeneous population that includes individuals with normal body weight ("lean MASLD") highlighting the complexity of its pathogenesis. This disease results from a complex interplay between genetic susceptibility, epigenetic modifications, and environmental factors, which converge to disrupt metabolic homeostasis. Adipose tissue dysfunction and insulin resistance trigger an overflow of lipids to the liver, leading to mitochondrial dysfunction, oxidative stress, and hepatocellular injury. These processes promote hepatic inflammation and fibrogenesis, driven by cross talk among hepatocytes, immune cells, and hepatic stellate cells, with key contributions from gut-liver axis perturbations. Recent advances have unraveled pivotal molecular pathways, such as transforming growth factor-β signaling, Notch-induced osteopontin, and sphingosine kinase 1-mediated responses, that orchestrate fibrogenic activation. Understanding these interconnected mechanisms is crucial for developing targeted therapies. This review integrates current knowledge on the pathophysiology of MASLD, emphasizing emerging concepts such as lean metabolic dysfunction-associated steatohepatitis (MASH), epigenetic alterations, hepatic extracellular vesicles, and the relevance of extrahepatic signals. It also discusses novel therapeutic strategies under investigation, aiming to provide a comprehensive and structured overview of the evolving MASLD landscape for both basic scientists and clinicians.
Collapse
Affiliation(s)
| | - Narendra Singh Choudhary
- Institute of Digestive and Hepatobiliary Sciences, Medanta-The Medicity Hospital, Gurugram, India
| | - Bruno Ramos-Molina
- Group of Obesity, Diabetes & Metabolism, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| |
Collapse
|
38
|
Zhao X, Xia F, Dong Z, Huang W, Kong X, Cui Z, Yan M, Gao H, Rong R, Wang M, Liu G, Zhang Z, Zhang J, Yuan T, Cai H, Yan Z, Zhu L, Qin W. A novel EndMT inhibitor, xanthotoxin, attenuates non-alcoholic fatty liver disease by acting as TGFβR2 antagonist. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156823. [PMID: 40347928 DOI: 10.1016/j.phymed.2025.156823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/12/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Endothelial-to-mesenchymal transition (EndMT) has emerged as a key process contributing to the pathology of non-alcoholic fatty liver disease (NAFLD). Thus, identifying EndMT inhibitors may help impede NAFLD progression. PURPOSE Our research aims to identify potent natural EndMT inhibitors and explore their therapeutic potential and mechanisms of action in NAFLD. METHODS A natural compound library was employed to screen potential EndMT inhibitors. High-fat diet (HFD)-induced ApoE-/- mice and free fatty acid (FFA)-treated human hepatic sinusoidal endothelial cells (HHSECs) were employed as animal and cellular models of NAFLD. EndMT was evaluated by western blotting, qRT-PCR, immunofluorescence staining, tube formation, wound healing, and transwell assays. LC-MS/MS was applied to screen for altered secreted proteins during EndMT. Molecular docking, CETSA, and SPR assays were employed to validate the combination of xanthotoxin with TGFβR2. RESULTS Xanthotoxin was identified as a novel EndMT inhibitor. Further investigation revealed that xanthotoxin ameliorates NAFLD in ApoE-/- mice. By inhibiting EndMT, xanthotoxin improves endothelial dysfunction, reduces the pro-NAFLD factor ANGPTL2 secretion, and increases the anti-NAFLD factor SOD2 secretion, thus reducing hepatocyte steatosis, inflammation, and hepatic stellate cell fibrosis. Additional studies demonstrated that xanthotoxin binds to TGFβR2 and acts as its antagonist to block EndMT. In mice, EC-specific overexpression of TGFβR2 negated xanthotoxin's therapeutic impact on NAFLD. CONCLUSION This study reveals for the first time that xanthotoxin attenuates NAFLD by acting as a TGFβR2 antagonist to inhibit EndMT. These findings highlight the significant therapeutic potential of xanthotoxin in NAFLD treatment.
Collapse
Affiliation(s)
- Xiaona Zhao
- School of Pharmacy, Shandong Second Medical University, Weifang 261000, Shandong, China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China
| | - Fangjie Xia
- School of Pharmacy, Shandong Second Medical University, Weifang 261000, Shandong, China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China
| | - Zixu Dong
- School of Pharmacy, Shandong Second Medical University, Weifang 261000, Shandong, China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China
| | - Wenyang Huang
- School of Pharmacy, Shandong Second Medical University, Weifang 261000, Shandong, China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China
| | - Xinxin Kong
- School of Pharmacy, Shandong Second Medical University, Weifang 261000, Shandong, China; School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China
| | - Zhoujun Cui
- Department of General Surgery, Rizhao People's Hospital, Rizhao 276800, China
| | - Maocai Yan
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China
| | - Honggang Gao
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China
| | - Ruixue Rong
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, China
| | - Minghui Wang
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, China
| | - Guoqing Liu
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, China
| | - Zejin Zhang
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China; School of Pharmacy, Binzhou Medical University, Yantai 264000, Shandong, China
| | - Jing Zhang
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China
| | - Tao Yuan
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China; School of Pharmacy, Shandong First Medical University, Jinan 250000, Shandong, China
| | - Huiying Cai
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, China
| | - Zhenzhen Yan
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, China
| | - Lin Zhu
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, China
| | - Wei Qin
- School of Pharmacy, Jining Medical University, Rizhao 276800, Shandong, China; Department of Cardiology (Shandong Provincial Key Laboratory for Cardiovascular Disease Diagnosis and Treatment) at Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University, Jinan 250000, Shandong, China.
| |
Collapse
|
39
|
Mao Y, Xu W, Chen L, Liao H. Computational Drug Repurposing Screening Targeting Profibrotic Cytokine in Acute Respiratory Distress Syndrome. Cell Biochem Biophys 2025:10.1007/s12013-025-01762-x. [PMID: 40304856 DOI: 10.1007/s12013-025-01762-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a severe lung disease with a high fatality rate and few treatment options. Targeting certain signalling pathways, notably the Transforming Growth Factor-beta (TGF-beta) signalling pathway, has emerged as a promising option for ARDS therapy. We identified TGF-beta Receptor 1 (TGFBR1) as a major target for ARDS treatment using the STRING and KEGG databases and validated TGFBR1's critical function in the TGF-beta signalling pathway, which is important in ARDS pathogenesis. To find prospective TGFBR1 inhibitors, we selected two FDA-approved medicines, Galunisertib and Vactosertib, which are established pharmacological profiles in cancer and fibrotic illnesses. Furthermore, the SwissSimilarity platform's ligand-based virtual screening revealed structurally related drugs in the DrugBank and ChEMBL databases. Among these, seven candidates were selected for further consideration. Molecular docking experiments found that DB08387 and CHEMBL14297639 had the strongest affinity for TGFBR1, creating strong hydrogen bonds at key sites. These findings point to their potential as TGFBR1 inhibitors in ARDS treatment. The pharmacokinetic screening revealed that most of the chosen compounds had favourable ADME features, with CHEMBL14297639 standing out for its low gastrointestinal absorption and limited cytochrome P450 inhibition. This study demonstrates the possibility of targeting TGFBR1 with Galunisertib, Vactosertib, and other prospective ARDS treatments. The findings lay the groundwork for additional experimental validation and the development of innovative therapeutics aimed at reducing ARDS severity.
Collapse
Affiliation(s)
- Yong Mao
- Department of Intensive Care Unit, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Wei Xu
- Department of Intensive Care Unit, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Li Chen
- Department of Intensive Care Unit, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Handi Liao
- Department of Intensive Care Unit, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
40
|
Gao X, Cai S, Li X, Wu G. Sepsis-induced immunosuppression: mechanisms, biomarkers and immunotherapy. Front Immunol 2025; 16:1577105. [PMID: 40364841 PMCID: PMC12069044 DOI: 10.3389/fimmu.2025.1577105] [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: 02/15/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Sepsis, a life-threatening organ dysfunction resulting from a dysregulated host response to infection, initiates a complex immune response that varies over time, characterized by sustained excessive inflammation and immunosuppression. Sepsis-induced immunosuppression is now recognized as a major cause of septic death, and identifying effective strategies to counteract it poses a significant challenge. This immunosuppression results from the disruption of immune homeostasis, characterized by the abnormal death of immune effector cells, hyperproliferation of immune suppressor cells, release of anti-inflammatory cytokines, and expression of immune checkpoints. Preclinical studies targeting immunosuppression, particularly with immune checkpoint inhibitors, have shown promise in reversing immunocyte dysfunctions and establishing host resistance to pathogens. Here, our review highlights the mechanisms of sepsis-induced immunosuppression and current diagnostic biomarkers, as well as immune-enhancing strategies evaluated in septic patients and therapeutics under investigation.
Collapse
Affiliation(s)
- Xun Gao
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
- Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Shijie Cai
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
- Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Xiao Li
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
- Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Guoqiu Wu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China
- Department of Laboratory Medicine, Medical School of Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
41
|
Liao W, Ang Y, Kee ACL, Lim V, Lim AYH, Chai CLL, Wong WSF. Calcaratarin D, A Labdane Diterpenoid, Attenuates Bleomycin-Induced Pulmonary Fibrosis by Blocking Wnt/β-Catenin Signaling Pathway. Pharmacol Res 2025; 216:107756. [PMID: 40311955 DOI: 10.1016/j.phrs.2025.107756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is one of the most common interstitial lung diseases with a high mortality rate. Calcaratarin D (CalD), a labdane diterpenoid, has been shown to possess anti-inflammatory properties. The present study evaluated the therapeutic potential of CalD in pulmonary fibrosis. A single dose of bleomycin (BLM, 2.5mg/kg) was instilled intratracheally in mice for up to 21 days to develop lung fibrosis. Oral CalD (50mg/kg) reduced BLM-induced inflammatory cell infiltration, especially pro-fibrotic Arg1-expressing interstitial macrophages in the bronchoalveolar lavage fluid. During the late fibrotic phase, CalD decreased BLM-induced mortality and body weight loss. In addition, CalD ameliorated lung histopathology, reduced collagen deposition and mucus hypersecretion, and improved lung functions in BLM-exposed mice. Furthermore, CalD modulated the levels of pro-inflammatory cytokines, chemokines, and growth factors in BAL fluid and lung tissues. In mouse lungs, BLM selectively upregulated Wnt10A level and promoted β-catenin nuclear translocation. CalD not only blocked Wnt10A/β-catenin signaling pathway but also reduced pro-fibrotic markers such as collagens, α-SMA and FHL2. In normal human lung fibroblasts, CalD inhibited TGF-β1-stimulated pro-fibrotic markers and Wnt/β-catenin signaling pathway by reducing Wnt10A production, upregulating endogenous Wnt antagonist DKK1 level, dephosphorylating Wnt ligand co-receptor LRP6, and preventing β-catenin and YAP/TAZ nuclear translocation. The antifibrotic action of CalD was shown to be dependent on its α,β-unsaturated γ-butyrolactone structure that is essential for CalD to form covalent interaction with cellular protein targets. Our results imply that CalD could be a novel antifibrotic agent for IPF, acting through blockade of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore
| | - Yuet Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore
| | - Adrian C L Kee
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Health System, Singapore
| | - Valencia Lim
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Health System, Singapore
| | - Albert Y H Lim
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christina L L Chai
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore; Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore; Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore.
| |
Collapse
|
42
|
Eraković M, Bekić M, Đokić J, Tomić S, Vučević D, Pavlović L, Duka M, Marković M, Bokonjić D, Čolić M. Biodentine Stimulates Calcium-Dependent Osteogenic Differentiation of Mesenchymal Stromal Cells from Periapical Lesions. Int J Mol Sci 2025; 26:4220. [PMID: 40362457 PMCID: PMC12072047 DOI: 10.3390/ijms26094220] [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: 03/11/2025] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Biodentine, a tricalcium silicate cement, has emerged as a retrograde root-end filling material to promote periapical lesion (PL) healing after apicoectomy. However, its underlying mechanisms remain unclear. This study tested the hypothesis that Biodentine stimulates the osteogenic differentiation of mesenchymal stromal cells (MSCs) derived from PLs. The Biodentine extract (B-Ex) was prepared by incubating polymerized Biodentine in RPMI medium (0.2 g/mL) for three days at 37 °C. B-Ex, containing both released microparticles and soluble components, was incubated with PL-MSCs cultured in either a basal MSC medium or suboptimal osteogenic medium. Osteogenic differentiation was assessed by Alizarin Red staining and the expression of 20 osteoblastogenesis-related genes. Non-cytotoxic concentrations of B-Ex stimulated the proliferation of PL-MSCs and induced their osteogenic differentiation in a dose-dependent manner, with a significantly enhanced effect in suboptimal osteogenic medium. B-Ex upregulated most early and late osteoblastic genes. However, the differentiation process was prolonged, as indicated by the delayed expression of wingless-type MMTV integration site family member 2 (WNT2), bone gamma-carboxyglutamate protein (BGLAP), bone morphogenic protein-2 (BMP-2), growth hormone receptor (GHR), and FOS-like 2, AP-1 transcription factor subunit (FOSL2), compared with their expression under optimal osteogenic conditions. The stimulatory effect of B-Ex was primarily calcium dependent, as it was reduced by 85% when B-Ex was treated with the calcium-chelating agent EGTA. In conclusion, Biodentine promotes the osteogenic differentiation of PL-MSCs in a calcium-dependent manner, supporting its stimulatory role in periapical healing.
Collapse
Affiliation(s)
- Mile Eraković
- Clinic for Stomatology, Medical Faculty of the Military Medical Academy, University of Defense, 11154 Belgrade, Serbia; (M.E.); (M.D.)
| | - Marina Bekić
- Institute for the Application of Nuclear Energy, University of Belgrade, 11080 Belgrade, Serbia; (M.B.); (S.T.); (L.P.); (M.M.)
| | - Jelena Đokić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia;
| | - Sergej Tomić
- Institute for the Application of Nuclear Energy, University of Belgrade, 11080 Belgrade, Serbia; (M.B.); (S.T.); (L.P.); (M.M.)
| | - Dragana Vučević
- Center for Medical Scientific Information, Faculty of Medicine of the Military Medical Academy, University of Defence, 11040 Belgrade, Serbia;
| | - Luka Pavlović
- Institute for the Application of Nuclear Energy, University of Belgrade, 11080 Belgrade, Serbia; (M.B.); (S.T.); (L.P.); (M.M.)
| | - Miloš Duka
- Clinic for Stomatology, Medical Faculty of the Military Medical Academy, University of Defense, 11154 Belgrade, Serbia; (M.E.); (M.D.)
| | - Milan Marković
- Institute for the Application of Nuclear Energy, University of Belgrade, 11080 Belgrade, Serbia; (M.B.); (S.T.); (L.P.); (M.M.)
| | - Dejan Bokonjić
- Medical Faculty Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina;
| | - Miodrag Čolić
- Medical Faculty Foča, University of East Sarajevo, 73300 Foča, Bosnia and Herzegovina;
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| |
Collapse
|
43
|
Yang Y, Zhao L, Gao F, Wu G, Luo Y, An M. Modulation of renal fibrosis-related signaling pathways by traditional Chinese medicine: molecular mechanisms and experimental evidence. Int Urol Nephrol 2025:10.1007/s11255-025-04532-z. [PMID: 40293615 DOI: 10.1007/s11255-025-04532-z] [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: 02/10/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Renal fibrosis (RF), characterized by excessive deposition of extracellular matrix leading to tissue damage and scar formation, represents a refractory disease and a pivotal pathological basis for the progression to end-stage renal disease. The pathogenesis of RF is intricate, prominently implicating multiple key signaling pathways, including adenosine monophosphate-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR), phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), transforming growth factor-β1/small mother against decapentaplegic (TGF-β1/Smad), toll-like receptor 4/nuclear factor kappa B (TLR4/NF-κB), wingless integrated/β-catenin (Wnt/β-catenin), hypoxia-inducible factor-1α (HIF-1α), Hedgehog, and mitogen-activated protein kinase (MAPK). The current Western medical practices primarily rely on supportive and replacement therapies, which are often costly and suboptimal in efficacy. In contrast, traditional Chinese medicine (TCM), with its inherent advantages of multi-target, multi-pathway, and multi-effect modulation, emerges as a promising new strategy for RF treatment. However, a systematic, comprehensive, and detailed summary of these advancements remains absent. Therefore, this review consolidates the recent research progress on TCM modulation of RF-related signaling pathways, aiming to provide a theoretical foundation for further investigations into RF and the development of TCM interventions.
Collapse
Affiliation(s)
- Yufei Yang
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Longshan Zhao
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
- Department of Pharmacy, Shenyang Pharmaceutical University, Benxi, 117004, China
| | - Fengli Gao
- Department of Pharmacy, Second Affiliated Hospital of Baotou Medical College, Baotou, 014030, China
| | - Guodong Wu
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Yiduo Luo
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China
| | - Ming An
- Department of Pharmacy, Baotou Medical College, 31 Jianshe Road, Donghe District, Baotou, 014040, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
44
|
Zhang YS, Chen HC, Cao JX, Zhou SW, Ma YZ, Jing YH. Oxytocin-Mediate Modulation of Splenic Immunosuppression in Chronic Social Stress Through Neuroendocrine Pathways. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500849. [PMID: 40285614 DOI: 10.1002/advs.202500849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Chronic social stress (CSS) is a significant public health challenge that negatively impacts behavior and immune function through brain-spleen interactions. Oxytocin (OT), a neuropeptide critical for social behavior and immune regulation, is upregulated during CSS, though its underlying mechanisms remain unclear. This study investigates the role of OT in splenic immune modulation using a murine model of CSS. Behavioral evaluations, serum oxytocin quantification, and splenic immunophenotypic analysis were performed. Splenic denervation confirmed OT's neuromodulatory role, whereas OTR antagonism revealed its endocrine function. CSS-induced OT elevation was associated with immunosuppression, characterized by increased Foxp3⁺ regulatory T cells and reduced CD4⁺ T and CD19⁺ B cells. OT also modulated macrophage polarization, inhibiting M1-like (pro-inflammatory) and enhancing M2-like (anti-inflammatory) phenotypes. Denervation or pharmacological blockade of OT signaling partly reversed CSS-induced splenic immunosuppression but adversely affected survival in CSS-exposed mice. Additionally, denervation or OTR antagonism reduced the mice's response to social defeat, as shown by decreased social avoidance behavior. These findings suggest that OT-mediated immunosuppression likely represents a compensatory mechanism in response to chronic social stress. Targeting the OT-immune axis could offer innovative therapeutic approaches for stress-associated disorders by restoring immune homeostasis while maintaining behavioral integrity.
Collapse
Affiliation(s)
- Yi-Shu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 212696, P. R. China
| | - Hai-Chao Chen
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 212696, P. R. China
| | - Jia-Xin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 212696, P. R. China
| | - Si-Wei Zhou
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 212696, P. R. China
| | - Yue-Zhang Ma
- Department of Immunization Program, Shaanxi Provincial Center for Disease Control and Prevention, Xi'an, P. R. China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 212696, P. R. China
- Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, Lanzhou, Gansu, 730000, P. R. China
| |
Collapse
|
45
|
Jing H, Gao Y, Jing L, Yang H, Liu S. Recent advances in therapeutic use of transforming growth factor-beta inhibitors in cancer and fibrosis. Front Oncol 2025; 15:1489701. [PMID: 40352593 PMCID: PMC12061708 DOI: 10.3389/fonc.2025.1489701] [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: 09/01/2024] [Accepted: 04/03/2025] [Indexed: 05/14/2025] Open
Abstract
Transforming growth factor-beta (TGF-β) has long been known to be associated with early embryonic development and organogenesis, immune supervision, and tissue repair and homeostasis in adults. TGF-β has complex roles in fibrosis and cancer that may be opposing at different stages of these diseases. Under pathological conditions, overexpression of TGF-β causes epithelial-mesenchymal transition, deposition of extracellular matrix, and formation of cancer-associated fibroblasts, leading to fibrotic disease or cancer. Fibroblasts, epithelial cells, and immune cells are the most common targets of TGF-β, while fibrosis and cancer are the most common TGF-β-associated diseases. Given the critical role of TGF-β and its downstream molecules in fibrosis and progression of cancer, therapies targeting TGF-β signaling appear to be a promising strategy. Preclinical and clinical studies have investigated therapies targeting TGF-β, including antisense oligonucleotides, monoclonal antibodies, and ligand traps. However, development of targeted TGF-β therapy has been hindered by systemic cytotoxicity. This review discusses the molecular mechanisms of TGF-β signaling and highlights targeted TGF-β therapy for cancer and fibrosis as a therapeutic strategy for related diseases.
Collapse
Affiliation(s)
- Hanhui Jing
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Gao
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Linyuan Jing
- Department of Integrated Chinese and Western Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Hanyu Yang
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
46
|
Han J, Rotenberg D. Multi-omics analysis reveals discordant proteome and transcriptome responses in larval guts of Frankliniella occidentalis infected with an orthotospovirus. INSECT MOLECULAR BIOLOGY 2025. [PMID: 40279100 DOI: 10.1111/imb.12992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025]
Abstract
The western flower thrips, Frankliniella occidentalis, is the principal thrips vector of Orthotospovirus tomatomaculae (order Bunyavirales, family Tospoviridae), a devastating plant-pathogenic virus commonly referred to as tomato spotted wilt virus (TSWV). The larval gut is the gateway for virus transmission by F. occidentalis adults to plants. In a previous report, gut expression at the transcriptome level was subtle but significant in response to TSWV in L1s. Since it has been well documented that the relationship between the expression of mRNA and associated protein products in eukaryotic cells is often discordant, we performed identical, replicated experiments to identify and quantify virus-responsive larval gut proteins to expand our understanding of insect host response to TSWV. While we documented statistically significant, positive correlations between the abundance of proteins (4189 identified) and their cognate mRNAs expressed in first and second instar guts, there was virtually no alignment of individual genes identified to be differentially modulated by virus infection at the transcriptome and proteome levels. Predicted protein-protein interaction networks associated with clusters of co-expressed proteins revealed wide variation in correlation strength between protein and cognate transcript abundance, which appeared to be associated with the type of cellular processes, cellular compartments and network connectivity represented by the proteins. In total, our findings indicate distinct and dynamic regulatory mechanisms of transcript and protein abundance (expression, modifications and/or turnover) in virus-infected gut tissues. This study provides molecular candidates for future functional analysis of thrips vector competence and underscores the necessity of examining complex virus-vector interactions at a systems level.
Collapse
Affiliation(s)
- Jinlong Han
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, North Carolina, USA
| | - Dorith Rotenberg
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
47
|
Ma Y, Song B, Peng J, Wei F, Hao Y, Wen Y, Lv H, Shi X, Wang Y, Peng T. Cornua cervi degelatinatum inhibits breast cancer stem-like cell properties and metastasis via miR-148a-3p-mediated TGF-β/Smad2 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119709. [PMID: 40157402 DOI: 10.1016/j.jep.2025.119709] [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: 02/10/2025] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cornua cervi degelatinatum (CCD) is formed by removing the gelatinous substance from deer antlers according to traditional methods. It was first recorded in the Shennong's Classic of Materia Medica and has been included in the Pharmacopoeia of the People's Republic of China. It is commonly used in clinical practice for the treatment of diseases such as cancer and infertility. AIM OF THE STUDY This study aims to investigate the impact of CCD aqueous extract on the proliferation and stemness of breast cancer (BC) cells, with an emphasis on its regulation of miR-148a-3p expression and associated molecular pathways. MATERIALS AND METHODS Breast cancer cells were treated with various concentrations of CCD to assess its effects on cancer stem cell (CSC) features, epithelial-mesenchymal transition (EMT) markers, and overall plasticity. The UALCAN platform was utilized to analyze the relationship between miR-148a-3p and Smad2 expression. Functional experiments involving miR-148a-3p overexpression were performed to elucidate CCD's modulatory effects on the TGF-β/Smad2 pathway. Furthermore, molecular docking analysis was conducted to predict the binding affinity of CCD's active components to Smad2. RESULTS The CCD aqueous extract significantly reduced BC cell viability in vitro and dose-dependently suppressed the expression of stemness- and EMT-related proteins. Bioinformatics analysis and luciferase reporter assays validated miR-148a-3p as a direct regulator of Smad2, inhibiting the TGF-β/Smad2 signaling pathway. Molecular docking revealed strong binding interactions between CCD's active components and Smad2. CONCLUSIONS CCD exhibits anti-BC effects by working synergistically with miR-148a-3p to inhibit the TGF-β/Smad2 pathway, thereby reducing BC stemness and EMT progression. These findings provide valuable insights into the molecular mechanisms underlying CCD's therapeutic potential in BC treatment.
Collapse
Affiliation(s)
- Yanmiao Ma
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, 030619, China; Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Taiyuan, 030013, China; Shanxi Provincial Key Laboratory of Prescription Compatibility and Functions, Taiyuan, 030619, China.
| | - Bo Song
- College of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Jiehao Peng
- College of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Fuxia Wei
- College of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Yuanhui Hao
- College of Third Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Ya Wen
- College of First Clinical Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Huiling Lv
- Experimental Management Center, Shanxi University of Chinese Medicine, Taiyuan, 030619, China
| | - Xiaoli Shi
- Department of Rehabilitation, Shanxi Acupuncture Hospital, Taiyuan, 030006, China
| | - Yonghui Wang
- College of Basic Medical Sciences, Shanxi University of Chinese Medicine, Taiyuan, 030619, China; Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Taiyuan, 030013, China.
| | - Tao Peng
- Shanxi Hospital of Integrated Traditional Chinese and Western Medicine, Taiyuan, 030013, China; Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Taiyuan, 030013, China; Shanxi Provincial Key Laboratory of Prescription Compatibility and Functions, Taiyuan, 030619, China.
| |
Collapse
|
48
|
Guo Y, Deng F, Jiang Y, Cao G, Zhang Y, Liu G, Alimujiang M, Ayati M, Chen Y, Chen L, Lv S, Dou X. IL-37 Alleviates Sepsis-Induced Lung Injury by Inhibiting Inflammatory Response Through the TGF-β/Smad3 Pathway. Immunol Invest 2025:1-15. [PMID: 40270428 DOI: 10.1080/08820139.2025.2495958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Introduction: Sepsis is caused by an uncontrolled inflammatory response and immune dysfunction, with lung injury being the most common complication and one of the leading causes of death in clinically ill patients. Interleukin 37 (IL-37) is a multifunctional cytokine that plays a vital role in various pathophysiological processes, including inflammation, infection, and immunity.Methods: The study involved both clinical and animal experiments (establishing an animal model of sepsis-induced lung injury). Firstly, 50 patients with sepsis-induced lung injury and 50 healthy controls were included. In addition, a more in-depth study was conducted using animal models.Results: IL-37, IL-6, PCT, and CRP levels were significantly higher in the sepsis-induced lung injury group. Correlation analysis revealed that IL-37 significantly correlated with IL-6, PCT, and CRP levels. In animal experiments, IL-37 significantly attenuated CLP-induced pulmonary edema and cellular injury while reducing the levels of inflammatory factors IL-6 and TNF-α, as well as sepsis-related inflammatory markers PCT and CRP. Moreover, IL-37 significantly downregulated the expression levels of genes and proteins of apoptosis-related molecules Caspase-3 and Bax and pathway molecules TGF-β and Smad3. Discussion: The TGF-β/Smad3 pathway is involved in the process of IL-37 inhibiting inflammatory response and ameliorating sepsis-induced lung injury.
Collapse
Affiliation(s)
- Yufang Guo
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| | - Feifei Deng
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Yali Jiang
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- Ili & Jiangsu Joint Institute of Health, Yili, Xinjiang, P.R. China
| | - Guodong Cao
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
- Ili & Jiangsu Joint Institute of Health, Yili, Xinjiang, P.R. China
| | - Yixin Zhang
- Department of Critical Care Medicine, Medical School of Shihezi University, Shihezi, Xinjiang, P.R. China
| | - Gaowu Liu
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Mayinur Alimujiang
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Mairhaba Ayati
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Yufeng Chen
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Lili Chen
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Su Lv
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Xueqin Dou
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| |
Collapse
|
49
|
Shen F, Chen Y, Xu Z, Wang W, Chen G, Ye F. Inhibition of M2 macrophage-mediated mesenchymal stem cell migration: Boldine attenuates elbow heterotopic ossification. Int J Biochem Cell Biol 2025; 185:106787. [PMID: 40287052 DOI: 10.1016/j.biocel.2025.106787] [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: 01/22/2025] [Revised: 03/26/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Heterotopic ossification (HO) is characterized by abnormal bone formation in soft tissues, often following trauma or surgery. Transforming growth factor-beta (TGF-β) signaling and M2 macrophage polarization play critical roles in the recruitment and differentiation of mesenchymal stromal/progenitor cells (MSPCs), promoting HO. METHODS An elbow joint trauma-induced HO mouse model was established, where model mice were treated with dichloromethylene-bisphosphonate (Cl2MBP) liposomes or PBS liposomes to deplete macrophages. In addition, boldine was administered to evaluate its therapeutic effect on HO formation. Bone marrow mesenchymal stem cells (BMSCs) were also extracted for in vitro experiments. Quantitative real-time PCR (qRT-PCR) and Western blot were conducted to assess gene and protein expression. In vivo methods included Micro-Computed Tomography (Micro-CT) to assess bone formation, histological staining to evaluate tissue changes, immunohistochemistry (IHC) and immunofluorescence to analyze macrophage, CD73+ and CD105+ cells infiltration. In vitro, BMSCs were identified by flow cytometry and treated with interleukin-10 (IL-10) and/or boldine, and assays such as cell viability (Cell Counting Kit 8 (CCK8)), migration (Transwell), immunofluorescence, ALP staining, and Alizarin Red S staining, were conducted to assess osteogenic differentiation. RESULTS Boldine treatment significantly reduced HO formation, decreased collagen deposition, and inhibited M2 macrophage infiltration (P < 0.05). In vitro, boldine reduced IL-10-induced cell activity, migration, and osteogenic differentiation of BMSCs and inhibited TGF-β and pSmad2/3/Smad2/3 protein (P < 0.05). CONCLUSION Boldine attenuates HO by inhibiting M2 macrophage-mediated MSPC migration and might involve the TGF-β signaling, suggesting its potential as a therapeutic approach for managing HO.
Collapse
Affiliation(s)
- Fengteng Shen
- Department of Orthopedics, Tongxiang First People's Hospital, China
| | - Yansong Chen
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou City, 311200, China
| | - Zhikun Xu
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou City, 311200, China
| | - Wei Wang
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou City, 311200, China
| | - Guofang Chen
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou City, 311200, China
| | - Fusheng Ye
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou City, 311200, China.
| |
Collapse
|
50
|
Gaydarski L, Petrova K, Stanchev S, Pelinkov D, Iliev A, Dimitrova IN, Kirkov V, Landzhov B, Stamenov N. Morphometric and Molecular Interplay in Hypertension-Induced Cardiac Remodeling with an Emphasis on the Potential Therapeutic Implications. Int J Mol Sci 2025; 26:4022. [PMID: 40362262 PMCID: PMC12071960 DOI: 10.3390/ijms26094022] [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: 01/30/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 05/15/2025] Open
Abstract
Hypertension-induced cardiac remodeling is a complex process driven by interconnected molecular and cellular mechanisms that culminate in hypertensive myocardium, characterized by ventricular hypertrophy, fibrosis, impaired angiogenesis, and myocardial dysfunction. This review discusses the histomorphometric changes in capillary density, fibrosis, and mast cells in the hypertensive myocardium and delves into the roles of key regulatory systems, including the apelinergic system, vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) pathways, and nitric oxide (NO)/nitric oxide synthase (NOS) signaling in the pathogenesis of hypertensive heart disease (HHD). Capillary rarefaction, a hallmark of HHD, contributes to myocardial ischemia and fibrosis, underscoring the importance of maintaining vascular integrity. Targeting capillary density (CD) through antihypertensive therapy or angiogenic interventions could significantly improve cardiac outcomes. Myocardial fibrosis, mediated by excessive collagen deposition and influenced by fibroblast growth factor-2 (FGF-2) and transforming growth factor-beta (TGF-β), plays a pivotal role in the structural remodeling of hypertensive myocardium. While renin-angiotensin-aldosterone system (RAAS) inhibitors show anti-fibrotic effects, more targeted therapies are needed to address fibrosis directly. Mast cells, though less studied in humans, emerge as critical regulators of cardiac remodeling through their release of pro-fibrotic mediators such as histamine, tryptase, and FGF-2. The apelinergic system emerges as a promising therapeutic target due to its vasodilatory, anti-fibrotic, and cardioprotective properties. The system counteracts the deleterious effects of the RAAS and has demonstrated efficacy in preclinical models of hypertension-induced cardiac damage. Despite its potential, human studies on apelin analogs remain limited, warranting further exploration to evaluate their clinical utility. VEGF signaling plays a dual role, facilitating angiogenesis and compensatory remodeling during the early stages of arterial hypertension (AH) but contributing to maladaptive changes when dysregulated. Modulating VEGF signaling through exercise or pharmacological interventions has shown promise in improving CD and mitigating hypertensive cardiac damage. However, VEGF inhibitors, commonly used in oncology, can exacerbate AH and endothelial dysfunction, highlighting the need for therapeutic caution. The NO/NOS pathway is essential for vascular homeostasis and the prevention of oxidative stress. Dysregulation of this pathway, particularly endothelial NOS (eNOS) uncoupling and inducible NOS (iNOS) overexpression, leads to endothelial dysfunction and nitrosative stress in hypertensive myocardium. Strategies to restore NO bioavailability, such as tetrahydrobiopterin (BH4) supplementation and antioxidants, hold potential for therapeutic application but require further validation. Future studies should adopt a multidisciplinary approach to integrate molecular insights with clinical applications, paving the way for more personalized and effective treatments for HHD. Addressing these challenges will not only enhance the understanding of hypertensive myocardium but also improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Lyubomir Gaydarski
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Kristina Petrova
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Stancho Stanchev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Dimitar Pelinkov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Alexandar Iliev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Iva N. Dimitrova
- Department of Cardiology, University Hospital “St. Ekaterina”, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Vidin Kirkov
- Department of Health Policy and Management, Faculty of Public Health ‘Prof. Dr. Tzekomir Vodenicharov’, Medical University of Sofia, 1527 Sofia, Bulgaria;
| | - Boycho Landzhov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| | - Nikola Stamenov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (K.P.); (S.S.); (D.P.); (A.I.); (B.L.); (N.S.)
| |
Collapse
|