1
|
Malak MN, Arafa EA, Abdel-Fattah MM, Khalaf MM, Arab HH, Hamzawy MA. Targeting EGFR/PI3K/AKT/mTOR and Bax/Bcl-2/caspase3 pathways with ivermectin mediates its anticancer effects against urethane-induced non-small cell lung cancer in BALB/c mice. Tissue Cell 2025; 95:102873. [PMID: 40174264 DOI: 10.1016/j.tice.2025.102873] [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: 12/02/2024] [Revised: 02/12/2025] [Accepted: 03/15/2025] [Indexed: 04/04/2025]
Abstract
Lung cancer's mortality is among the highest compared to other cancers globally. However, a recent study has shown that ivermectin, an antiparasitic drug, may have a promising anticancer effect on lung cancer. The present study aimed to investigate the impact of ivermectin on EGFR.3/PI3K4/AKT5/mTOR6 signaling pathway in NSCLC.7 Mice were divided into four groups; (1) normal; (2) oral ivermectin alone (5 mg/kg) daily; (3) NSCLC was induced by urethane (1.5 g/kg, i.p.) at days one and sixty; (4) NSCLC group treated with ivermectin. The effect of ivermectin on macroscopic, microscopic, and lung index was assessed. The antitumor and antiproliferative effects of ivermectin were investigated by CYFRA 21-1 level and Ki-67, respectively. IHC determined the molecular expression of EGFR8, while phosphorylated PI3K, AKT, and mTOR were quantified by Western blotting assay. ELISA assay of active caspase 3, Bcl-29, and BAX10 was used to assess the apoptotic effect of ivermectin. Finally, VEGF11 lung content was measured. Findings showed that ivermectin improved macro and microscopic pathological changes. Ivermectin induced cytotoxic effect as indicated by CYFRA 21-1 suppression besides enhancing BAX/Bcl-2 ratio and active caspase 3. The immunoexpression of Ki-67 and EGFR declined. Ivermectin remarkably reduced p-PI3K, p-AKT, p-mTOR, and VEGF expressions. Overall, the study proposes ivermectin as a promising drug for lung cancer through its orchestral regulation of EGFR/PI3K/AKT/mTOR/VEGF signaling.
Collapse
Affiliation(s)
- Marina N Malak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Elshaimaa A Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates.
| | - Maha M Abdel-Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Marwa M Khalaf
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Hany H Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Mohamed A Hamzawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Fayoum University, Fayoum 63514, Egypt
| |
Collapse
|
2
|
Li J, Gu J, Pan S, Deng N, Khan M, Li L, Wu X, Li Y. Synergic effect of the combination of isoliquiritigenin and arsenic trioxide in HepG2 liver cancer cells. Cell Signal 2025; 131:111752. [PMID: 40107478 DOI: 10.1016/j.cellsig.2025.111752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/06/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
Despite continuous therapeutic interventions, the prognosis of hepatocellular carcinoma (HCC) remains very poor. Thus, quest for novel treatment strategies to improve therapeutic window of HCC therapy is paramount. Arsenic trioxide (ATO) is commonly used as the first-line treatment for acute promyelocytic leukemia (APL). Isoliquiritigenin (ISL) is a potential plant-based bioactive molecule with versatile biological and pharmacological effects including anticancer effect. The present study aimed to investigate the potential synergistic effects of combination of ISL and ATO in HCC cells. The data revealed that the combination of ISL and ATO synergistically inhibited HCC cell proliferation. The collective data demonstrate that synergistic anticancer effect of combined treatment of ISL + ATO was achieved via cooperative induction of mitochondrial apoptosis through ROS generation and inhibition of PI3K/Akt/mTOR pathway. In addition, ROS generation and suppression of PI3K/Akt/mTOR pathway were found to be two independent events in induction of apoptosis. Finally, we observed that combination treatment effectively suppressed tumor growth in nude mice xenograft model through induction of intrinsic apoptosis and inhibition of PI3K/Akt/mTOR pathway. In conclusion, the findings of this study suggest that both drugs work synergistically to exert anti-tumor effect in HCC, both in-vitro and in-vivo and could offer novel strategy for liver cancer treatment.
Collapse
Affiliation(s)
- Jingjing Li
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Juan Gu
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sijia Pan
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Nuo Deng
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Muhammad Khan
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan.
| | - Lingyan Li
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiao Wu
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yongming Li
- School of Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Lu N, Guo Y, Ren L, Zhao H, Yan L, Han H, Zhang S. CORO1C Regulates the Malignant Biological Behavior of Ovarian Cancer Cells and Modulates the mRNA Expression Profile through the PI3K/AKT Signaling Pathway. Cell Biochem Biophys 2025; 83:1819-1833. [PMID: 39433598 DOI: 10.1007/s12013-024-01591-4] [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] [Accepted: 10/03/2024] [Indexed: 10/23/2024]
Abstract
Ovarian cancer (OC) is a frequently occurring gynecological tumor, and its global incidence has recently increased. Coronin-like actin-binding protein 1C (CORO1C) is known to activate the phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT) pathway and promote tumor progression. However, its role in OC remains unclear. This study investigated the role of CORO1C in OC malignancy. In this study, quantitative real-time polymerase chain reaction (qRT-PCR) was used to examine AKT and CORO1C mRNA expression in clinical OC tissues and cells. Immunohistochemical analysis and western blotting were used to examine protein expression in OC tissues and cells, respectively. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), scratch wound-healing, and Transwell assays were performed to examine cell proliferation and migration. RNA-Seq was used to validate the relationship between AKT and CORO1C expression. The results showed that CORO1C was highly expressed in clinical OC tissues and SKOV3 cells, correlating with the International Federation of Gynecology and Obstetrics (FIGO) stage. Furthermore, CORO1C knockout inhibited the proliferation, migration, and invasion of SKOV3 cells; altered the gene expression patterns in these cells; and was closely associated with the PI3K/AKT pathway. Western blotting confirmed that CORO1C knockout reduced the levels of phosphorylated PI3K and AKT. Additionally, CORO1C knockout increased phosphatase and tensin homologs deleted on chromosome 10 (PTEN) protein expression, whereas CORO1C overexpression decreased it. In conclusion, this study demonstrated that high CORO1C levels in OC are associated with greater metastasis and worse prognosis. CORO1C negatively regulates PTEN expression, activates the PI3K/AKT pathway, and promotes OC cell malignancy In patients with OC, CORO1C may function as an effective therapeutic and predictive biomarker.
Collapse
Affiliation(s)
- Na Lu
- Gynecology and oncology department, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Yongfeng Guo
- Gynecology and oncology department, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Lixin Ren
- General surgery department, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Hongwei Zhao
- Gynecology and oncology department, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Lijun Yan
- Gynecology and oncology department, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Haiqiong Han
- Gynecology and oncology department, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Sanyuan Zhang
- Department of gynecology and obstetrics, The First Clinical Medical College of Shanxi Medical University, Taiyuan, 030000, China.
| |
Collapse
|
4
|
Liu L, Song L, Liu T, Hui K, Hu C, Yang J, Pi X, Yan Y, Liu S, Zhang Y, Chen H, Cao Y, Zhou L, Qiao Y, Yu D, Yin C, Li X, Zhang C, Li D, Wang Z, Liu Z, Jiang X. Recombinant oncolytic virus NDV-anti-VEGFR2 enhances radiotherapy sensitivity in NSCLC by targeting VEGF signaling and impairing DNA repair. Gene Ther 2025:10.1038/s41434-025-00540-x. [PMID: 40382521 DOI: 10.1038/s41434-025-00540-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 04/09/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Resistance to radiotherapy is a significant challenge in the clinical management of non-small cell lung cancer (NSCLC). This study investigates a novel multimodal therapeutic strategy that combines oncolytic Newcastle disease virus (NDV) with an anti-VEGFR2 single-chain variable fragment (NDV-anti-VEGFR2) to enhance radiosensitivity in NSCLC. We engineered NDV-anti-VEGFR2 and assessed its efficacy in sensitizing Calu-1 cells to radiation. In vitro results demonstrated that NDV-anti-VEGFR2 significantly inhibited tumor cell proliferation when combined with radiotherapy. In vivo experiments revealed that NDV-anti-VEGFR2, combined with radiation, achieved a tumor growth inhibition rate of 86.48%, surpassing the effects of NDV or radiation alone. Mechanistic investigations indicated that NDV-anti-VEGFR2 mitigated hypoxia by downregulating HIF-1α and impaired DNA repair pathways, as evidenced by reduced levels of RAD51 and γ-H2AX. These findings suggest that NDV-anti-VEGFR2 not only normalizes tumor vasculature but also enhances the cytotoxic effects of radiation by compromising DNA repair mechanisms. Collectively, our results support the clinical potential of NDV-anti-VEGFR2 combined with radiotherapy as a promising strategy to overcome radiotherapy resistance in NSCLC. Future studies in immunocompetent models are warranted to elucidate the immune-mediated effects of this innovative therapeutic approach.
Collapse
Affiliation(s)
- Liang Liu
- Lianyungang Clinical College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Liying Song
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Tianyan Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Kaiyuan Hui
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Chenxi Hu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Jiarui Yang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Xuelei Pi
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yuanyuan Yan
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Shishi Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yating Zhang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Hongna Chen
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Yukai Cao
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Lihua Zhou
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Yun Qiao
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China
| | - Dan Yu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Chengkai Yin
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Xu Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Chenfeng Zhang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Deshan Li
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China
| | - Zhihang Liu
- Jiangsu Kanion Pharmaceutical Co., Ltd., Jiangning Industrial City, Economic and Technological Development Zone, Lianyungang City, Jiangsu province, China.
| | - Xiaodong Jiang
- Lianyungang Clinical College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang City, Jiangsu Province, China.
| |
Collapse
|
5
|
Wang Y, Ohnuki H, Tran AD, Wang D, Ha T, Feng JX, Sim M, Barnhill R, Lugassy C, Sargen MR, Salazar-Cavazos E, Kruhlak M, Tosato G. Induced clustering of SHP2-depleted tumor cells in vascular islands restores sensitivity to MEK/ERK inhibition. J Clin Invest 2025; 135:e181609. [PMID: 40131370 PMCID: PMC12077907 DOI: 10.1172/jci181609] [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: 04/01/2024] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Allosteric inhibitors of the tyrosine phosphatase Src homology 2 domain-containing protein tyrosine phosphatase 2 (SHP2) hold therapeutic promise in cancers with overactive RAS/ERK signaling, but adaptive resistance to SHP2 inhibitors may limit benefits. Here, we utilized tumor cells that proliferate similarly with or without endogenous SHP2 to explore means to overcome this growth independence from SHP2. We found that SHP2 depletion profoundly altered the output of vascular regulators, cytokines, chemokines, and other factors from SHP2 growth-resistant cancer cells. Tumors derived from inoculation of SHP2-depleted, but SHP2 growth-independent, mouse melanoma and colon carcinoma cell lines displayed a typically subverted architecture, in which proliferative tumor cells surrounding a remodeled vessel formed "vascular islands", each limited by surrounding hypoxic and dead tumor tissue, where inflammatory blood cells were limited. Although vascular islands generally reflect protected sanctuaries for tumor cells, we found that vascular island-resident, highly proliferative, SHP2-depleted tumor cells acquired an increased sensitivity to blockage of MEK/ERK signaling, resulting in reduced tumor growth. Our results show that the response to targeted therapies in resistant tumor cells was controlled by tumor cell-induced vascular changes and tumor architectural reorganization, providing a compelling approach to elicit tumor responses by exploiting tumor- and endothelium-dependent biochemical changes.
Collapse
MESH Headings
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency
- Animals
- Mice
- MAP Kinase Signaling System/drug effects
- Humans
- Cell Line, Tumor
- Drug Resistance, Neoplasm
- Colonic Neoplasms/pathology
- Colonic Neoplasms/drug therapy
- Melanoma, Experimental/pathology
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/enzymology
- Melanoma, Experimental/genetics
- Cell Proliferation
- Neovascularization, Pathologic/enzymology
Collapse
Affiliation(s)
- Yuyi Wang
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Hidetaka Ohnuki
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Andy D. Tran
- Center for Cancer Research Microscopy Core, Laboratory of Cancer Biology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Dunrui Wang
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Taekyu Ha
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Jing-Xin Feng
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Minji Sim
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| | - Raymond Barnhill
- Department of Translational Research, Institut Curie, Paris, France
| | - Claire Lugassy
- Department of Translational Research, Institut Curie, Paris, France
| | - Michael R. Sargen
- Division of Cancer Epidemiology and Genetics, NCI, NIH, Rockville, Maryland, USA
| | - Emanuel Salazar-Cavazos
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, NIH, Bethesda, Maryland, USA
| | - Michael Kruhlak
- Center for Cancer Research Microscopy Core, Laboratory of Cancer Biology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, and
| |
Collapse
|
6
|
Wang BQ, Duan YY, Chen M, Ma YF, Chen R, Huang C, Gao F, Xu R, Duan CM. Endothelial Cell Integrin α6 Regulates Vascular Remodeling Through the PI3K/Akt-eNOS-VEGFA Axis After Stroke. Neurosci Bull 2025:10.1007/s12264-025-01403-6. [PMID: 40316875 DOI: 10.1007/s12264-025-01403-6] [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: 09/10/2024] [Accepted: 12/28/2024] [Indexed: 05/04/2025] Open
Abstract
The angiogenic response is essential for the repair of ischemic brain tissue. Integrin α6 (Itga6) expression has been shown to increase under hypoxic conditions and is expressed exclusively in vascular structures; however, its role in post-ischemic angiogenesis remains poorly understood. In this study, we demonstrate that mice with endothelial cell-specific knockout of Itga6 exhibit reduced neovascularization, reduced pericyte coverage on microvessels, and accelerated breakdown of microvascular integrity in the peri-infarct area. In vitro, endothelial cells with ITGA6 knockdown display reduced proliferation, migration, and tube-formation. Mechanistically, we demonstrated that ITGA6 regulates post-stroke angiogenesis through the PI3K/Akt-eNOS-VEGFA axis. Importantly, the specific overexpression of Itga6 in endothelial cells significantly enhanced neovascularization and enhanced the integrity of microvessels, leading to improved functional recovery. Our results suggest that endothelial cell Itga6 plays a crucial role in key steps of post-stroke angiogenesis, and may represent a promising therapeutic target for promoting recovery after stroke.
Collapse
Affiliation(s)
- Bing-Qiao Wang
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Yang-Ying Duan
- Department of Ultrasound Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Mao Chen
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Yu-Fan Ma
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Ru Chen
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Cheng Huang
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Fei Gao
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China
| | - Rui Xu
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China.
| | - Chun-Mei Duan
- Department of Neurology, Xinqiao Hospital, The Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
7
|
Lee D, Kim J, Baek S, Lee JW, Lee C, Kang KS, Shim SH. 1,3,5-Tricaffeoylquinic Acid from Ipomoea batatas Vines Induced Ovarian Cancer Cell Apoptosis and Inhibited Endothelial Tube Formation. Biomol Ther (Seoul) 2025; 33:483-493. [PMID: 40195283 PMCID: PMC12059361 DOI: 10.4062/biomolther.2024.239] [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: 12/11/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 04/09/2025] Open
Abstract
Ovarian cancer usually metastasizes from the ovary to adjacent organs through direct invasion with blood vessels formed by endothelial cells. Targeting apoptosis of ovarian cancer and angiogenesis is promising for anticancer therapy. Leaves of Ipomoea sp. have reportedly shown promise in treating ovarian cancer. Here, we investigated the apoptosis-inducing and anti-angiogenic effects of compounds isolated from Ipomoea batatas vines (IBV). Phytochemical examination of IBV led to the isolation and verification of eight compounds (1-8): chlorogenic acid (1), 3,4-dicaffeoylquinic acid (2), 3,5-dicaffeoylquinic acid (3), 4,5-dicaffeoylquinic acid (4), 1,3,5-tricaffeoylquinic acid (5), N-trans-feruloyltyramine (6), scopoletin (7), and esculetin (8). Of these, 1,3,5-tricaffeoylquinic acid (5) showed the highest cytotoxicity in A2780 human ovarian cancer cells, inducing apoptotic death in more than 37% cells and decreasing viability to less than 25% at 100 μM. Compound 5 increased the levels of cleaved caspase-8, Bax, cleaved PARP, and caspase-3/9, and decreased the levels of cleaved Bcl-2. Further, 5 inhibited tubule formation in HUVECs. VEGFR2, ERK, PI3K, Akt, and mTOR protein expression was also suppressed by 5. Then, a simple, rapid, and reliable LC-MS/ MS method was developed to determine the contents of the isolated compounds from IBV. Overall, 5 has potential for treating ovarian cancer as it induces apoptosis in ovarian cancer cells and inhibits tube formation.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Jaekyoung Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Soyoon Baek
- R&D Complex, Kolmar Korea, Seoul 06800, Republic of Korea
| | - Jin Woo Lee
- College of Pharmacy, Duksung Women’s University, Seoul 01369, Republic of Korea
| | - Changyeol Lee
- Herbal Medicine Resources Center, Korea Institute of Oriental Medicine, Naju 58245, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Sang Hee Shim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
8
|
Thomas WR, Richter T, O'Neil ET, Baldoni C, Corthals A, von Elverfeldt D, Nieland JD, Dechmann D, Hunter R, Davalos LM. Seasonal and comparative evidence of adaptive gene expression in mammalian brain size plasticity. eLife 2025; 13:RP100788. [PMID: 40310674 PMCID: PMC12045622 DOI: 10.7554/elife.100788] [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: 05/02/2025] Open
Abstract
Contrasting almost all other mammalian wintering strategies, Eurasian common shrews, Sorex araneus, endure winter by shrinking their brain, skull, and most organs, only to then regrow to breeding size the following spring. How such tiny mammals achieve this unique brain size plasticity while maintaining activity through the winter remains unknown. To discover potential adaptations underlying this trait, we analyzed seasonal differential gene expression in the shrew hypothalamus, a brain region that both regulates metabolic homeostasis and drastically changes size, and compared hypothalamus gene expression across species. We discovered seasonal variation in suites of genes involved in energy homeostasis and apoptosis, shrew-specific upregulation of genes involved in the development of the hypothalamic blood-brain barrier and calcium signaling, as well as overlapping seasonal and comparative gene expression divergence in genes implicated in the development and progression of human neurological and metabolic disorders, including CCDC22. With high metabolic rates and facing harsh winter conditions, S. araneus have evolved both adaptive and plastic mechanisms to sense and regulate their energy budget. Many of these changes mirrored those identified in human neurological and metabolic disease, highlighting the interactions between metabolic homeostasis, brain size plasticity, and longevity.
Collapse
Affiliation(s)
- William R Thomas
- Department of Ecology and Evolution, Stony Brook UniversityNew YorkUnited States
| | - Troy Richter
- Department of Psychology, Developmental and Brain Sciences Program, University of Massachusetts BostonBostonUnited States
| | - Erin T O'Neil
- Department of Psychology, Developmental and Brain Sciences Program, University of Massachusetts BostonBostonUnited States
| | - Cecilia Baldoni
- Max Planck Institute of Animal BehaviorRadolfzellGermany
- University of KonstanzRadolfzellGermany
| | | | - Dominik von Elverfeldt
- Division of Medical Physics, Department of Dignostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University FreiburgFreiburgGermany
| | - John D Nieland
- Health Science and Technology, Aalborg UniversityAalborgDenmark
| | - Dina Dechmann
- Max Planck Institute of Animal BehaviorRadolfzellGermany
- University of KonstanzRadolfzellGermany
| | - Richard Hunter
- Department of Psychology, Developmental and Brain Sciences Program, University of Massachusetts BostonBostonUnited States
| | - Liliana M Davalos
- Department of Ecology and Evolution, Stony Brook UniversityNew YorkUnited States
- Consortium for Inter-Disciplinary Environmental Research, Stony Brook UniversityNew YorkUnited States
| |
Collapse
|
9
|
Attique I, Haider Z, Khan M, Hassan S, Soliman MM, Ibrahim WN, Anjum S. Reactive Oxygen Species: From Tumorigenesis to Therapeutic Strategies in Cancer. Cancer Med 2025; 14:e70947. [PMID: 40377005 DOI: 10.1002/cam4.70947] [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: 11/26/2024] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Reactive oxygen species (ROS), a class of highly reactive molecules, are closely linked to the pathogenesis of various cancers. While ROS primarily originate from normal cellular processes, external stimuli can also contribute to their production. Cancer cells typically exhibit elevated ROS levels due to disrupted redox homeostasis, characterized by an imbalance between antioxidant and oxidant species. ROS play a dual role in cancer biology: at moderate levels, they facilitate tumor progression by regulating oncogenes and tumor suppressor genes, inducing mutations, promoting proliferation, extracellular matrix remodeling, invasion, immune modulation, and angiogenesis. However, excessive ROS levels can cause cellular damage and initiate apoptosis, necroptosis, or ferroptosis. METHODS This review explores molecular targets involved in redox homeostasis dysregulation and examines the impact of ROS on the tumor microenvironment (TME). Literature from recent in vitro and in vivo studies was analyzed to assess how ROS modulation contributes to cancer development and therapy. RESULTS Findings indicate that ROS influence cancer progression through various pathways and cellular mechanisms. Targeting ROS synthesis or enhancing ROS accumulation in tumor cells has shown promising anticancer effects. These therapeutic strategies exhibit significant potential to impair tumor growth while also interacting with elements of the TME. CONCLUSION The ROS serve as both promoters and suppressors of cancer depending on their intracellular concentration. Their complex role offers valuable opportunities for targeted cancer therapies. While challenges remain in precisely modulating ROS for therapeutic benefit, they hold promise as synergistic agents alongside conventional treatments, opening new avenues in cancer management.
Collapse
Affiliation(s)
- Iqra Attique
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| | - Zahra Haider
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| | - Maha Khan
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
| | - Samina Hassan
- Department of Botany, Kinnaird College for Women University, Lahore, Pakistan
| | - Mohamed Mohamed Soliman
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, Taif, Saudi Arabia
- Biochemistry Department, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Sumaira Anjum
- Department of Biotechnology, Kinnaird College for Women University, Lahore, Pakistan
| |
Collapse
|
10
|
Atarbashi-Moghadam F, Mahmoudian A, Taghipour N, Hakimiha N, Azadi A, Nokhbatolfoghahaei H. Enhancement of the angiogenic differentiation in the periodontal ligament stem cells using fibroblast growth factor 2 and photobiomodulation: An in vitro investigation. Photochem Photobiol 2025; 101:660-672. [PMID: 39435497 DOI: 10.1111/php.14032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/10/2024] [Accepted: 09/29/2024] [Indexed: 10/23/2024]
Abstract
This study aims to evaluate and compare the effect of fibroblastic growth factor 2 (FGF-2) and photobiomodulation, solely or in combination, in angiogenic differentiation of human periodontal ligament stem cells (hPDLSCs). The study comprises the following groups: control group (hPDLSCs only), FGF-2 (50 ng/mL) group, two photobiomodulation groups with a 4 J/cm2 energy density of 808 nm diode laser (1-Session or 2-Session), and two groups with the combination of each 1-Session or 2-Session photobiomodulation with FGF-2 (50 ng/mL). The 4',6-diamidino-2-phenylindole (DAPI) staining, and Methyl Thiazolyl Tetrazolium (MTT) assay were undertaken on days 2, 4, and 6. Quantitative Real-time Polymerase Chain Reaction (RT-qPCR) analysis on days 2, 4, 6, 8, and 11 was conducted to investigate VEGF-A and ANG-I genes. Coherently, the results of the DAPI and MTT showed the Laser (2-Session) group had higher cell viability than others on day 6. All groups demonstrated a growth pattern in the expression of VEGF-A and ANG-I from day 2 to 8 and, afterward, a significant downgrowth to day 11 (p < 0.05). The most amounts of expression of VEGF-A and ANG-I on day 8 were seen in the Laser (2-Session) group. Two-time application of photobiomodulation using a diode laser with 808 nm wavelength after 2 and 4 days of cell seeding can be associated with higher cell viability and angiogenic differentiation of hPDLSCs compared to the one-time application of photobiomodulation and administration of FGF-2.
Collapse
Affiliation(s)
- Fazele Atarbashi-Moghadam
- Department of Periodontics, Dental School of Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhosein Mahmoudian
- Department of Periodontics, Dental School of Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niloofar Taghipour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Hakimiha
- Laser Application in Medical Sciences Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Azadi
- Dentofacial Deformities Research Center, Research Institute for Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hanieh Nokhbatolfoghahaei
- Dental Research Center, Research Institute for Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Zhang J, Guo J, Qian Y, Yu L, Ma J, Gu B, Tang W, Li Y, Li H, Wu W. Quercetin Induces Apoptosis Through Downregulating P4HA2 and Inhibiting the PI3K/Akt/mTOR Axis in Hepatocellular Carcinoma Cells: An In Vitro Study. Cancer Rep (Hoboken) 2025; 8:e70220. [PMID: 40347062 PMCID: PMC12065022 DOI: 10.1002/cnr2.70220] [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: 09/28/2024] [Revised: 03/25/2025] [Accepted: 04/22/2025] [Indexed: 05/12/2025] Open
Abstract
BACKGROUND Quercetin is a natural product with multiple activities, which possesses a promising antitumor effect on malignancies. The involvement of proline 4-hydroxylase II (P4HA2) in collagen synthesis is crucial in the growth of tumor cells. Apoptosis is a programmed cell death requisite for the stability of the intracellular environment. However, the relationship between quercetin and cell apoptosis, as well as the impact of P4HA2 in this connection, has not yet been specified in hepatocellular carcinoma(HCC). AIMS The present study used HCC cells to investigate how quercetin regulates P4HA2 and influences cell proliferation and apoptosis. METHODS AND RESULTS The outcomes reveal that quercetin can impede the viability and growth of HCC cells and generate cell apoptosis in a dose-dependent manner. Additionally, quercetin prompts downregulation of P4HA2, leading to cell apoptosis in HCC cells, and knocking down P4HA2 can enhance this effect. Furthermore, we pretreated HCC cells with inhibitors (Z-VAD-FMK, LY294002) or activators (740Y-P) and found that the PI3K/Akt/mTOR pathway was occupied with quercetin-induced cell apoptosis. CONCLUSION This investigation reveals that quercetin compels apoptosis in HCC cells by diminishing P4HA2 and restraining the PI3K/Akt/mTOR axis.
Collapse
Affiliation(s)
- Junli Zhang
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical UniversityBengbuChina
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and TreatmentBengbuChina
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
| | - Jiayi Guo
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
| | - Ying Qian
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
| | - Lianchen Yu
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
| | - Junrao Ma
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
| | - Biao Gu
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical UniversityBengbuChina
| | - Weichun Tang
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical UniversityBengbuChina
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and TreatmentBengbuChina
| | - Yi Li
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical UniversityBengbuChina
| | - Hongwei Li
- The Third People's Hospital of Bengbu Affiliated to Bengbu Medical UniversityBengbuChina
| | - Wenjuan Wu
- Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory DiagnosisBengbu Medical UniversityBengbuChina
- Department of Biochemistry and Molecular BiologySchool of Laboratory Medicine, Bengbu Medical UniversityBengbuChina
| |
Collapse
|
12
|
Boff MO, Xavier FAC, Diz FM, Gonçalves JB, Ferreira LM, Zambeli J, Pazzin DB, Previato TTR, Erwig HS, Gonçalves JIB, Bruzzo FTK, Marinowic D, da Costa JC, Zanirati G. mTORopathies in Epilepsy and Neurodevelopmental Disorders: The Future of Therapeutics and the Role of Gene Editing. Cells 2025; 14:662. [PMID: 40358185 PMCID: PMC12071303 DOI: 10.3390/cells14090662] [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: 01/20/2025] [Revised: 02/06/2025] [Accepted: 02/06/2025] [Indexed: 05/15/2025] Open
Abstract
mTORopathies represent a group of neurodevelopmental disorders linked to dysregulated mTOR signaling, resulting in conditions such as tuberous sclerosis complex, focal cortical dysplasia, hemimegalencephaly, and Smith-Kingsmore Syndrome. These disorders often manifest with epilepsy, cognitive impairments, and, in some cases, structural brain anomalies. The mTOR pathway, a central regulator of cell growth and metabolism, plays a crucial role in brain development, where its hyperactivation leads to abnormal neuroplasticity, tumor formation, and heightened neuronal excitability. Current treatments primarily rely on mTOR inhibitors, such as rapamycin, which reduce seizure frequency and tumor size but fail to address underlying genetic causes. Advances in gene editing, particularly via CRISPR/Cas9, offer promising avenues for precision therapies targeting the genetic mutations driving mTORopathies. New delivery systems, including viral and non-viral vectors, aim to enhance the specificity and efficacy of these therapies, potentially transforming the management of these disorders. While gene editing holds curative potential, challenges remain concerning delivery, long-term safety, and ethical considerations. Continued research into mTOR mechanisms and innovative gene therapies may pave the way for transformative, personalized treatments for patients affected by these complex neurodevelopmental conditions.
Collapse
Affiliation(s)
- Marina Ottmann Boff
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Fernando Antônio Costa Xavier
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Medicine and Health Sciences, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Fernando Mendonça Diz
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Júlia Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Laura Meireles Ferreira
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Jean Zambeli
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Medicine, University of the Valley of the Rio dos Sinos (UNISINOS), São Leopoldo 93022-750, RS, Brazil
| | - Douglas Bottega Pazzin
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Thales Thor Ramos Previato
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Helena Scartassini Erwig
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Health and Life, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Fernanda Thays Konat Bruzzo
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Daniel Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
- School of Health and Life, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, RS, Brazil
| | - Jaderson Costa da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| | - Gabriele Zanirati
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (M.O.B.); (F.A.C.X.); (F.M.D.); (J.B.G.); (L.M.F.); (J.Z.); (D.B.P.); (T.T.R.P.); (H.S.E.); (J.I.B.G.); (F.T.K.B.); (D.M.); (J.C.d.C.)
| |
Collapse
|
13
|
Yang Y, Wu G, Wang Y, Mao Q, Zhang D, Wu J. LL37 promotes angiogenesis: a potential therapeutic strategy for lower limb ischemic diseases. Front Pharmacol 2025; 16:1587351. [PMID: 40337519 PMCID: PMC12055537 DOI: 10.3389/fphar.2025.1587351] [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: 03/04/2025] [Accepted: 04/08/2025] [Indexed: 05/09/2025] Open
Abstract
Purpose To study the angiogenic capacity of antimicrobial peptide LL37 (cathelicidin antimicrobial peptide), explore its molecular mechanisms, and provide new ideas for treating lower limb ischemic diseases. Methods LL37 was applied exogenously to human umbilical vein endothelial cells (HUVECs), and its effects on cell proliferation, migration, and angiogenesis were assessed using Cell Counting Kit-8 (CCK-8), plate cloning, scratch, and angiogenesis assays. A mouse lower limb ischemia model was established, with LL37 injected intramuscularly on days 0, 4, and 8. Blood flow recovery was evaluated by laser Doppler flowmetry. Immunofluorescence staining detected cluster of differentiation 31 (CD31) and cluster of differentiation 34 (CD34) expression, while Hematoxylin and Eosin (H&E) staining assessed muscle cell morphology. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting analyzed gene and protein expression changes in HUVECs. Results LL37 enhanced the proliferative, migratory, and pro-angiogenic abilities of HUVECs. It significantly improved blood flow recovery in ischemic limbs, with higher CD31/CD34 expression and more intact muscle morphology. qRT-PCR analysis demonstrated elevated expression of angiogenesis-related genes in LL37-treated HUVECs. Western blotting revealed increased vascular endothelial growth factor A (VEGFA) expression and enhanced phosphorylation levels of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway in LL37-treated cells. Conclusion LL37 promotes angiogenesis via the VEGFA-PI3K/AKT/mTOR pathway, showing potential for treating lower limb ischemia by improving perfusion.
Collapse
Affiliation(s)
| | | | | | | | | | - Jitao Wu
- *Correspondence: Dongxu Zhang, ; Jitao Wu,
| |
Collapse
|
14
|
Haratake T, Nishimura MF, Nishikori A, Gonzalez MV, Ennishi D, Lai YC, Ochi S, Tsunoda M, Fajgenbaum DC, van Rhee F, Momose S, Sato Y. The Involvement of PI3K-Akt Signaling in the Clinical and Pathological Findings of Idiopathic Multicentric Castleman Disease-Thrombocytopenia, Anasarca, Fever, Reticulin Fibrosis, and Organomegaly and Not Otherwise Specified Subtypes. Mod Pathol 2025; 38:100782. [PMID: 40274066 DOI: 10.1016/j.modpat.2025.100782] [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: 01/30/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
Idiopathic multicentric Castleman disease is a rare lymphoproliferative disorder that is clinically classified into idiopathic plasmacytic lymphadenopathy (IPL); thrombocytopenia, anasarca, fever, reticulin fibrosis, and organomegaly (TAFRO); and not otherwise specified (NOS). Although each subtype shows varying degrees of hypervascularity, no statistical data on the degree of vascularization have been reported. Additionally, the mechanisms underlying vascularization in each clinical subtype are poorly understood. Here, we aimed to clarify these mechanisms by evaluating the histopathological characteristics of each clinical subtype across 37 patients and performing a whole-transcriptome analysis focusing on angiogenesis-related gene expression. Histologically, TAFRO and NOS exhibited a significantly higher degree of vascularization than IPL (IPL vs TAFRO, P < .001; IPL vs NOS, P = .002). In addition, the germinal centers (GCs) were significantly more atrophic in TAFRO than in IPL. In TAFRO and NOS, "whirlpool vessels" in GCs were seen in most cases (TAFRO, 9/9, 100%; NOS, 6/8, 75%) but not in IPL (IPL vs TAFRO, P < .001; IPL vs NOS, P = .007). Likewise, immunostaining for Ets-related gene revealed higher levels in endothelial cells of GCs in TAFRO than in IPL (P = .014), and TAFRO and NOS were associated with a significantly higher number of endothelial cells in interfollicular areas compared with that in IPL (TAFRO vs IPL, P < .001; NOS vs IPL, P = .002). Gene expression analysis revealed that the PI3K-Akt signaling pathway was significantly enriched in the TAFRO and NOS (TAFRO/NOS) groups. This pathway, which may be activated by vascular endothelial growth factor A and some integrins, is known to affect angiogenesis by increasing vascular permeability, which may explain the clinical manifestations of anasarca and/or fluid retention in TAFRO/NOS. These results suggest that the PI3K-Akt pathway plays an important role in the pathogenesis of TAFRO/NOS.
Collapse
Affiliation(s)
- Tomoka Haratake
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, Okayama, Japan
| | - Midori Filiz Nishimura
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, Okayama, Japan
| | - Asami Nishikori
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, Okayama, Japan
| | - Michael V Gonzalez
- Center for Cytokine Storm Treatment and Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan; Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - You Cheng Lai
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
| | - Sayaka Ochi
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, Okayama, Japan
| | - Manaka Tsunoda
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, Okayama, Japan
| | - David C Fajgenbaum
- Center for Cytokine Storm Treatment and Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Castleman Disease Collaborative Network, Philadelphia, Pennsylvania
| | - Frits van Rhee
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Shuji Momose
- Department of Pathology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Yasuharu Sato
- Department of Molecular Hematopathology, Okayama University Graduate School of Health Sciences, Okayama, Japan.
| |
Collapse
|
15
|
Indelicato S, Bongiorno D, Mauro M, Cascioferro S. Recent Developments of 1,3,4-Thiadiazole Compounds as Anticancer Agents. Pharmaceuticals (Basel) 2025; 18:580. [PMID: 40284015 PMCID: PMC12030488 DOI: 10.3390/ph18040580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
The World Health Organization has recently underlined the increasing global burden of cancer, with a particularly alarming impact on underserved populations. In recent years, 1,3,4-thiadiazole has emerged as a versatile pharmacophore to obtain bioactive compounds. The pharmacological properties of this ring are primarily attributed to its role as a bioisostere of pyrimidine, the core structure of three nucleic bases. This structural feature endows 1,3,4-thiadiazole derivatives with the ability to interfere with DNA replication processes. Additionally, the mesoionic behavior of this heterocycle gives it important properties, such as the ability to cross biological membranes and interact with target proteins. Noteworthy, in analogy to the other sulfur heterocycles, the presence of C-S σ* orbitals, conferring small regions of low electron density on the sulfur atom, makes interaction with the target easier. This review focuses on the most promising anticancer agents with 1,3,4-thiadiazole structure reported in the past five years, providing information that may be useful to medicinal chemists who intend to develop new anticancer derivatives.
Collapse
Affiliation(s)
| | | | | | - Stella Cascioferro
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy; (S.I.); (D.B.); (M.M.)
| |
Collapse
|
16
|
Quelhas P, Morgado D, dos Santos J. Primary Cilia, Hypoxia, and Liver Dysfunction: A New Perspective on Biliary Atresia. Cells 2025; 14:596. [PMID: 40277920 PMCID: PMC12026149 DOI: 10.3390/cells14080596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
Ciliopathies are disorders that affect primary or secondary cellular cilia or structures associated with ciliary function. Primary cilia (PC) are essential for metabolic regulation and embryonic development, and pathogenic variants in cilia-related genes are linked to several pediatric conditions, including renal-hepatic diseases and congenital defects. Biliary atresia (BA) is a progressive infantile cholangiopathy and the leading cause of pediatric liver transplantation. Although the exact etiology of BA remains unclear, evidence suggests a multifactorial pathogenesis influenced by both genetic and environmental factors. Patients with BA and laterality defects exhibit genetic variants associated with ciliopathies. Interestingly, even isolated BA without extrahepatic anomalies presents morphological and functional ciliary abnormalities, suggesting that environmental triggers may disrupt the ciliary function. Among these factors, hypoxia has emerged as a potential modulator of this dysfunction. Hypoxia-inducible factor 1-alpha (HIF-1α) plays a central role in hepatic responses to oxygen deprivation, influencing bile duct remodeling and fibrosis, which are key processes in BA progression. This review explores the crosstalk between hypoxia and hepatic ciliopathies, with a focus on BA. It discusses the molecular mechanisms through which hypoxia may drive disease progression and examines the therapeutic potential of targeting hypoxia-related pathways. Understanding how oxygen deprivation influences ciliary function may open new avenues for treating biliary ciliopathies and improving patient outcomes.
Collapse
Affiliation(s)
| | | | - Jorge dos Santos
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (P.Q.); (D.M.)
| |
Collapse
|
17
|
Zhang Y, Yang H, Chen Y, Tang Y, Chen J, Huang J, Feng A, Weng Z, Li F, Lin J, Xie J, Zhang C, Chen J, Gao C, Nie X. Construction and diagnostic efficacy assessment of the urinary exosomal miRNA-mRNA network in children with IgA vasculitis nephritis. FASEB J 2025; 39:e70492. [PMID: 40166907 PMCID: PMC11959522 DOI: 10.1096/fj.202403111r] [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: 12/05/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
This study aimed to comprehensively evaluate the diagnostic potential of urinary exosomal microRNA (miRNA) in IgA vasculitis (IgAV) kidney injury by meticulously comparing the miRNA expression profiles in urine exosomes between children diagnosed with IgAV and those with IgA vasculitis nephritis (IgAVN). Urine samples were obtained from children with IgAV who were treated at our hospital from October 2022 to October 2023. These samples were then categorized into the IgAV group and the IgAVN group. High-throughput sequencing and bioinformatics analysis techniques were employed to conduct a thorough analysis of the differentially expressed miRNAs between the two groups. Additionally, the correlation between urinary exosomal miRNA and clinical parameters was evaluated. A total of 57 urinary exosomal miRNAs exhibited differential expression between the IgAV and IgAVN groups. Specifically, in the IgAVN group, 42 miRNAs were upregulated, while 15 were downregulated. Lasso regression analysis and ROC analysis identified five candidate urinary exosomal miRNAs with high diagnostic accuracy. A prediction of 95 target genes related to the candidate miRNAs led to the construction of an exosomal miRNA-mRNA regulatory network consisting of four key miRNAs and ten hub genes. Gene function and metabolic pathway analyses indicated that these ten hub genes were predominantly enriched in pro-fibrotic and inflammatory pathways. The analysis incorporating clinical parameters demonstrated a significant correlation between hsa-miR-383-5p and urinary protein levels. This research identified exosomal miRNAs and mRNAs with differential expression patterns associated with IgAVN and constructed the corresponding exosomal miRNA-mRNA network. It was determined that hsa-miR-3065-5p, hsa-miR-383-5p, hsa-miR-25-3p, and hsa-miR-450b-5p might mediate the pathogenesis of IgAVN by targeting pro-fibrotic and inflammatory pathways. Among them, exosomal hsa-miR-383-5p is highly likely to serve as a novel non-invasive biomarker for assessing the disease status of IgAVN, thereby offering new perspectives on the non-invasive diagnosis and treatment of IgAVN.
Collapse
Affiliation(s)
- Yunfan Zhang
- Department of PediatricsFuzong Clinical Medical College of Fujian Medical UniversityFuzhouChina
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Huanhuan Yang
- Department of PediatricsFuzong Clinical Medical College of Fujian Medical UniversityFuzhouChina
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Yi Chen
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Yuxian Tang
- Department of PediatricsFuzong Clinical Medical College of Fujian Medical UniversityFuzhouChina
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Junyan Chen
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Jun Huang
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Ai Feng
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Zengfeng Weng
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Fenrong Li
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Jinfeng Lin
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Jingqi Xie
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Chunfang Zhang
- Department of Pediatrics900th Hospital of PLA Joint Logistic Support ForceFuzhouChina
| | - Jie Chen
- Department of PediatricsFujian Provincial HospitalFuzhouChina
| | - Chunlin Gao
- Department of PediatricsJinling Hospital, Medical School of Nanjing UniversityNanjingChina
| | - Xiaojing Nie
- Department of PediatricsFuzong Clinical Medical College of Fujian Medical UniversityFuzhouChina
- Department of PediatricsFujian Provincial HospitalFuzhouChina
- Department of PediatricsDongfang Hospital of Xiamen University, School of Medical, Xiamen UniversityFuzhouChina
| |
Collapse
|
18
|
Brunmaier LAE, Ozdemir T, Walker TW. Angiogenesis: Biological Mechanisms and In Vitro Models. Ann Biomed Eng 2025:10.1007/s10439-025-03721-2. [PMID: 40210793 DOI: 10.1007/s10439-025-03721-2] [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: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
The translation of biomedical devices and drug research is an expensive and long process with a low probability of receiving FDA approval. Developing physiologically relevant in vitro models with human cells offers a solution to not only improving the odds of FDA approval but also to expand our ability to study complex in vivo systems in a simpler fashion. Animal models remain the standard for pre-clinical testing; however, the data from animal models is an unreliable extrapolation when anticipating a human response in clinical trials, thus contributing to the low rates of translation. In this review, we focus on in vitro vascular or angiogenic models because of the incremental role that the vascular system plays in the translation of biomedical research. The first section of this review discusses the most common angiogenic cytokines that are used in vitro to initiate angiogenesis, followed by angiogenic inhibitors where both initiators and inhibitors work to maintain vascular homeostasis. Next, we evaluate previously published in vitro models, where we evaluate capturing the physical environment for biomimetic in vitro modeling. These topics provide a foundation of parameters that must be considered to improve and achieve vascular biomimicry. Finally, we summarize these topics to suggest a path forward with the goal of engineering human in vitro models that emulate the in vivo environment and provide a platform for biomedical device and drug screening that produces data to support clinical translation.
Collapse
Affiliation(s)
- Laura A E Brunmaier
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Travis W Walker
- Karen M. Swindler Department of Chemical and Biological Engineering, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA.
| |
Collapse
|
19
|
Tang X, Ma C, Ren Y, Lv Y, He Y, Han L, Wu J. Revealing the Potential of Solamargine for Anti Metastasis and Angiogenesis Inhibition in Nasopharyngeal Carcinoma. J Inflamm Res 2025; 18:4879-4898. [PMID: 40224395 PMCID: PMC11992990 DOI: 10.2147/jir.s485244] [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: 10/24/2024] [Accepted: 02/06/2025] [Indexed: 04/15/2025] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is a major global health issue, especially in Southeast Asia. Solamargine (SM), an alkaloid from natural plants, inhibits various cancer cells. This study evaluates SM's effects on invasion, migration, EMT markers, angiogenesis, and related pathways in the NPC-specific C666-1 cell line. Methods In vitro assays, including wound healing, Transwell invasion, Western blot, and tube formation, were used to assess SM's impact on C666-1 NPC and HUVEC cells. SM concentrations were 2 µM and 5 µM, with axitinib (4 µM) as the control. Network pharmacology and GO-KEGG enrichment analyses were conducted to explore SM's targets and mechanisms in NPC. Results SM significantly inhibited C666-1 NPC cell invasion and migration by reducing EMT markers Vimentin and Snail. In HUVEC cells, SM decreased viability, invasion, migration, and tube formation, likely through VEGF signaling inactivation, EZH2 inhibition, and miR-203a-3p upregulation. Network pharmacology and GO-KEGG analyses identified key targets and pathways, suggesting SM's anti-NPC effects through multiple mechanisms. Discussion SM inhibits NPC cell invasion and migration by regulating EMT, suppressing angiogenesis, and modulating key pathways. These findings highlight SM's potential as an anti-cancer agent for NPC and provide new insights into its mechanisms. Network pharmacology and GO-KEGG analysis further identify its therapeutic targets, offering valuable directions for future drug development.
Collapse
Affiliation(s)
- Xiaojuan Tang
- Central Laboratory, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| | - Changju Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| | - Yuan Ren
- Central Laboratory, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
| | - Yuan Lv
- Central Laboratory, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
| | - Yongheng He
- Department of Anorectal Surgery, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
| | - Ling Han
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| | - Jingjing Wu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| |
Collapse
|
20
|
Yang N, Hua R, Lai Y, Zhu P, Ding J, Ma X, Yu G, Xia Y, Liang C, Gao W, Wang Z, Zhang H, Yang L, Zhou K, Ge L. Microenvironment-adaptive nanomedicine MXene promotes flap survival by inhibiting ROS cascade and endothelial pyroptosis. J Nanobiotechnology 2025; 23:282. [PMID: 40197477 PMCID: PMC11978011 DOI: 10.1186/s12951-025-03343-9] [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: 12/27/2024] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
In the field of large-area trauma flap transplantation, preventing avascular necrosis remains a critical challenge. Key mechanisms for improving flap viability include angiogenesis promotion, oxidative stress inhibition, and cell death prevention. Recently, two-dimensional ultrathin Ti3C2TX (MXene) nanosheets have gained attention for their potential contributions to these processes, though MXene's physiological impact on flap survival had not been previously investigated. This study is the first to confirm MXene's biological effects on the ischaemic microenvironment post-skin flap transplantation. Findings indicated that MXene significantly decreased the necrotic area in ischaemic flaps (37.96% ± 2.00%), with reductions of 30.40% ± 1.86% at 1 mg/mL and 20.19% ± 2.11% at 2 mg/mL in a concentration-dependent manner. Mechanistically, MXene facilitated in situ angiogenesis, mitigated oxidative stress, suppressed pro-inflammatory pyroptosis, and activated the PI3K-Akt pathway, particularly influencing vascular endothelial cells. Comparative transcriptome analysis of skin tissues with and without MXene treatment provided additional evidence, highlighting mechanisms such as pro-inflammatory pyroptosis, ROS metabolic processes, endothelial cell proliferation regulation, and PI3K-Akt signaling pathway activation. Overall, MXene demonstrated biological activity, effectively promoting ischaemic flaps survival and presenting a novel strategy for addressing ischaemic necrosis in skin flaps.
Collapse
Affiliation(s)
- Ningning Yang
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, 325027, Zhejiang, China
- State Key Laboratory of Macromolecular Drugs and Large-Scale Preparation, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Rongrong Hua
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yingying Lai
- Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, 325027, Zhejiang, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Peijun Zhu
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jian Ding
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China
- Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, 325027, Zhejiang, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Xianhui Ma
- Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, 325027, Zhejiang, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Gaoxiang Yu
- Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, 325027, Zhejiang, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Yiheng Xia
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China
| | - Chao Liang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China
| | - Weiyang Gao
- Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, 325027, Zhejiang, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Zhouguang Wang
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- State Key Laboratory of Macromolecular Drugs and Large-Scale Preparation, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China.
| | - Liangliang Yang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China.
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China.
| | - Kailiang Zhou
- Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, 325027, Zhejiang, China.
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China.
| | - Lu Ge
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China.
- School of Pharmaceutical Sciences, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, Zhejiang, China.
| |
Collapse
|
21
|
Shafeeque CM, Harmanci AO, Thomas S, Dienel AC, McBride DW, Peeyush KT, Blackburn SL. Gene expression changes in human cerebral arteries following hemoglobin exposure: implications for vascular responses in SAH. Front Physiol 2025; 16:1529113. [PMID: 40247928 PMCID: PMC12003393 DOI: 10.3389/fphys.2025.1529113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Subarachnoid hemorrhage (SAH), characterized by the presence of hemoglobin (Hb) in the subarachnoid space, significantly impacts cerebral vessels, leading to various pathological outcomes. The toxicity of cell-free Hb released from erythrocytes and its metabolites after SAH causes vasoconstriction and neuronal damage, and correlates with delayed ischemic neurological deficits (DIND). While animal models have provided substantial and invaluable data in the research of aneurysmal SAH, the specific effects of subarachnoid blood on cerebral arteries remain greatly understudied. Here, we describe the changes in the genetic profile of human cerebral arteries exposed to free Hb for 48 h. We performed an ex vivo exposure, followed by mRNA sequencing of the vessels. Compared to controls 54 genes were downregulated, and 53 genes were upregulated in human cerebral arteries after Hb exposure. Enrichment analysis identified the ferroptosis pathway as the most significantly affected. Further lipid peroxidation (LPO) assays and elevated ACSL4 gene expression support a ferroptosis pathway. Additionally, Hb exposure altered key signaling pathways essential for vascular stability (PI3K-Akt, MAPK), modified G-protein signaling mediated by RGS1/2, and suppressed key transcription factors such as KLF5, NR4A1, and FOS. Our results underscore the critical role of Hb in driving pathological responses in brain vessels. Furthermore, our dataset could be valuable for developing interventions after SAH and may help identify the underlying causes of vascular injury.
Collapse
Affiliation(s)
- Chathathayil M. Shafeeque
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Arif O. Harmanci
- D. Bradley McWilliams School of Biomedical Informatics, University of Texas Health Science Center, Fannin, TX, United States
| | - Sithara Thomas
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Ari C. Dienel
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Devin W. McBride
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Kumar T. Peeyush
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Spiros L. Blackburn
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
22
|
Gao J, Lu X, Wang G, Huang T, Tuo Z, Meng W. APOC1 knockdown induces apoptosis and decreases angiogenesis in diffuse large B-cell lymphoma cells through blocking the PI3K/AKT/mTOR pathway. BIOMOLECULES & BIOMEDICINE 2025; 25:1205-1217. [PMID: 39873475 PMCID: PMC11984368 DOI: 10.17305/bb.2024.11550] [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: 10/28/2024] [Revised: 01/11/2024] [Accepted: 01/11/2025] [Indexed: 01/30/2025]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a highly heterogeneous metastatic lymphoma that can be treated by targeting angiogenesis. Apolipoprotein C1 (APOC1) plays a significant role in the proliferation and metastasis of various malignant tumors; however, its role in DLBCL-particularly its effects on angiogenesis-remains largely unexplored. This study investigates the correlation between APOC1 expression and patient prognosis in DLBCL. Using APOC1 gene knockdown, apoptosis, migration, and invasion were assessed through flow cytometry, the EDU assay, wound healing, and Transwell assays. Additionally, human umbilical vein endothelial cells (HUVEC) angiogenesis was evaluated. Advanced techniques, such as immunofluorescence, TUNEL assay, and immunohistochemical labeling were employed to analyze the effects of APOC1 knockdown on the PI3K/AKT/mTOR signaling pathway and tumor formation in nude mice. Results showed that APOC1 is overexpressed in DLBCL tissues and cells, with high APOC1 levels associated with poor patient prognosis. In vitro experiments revealed that APOC1 knockdown increased apoptosis and inhibited cell proliferation, migration, invasion, HUVEC angiogenesis, and PI3K/AKT/mTOR signaling pathway protein expression in DLBCL cells. Similarly, in vivo studies demonstrated that APOC1 knockdown significantly reduced tumor growth, angiogenesis-related proteins, and phosphorylated PI3K/AKT/mTOR pathway proteins in nude mice. APOC1 knockdown promotes apoptosis and suppresses angiogenesis in DLBCL cells by inhibiting the PI3K/AKT/mTOR pathway.
Collapse
MESH Headings
- Humans
- TOR Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Apoptosis/genetics
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Animals
- Signal Transduction
- Phosphatidylinositol 3-Kinases/metabolism
- Mice
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Apolipoprotein C-I/genetics
- Apolipoprotein C-I/metabolism
- Gene Knockdown Techniques
- Cell Proliferation/genetics
- Cell Line, Tumor
- Mice, Nude
- Human Umbilical Vein Endothelial Cells
- Male
- Female
- Cell Movement/genetics
- Gene Expression Regulation, Neoplastic
- Angiogenesis
Collapse
Affiliation(s)
- Jing Gao
- Clinical Laboratory, Shenzhen Baoan Shiyan People’s Hospital, Guangdong Province, China
| | - Xiaojuan Lu
- Clinical Laboratory, Shenzhen Baoan Shiyan People’s Hospital, Guangdong Province, China
| | - Guanglei Wang
- Clinical Laboratory, Shenzhen Baoan Shiyan People’s Hospital, Guangdong Province, China
| | - Tanling Huang
- Clinical Laboratory, Shenzhen Baoan Shiyan People’s Hospital, Guangdong Province, China
| | - Zhongyu Tuo
- Clinical Laboratory, Shenzhen Baoan Shiyan People’s Hospital, Guangdong Province, China
| | - Weiwei Meng
- Clinical Laboratory, Shenzhen Baoan Shiyan People’s Hospital, Guangdong Province, China
| |
Collapse
|
23
|
Lee K, Kim K, Kim JY, Kim J, Kang Y, Kim YH, Kim S. Mechanisms Underlying Medication-Related Osteonecrosis of the Jaw. Oral Dis 2025; 31:1073-1083. [PMID: 39552606 PMCID: PMC12022389 DOI: 10.1111/odi.15198] [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: 04/22/2024] [Revised: 09/18/2024] [Accepted: 10/31/2024] [Indexed: 11/19/2024]
Abstract
OBJECTIVE Medication-related osteonecrosis of the jaw (MRONJ) is a rare but debilitating disease characterized by a progressive necrosis of jaw bones in patients who have received anti-resorptive or anti-angiogenic therapies. Unfortunately, we still have no validated preventive or pharmaceutical interventions to help these patients, primarily due to our limited understanding of MRONJ pathogenesis. Here, we offer an extensive review of recent studies relevant to MRONJ pathogenesis. We present a hypothesis regarding the coupling of bone resorption and angiogenesis that relies on osteoblast-derived, matrix-bound vascular endothelial growth factors to explain why ONJ is associated with both anti-resorptive and anti-angiogenic agents. METHODS A narrative review was conducted by searching databases, including PubMed, Scopus, Google Scholar, and Web of Science, to retrieve relevant reports. RESULTS Reduced bone resorption leads to reduced angiogenesis, and vice versa, creating a vicious cycle that ultimately results in ischemic necrosis of the jaw. Additionally, we suggest that reduced angiogenesis, induced by anti-resorptive or anti-angiogenic agents, aggravates bacterial infection-induced bone necrosis, explaining why the jaw bone is particularly susceptible to necrosis. CONCLUSION Our novel hypothesis will facilitate the advancement of future research and the development of more targeted approaches to managing MRONJ.
Collapse
Affiliation(s)
- Kyeongho Lee
- Department of Oral Histology and Developmental Biology, School of Dentistry and Dental Research InstituteSeoul National UniversitySeoulKorea
| | - Kihun Kim
- Department of Biomedical Informatics, School of MedicinePusan National UniversityYangsanKorea
- Department of Anatomy, School of MedicinePusan National UniversityYangsanKorea
| | - June Yeon Kim
- Department of Oral Histology and Developmental Biology, School of Dentistry and Dental Research InstituteSeoul National UniversitySeoulKorea
| | - Jin‐Woo Kim
- Department of Oral and Maxillofacial Surgery, Research Institute for Intractable Osteonecrosis of the Jaw, College of MedicineEwha Womans UniversitySeoulKorea
| | - Young‐Hoon Kang
- Department of Oral and Maxillofacial SurgeryChangwon Gyeongsang National University Hospital, Gyeongsang National University School of MedicineJinjuKorea
| | - Yun Hak Kim
- Department of Biomedical Informatics, School of MedicinePusan National UniversityYangsanKorea
- Department of Anatomy, School of MedicinePusan National UniversityYangsanKorea
| | - Sung‐Jin Kim
- Department of Oral Histology and Developmental Biology, School of Dentistry and Dental Research InstituteSeoul National UniversitySeoulKorea
| |
Collapse
|
24
|
Zhang P, Zhang N, Hu Y, Deng X, Zhu M, Lai C, Zeng W, Ke M. Role of PI3K/AKT/MAOA in glucocorticoid-induced oxidative stress and associated premature senescence of the trabecular meshwork. Aging Cell 2025; 24:e14452. [PMID: 39688282 PMCID: PMC11984687 DOI: 10.1111/acel.14452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/15/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
The oxidative stress-induced premature senescence of trabecular meshwork (TM) represents a pivotal risk factor for the development of glucocorticoid-induced glaucoma (GIG). This study aimed to elucidate the pathogenesis of TM senescence in GIG. MethodsIntraocular pressure (IOP), transmission electron microscopy and senescence-associated protein expression in TM were evaluated in GIG mice. Protein expression of phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1) and monoamine oxidase A (MAOA), phosphorylation of AKT were quantified. ROS and mitochondrial superoxide levels were measured to evaluate cellular oxidative stress. Cell cycle analysis, β-galactosidase staining, senescence-associated protein expression were employed to assess the aging status of primary human trabecular meshwork cells (pHTMs). ResultsmRNA-seq and KEGG analysis indicating PI3K/AKT pathway as a key regulator in TM of GIG. PI3K inhibitor significantly prevented IOP elevation and abnormal mitochondrial morphology of TM in the GIG mouse model. PI3K inhibitor or selective silencing of PIK3R1 alleviated dexamethasone (DEX)-induced oxidative stress, also mitochondrial dysfunction, inhibiting MAOA expression in pHTMs. The same phenomenon was observed in the GIG models with inhibition of MAOA. Further KEGG analysis indicates that cellular senescence is the key factor in the pathogenesis of GIG. TM senescence was observed in both GIG mouse and cell models. Inhibition of the PI3K/AKT/MAOA pathway significantly alleviated DEX-induced premature cellular senescence of TM in GIG models. Glucocorticoids activated the PI3K/AKT/MAOA pathway, leading to mitochondrial dysfunction, oxidative stress, and premature aging in TM, elevating IOP. This mechanism could be associated with the onset and progression of GIG, providing a potential approach for its treatment.
Collapse
Affiliation(s)
- Pengyu Zhang
- Department of OphthalmologyZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Nan Zhang
- Department of OphthalmologyZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Yixin Hu
- Department of OphthalmologyZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Xizhi Deng
- Department of OphthalmologyZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Min Zhu
- Department of OphthalmologyZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Cheng Lai
- Department of OphthalmologyZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Wen Zeng
- Department of OphthalmologyZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Min Ke
- Department of OphthalmologyZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| |
Collapse
|
25
|
Varlamova EG. Selenium-containing compounds, selenium nanoparticles and selenoproteins in the prevention and treatment of lung cancer. J Trace Elem Med Biol 2025; 88:127620. [PMID: 39970692 DOI: 10.1016/j.jtemb.2025.127620] [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: 09/15/2024] [Revised: 01/25/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
THE OBJECTIVE Is to review the latest data on the role of key organic and inorganic compounds of the essential trace element selenium, selenium-containing nanocomposites and nanoparticles, and selenoproteins in lung cancer therapy. OBJECT OF RESEARCH Sodium selenite, methylselenic acid, selenomethionine, selenium nanoparticles, mammalian selenoproteins KEY OBJECTIVES:: To describe the molecular mechanisms of the cytotoxic effect of sodium selenite, methylselenic acid and selenomethionine on lung cancer cells, to discuss the latest advances in lung cancer nanomedicine using selenium-based nanoparticles and nanocomposites and to assess the prospects for creating antitumor drugs based on them, to assess the role of selenoproteins in the progression or inhibition of lung cancer and to study the molecular mechanisms of such regulation CONCLUSIONS:: This review provides a complete picture of the role of selenium and selenium-containing agents of various natures in the regulation of carcinogenesis and therapy of lung cancer, which significantly complements the fundamental data on the functions of these compounds, on the molecular mechanisms of regulation of processes associated with lung cancer. This knowledge provides insight into the latest developments and future prospects in the treatment and prevention of lung cancer with the active participation of the trace element selenium.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", st. Institutskaya 3, Pushchino, 142290, Russia.
| |
Collapse
|
26
|
Qu F, Shen YY, Zhang XY, Wu YQ, Xu ZH, Zhai ZD, Zhong Q, Fan Z, Xu C. Ephrin-B2 acts as a positive regulator of osteogenesis-angiogenesis coupling. Int J Biol Macromol 2025; 302:140555. [PMID: 39894124 DOI: 10.1016/j.ijbiomac.2025.140555] [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: 11/04/2024] [Revised: 01/12/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
AIMS The aim of this study was to explore the role and potential mechanisms of Ephrin-B2 signaling in osteogenesis-angiogenesis coupling and to provide guidance for periodontal tissue engineering. MATERIALS AND METHODS Considering the close relationship between periodontitis and periodontal bone defects, single-cell transcriptomic data from periodontal tissues in published databases (GEO number: GSE171213) were analyzed to characterize Ephrin-B2 signaling between osteoblastic lineages and endothelial cells. To observe the effects and mechanisms of Ephrin-B2 in angiogenesis and periodontal bone regeneration, osteoblasts were constructed that overexpress Ephrin-B2 and were then co-cultured with human umbilical vein cells (HUVECs). RESULTS Single-cell sequencing revealed a deficiency in Ephrin-B2 signaling from osteoblastic lineages in periodontitis, resulting in impaired transduction of Ephrin-B2 signaling between osteoblasts and endothelial cells. In an osteoblast-endothelial cell co-culture system, Ephrin-B2 signaling from osteoblasts promoted angiogenesis. The downstream pathway might be related to VEGFR2-PI3K. The cotransplantation of osteoblasts overexpressing Ephrin-B2 and HUVECs facilitated angiogenesis and new bone generation in vivo. CONCLUSIONS This study confirmed that Ephrin-B2 signaling is an indispensable positive regulatory factor in the osteogenesis-angiogenesis coupling of periodontal tissue remodeling. These findings may help to optimize transplantation strategies and provide a new solution for the targeted treatment of periodontal bone defects. LAY SUMMARY Sufficient vascularization is a prerequisite for periodontal bone regeneration. The cotransplantation of angiogenic endothelial cells and bone-forming cells is a promising tissue engineering strategy. To facilitate the effect of the transplantation, we found evidence of Ephrin-B2 signaling deficiency in osteoblastic lineages and vascular endothelial cells of periodontitis patients from single-cell sequencing data. Moreover, by constructing a cotransplantation system with overexpression of Ephrin-B2, we found its positive role in promoting periodontal vascularized bone regeneration. The present study elucidates the positive role and potential mechanisms of Ephrin-B2 signaling in osteogenesis-angiogenesis coupling, which will help to promote the effect of periodontal tissue engineering.
Collapse
Affiliation(s)
- Fang Qu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Ying-Yi Shen
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Xin-Yu Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Ya-Qin Wu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Zi-Hang Xu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Zi-Di Zhai
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Qi Zhong
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Zhen Fan
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China.
| | - Chun Xu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.
| |
Collapse
|
27
|
Moon S, Ito Y. A simplified in vitro disease-mimicking culture system can determine the angiogenic effect of medicines on vascular diseases. Cytotechnology 2025; 77:75. [PMID: 40062227 PMCID: PMC11889311 DOI: 10.1007/s10616-025-00736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/27/2025] [Indexed: 03/21/2025] Open
Abstract
Many patients undergoing clinical regenerative treatments experience severe conditions arising from endothelial disruption. In chronic cardiac and perivascular diseases, deficiencies in vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF), and heparin, which are essential for maintaining and activating endothelial cells, can lead to angiogenic dysregulation. Endothelial disruption caused by ischemic hypoxia and a deficiency in these factors is associated with many vascular diseases. However, their pathogenic processes remain unclear at the cellular level. Therefore, the present study aimed to develop a culture system that mimics the disease environment to test the effectiveness of drug candidates in restoring damaged blood vessels in chronic vascular diseases, including coronary artery disease and peripheral vascular disease. This study focused on VEGF, IGF, and heparin and developed a pseudo-disease culture system by pre-treating human umbilical vein endothelial cells (HUVECs) with a starvation medium (EGM-2™ medium lacking VEGF, IGF, and heparin) to examine the ability of HUVECs to form a traditional 2D vascular network. The results indicated that a deficiency in these proteins results in disruptions in tube morphogenesis. Moreover, the results suggested that dysregulation of the PI3K/AKT pathway plays a key role for in vascular disruption in HUVECs. The proposed pseudo-disease starvation system provides a simple way to visualize pathological disruptions to blood vessels and assess the efficacy of drugs for vascular regeneration. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00736-4.
Collapse
Affiliation(s)
- SongHo Moon
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki Japan
| | - Yuzuru Ito
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki Japan
- Life Science Development Department, CHIYODA Corporation, Yokohama, Kanagawa Japan
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki Japan
| |
Collapse
|
28
|
Wen Y, Ma L, Liu Y, Xiong H, Shi D. Decoding the enigmatic role of T-cadherin in tumor angiogenesis. Front Immunol 2025; 16:1564130. [PMID: 40230838 PMCID: PMC11994602 DOI: 10.3389/fimmu.2025.1564130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/14/2025] [Indexed: 04/16/2025] Open
Abstract
The cadherin family, which includes T-cadherin, plays a significant role in angiogenesis, a critical process involved in tumor growth, metastasis, and recurrence. T-cadherin is extensively expressed in both normal and tumor vascular tissues and has been shown to facilitate the proliferation and migration of vascular cells in some studies. However, T-cadherin also exerts inhibitory effects on angiogenesis in various tumor tissues. The functional role of T-cadherin may vary depending on the tumor type and the interaction between tumor cells and vascular cells, suggesting that it acts as a modulator rather than a primary driver of angiogenesis. Additionally, T-cadherin exhibits distinct characteristics depending on the tumor microenvironment. This review provides an overview of recent research on the role of T-cadherin in tumor angiogenesis and discusses its potential as a diagnostic or therapeutic marker in the field of tumor biology.
Collapse
Affiliation(s)
- Yiyang Wen
- The Laboratory of Medical Mycology, Jining No.1 People’s Hospital, Jining, Shandong, China
- Department of Pathology, Jining No.1 People’s Hospital, Jining, Shandong, China
| | - Li Ma
- The Laboratory of Medical Mycology, Jining No.1 People’s Hospital, Jining, Shandong, China
| | - Yuanyuan Liu
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Dongmei Shi
- Department of Pathology, Jining No.1 People’s Hospital, Jining, Shandong, China
- Department of Dermatology, Jining No.1 People’s Hospital, Jining, Shandong, China
| |
Collapse
|
29
|
Korde A, Ramaswamy A, Anderson S, Jin L, Zhang JG, Hu B, Velasco WV, Diao L, Wang J, Pisani MA, Sauler M, Boffa DJ, Puchalski JT, Yan X, Moghaddam SJ, Takyar SS. Cigarette smoke induces angiogenic activation in the cancer field through dysregulation of an endothelial microRNA. Commun Biol 2025; 8:511. [PMID: 40155749 PMCID: PMC11953391 DOI: 10.1038/s42003-025-07710-y] [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: 08/23/2024] [Accepted: 02/10/2025] [Indexed: 04/01/2025] Open
Abstract
Cigarette smoke (CS) creates a "cancer field" in the lung that promotes malignant transformation. The molecular changes within this field are not fully characterized. We examined the significance of microRNA-1 (miR-1) downregulation as one of these changes. We found that tumor miR-1 levels in three non-small cell lung cancer cohorts show inverse correlations with the smoking burden. Lung MiR-1 levels follow a spatial gradient, have prognostic significance, and correlate inversely with the molecular markers of injury. In CS-exposed lungs, miR-1 is specifically downregulated in the endothelium. Exposure to CS induces angiogenesis by selectively degrading mature miR-1 via a vascular endothelial growth factor-driven pathway. Applying a multi-step molecular screen, we identified angiogenic genes regulated by miR-1 in the lungs of smokers. Knockdown of one of these genes, Notch homolog protein 3, simulates the anti-angiogenic effects of miR-1. These findings suggest that miR-1 can be used as an indicator of malignant transformation.
Collapse
Affiliation(s)
- Asawari Korde
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Anuradha Ramaswamy
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Seth Anderson
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Lei Jin
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jian-Ge Zhang
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Buqu Hu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Walter V Velasco
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lixia Diao
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Margaret A Pisani
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Maor Sauler
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Daniel J Boffa
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Jonathan T Puchalski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Xiting Yan
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shervin S Takyar
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
30
|
Shen S, Pan T, Liu P, Tian Y, Shi Y, Zhu W. The mechanisms and applications of endothelial progenitor cell therapy in the treatment of intracranial aneurysm. J Transl Med 2025; 23:377. [PMID: 40148864 PMCID: PMC11951544 DOI: 10.1186/s12967-025-06401-w] [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: 02/08/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
The pathophysiological mechanism of intracranial aneurysm (IA) involves the dynamic interaction of ECM abnormalities, hemodynamic stress, and inflammatory response. The rupture of intracranial aneurysm will cause serious consequences. Multiple studies have confirmed the important role and potential application of endothelial progenitor cells (EPCs) in vascular repair. This review focuses on the specific mechanism of EPCs in the treatment of intracranial aneurysms, which promote re-endothelialization and angiogenesis through bone marrow mobilization, targeted migration to the site of injury, differentiation into mature endothelial cells, and secretion of angiogenic factors. In addition, EPCs maintain ECM homeostasis by regulating MMP/IMP balance, inhibiting aneurysm wall thinning and structural damage. Based on the vascular repair mechanism of EPCs, new treatment strategies such as "biologically active" spring coils (loaded with EPCs or SDF-1α) and flow diverters(FDs) combined with EPCs therapy have been developed to synergistically promote carotid endothelialization of aneurysms and reduce the risk of recurrence. Future research needs to further validate the long-term efficacy and precise regulatory mechanisms of EPCs in clinical translation, providing new directions for IA treatment.
Collapse
Affiliation(s)
- Shiyu Shen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Tonglin Pan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Peixi Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yanlong Tian
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yuan Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| |
Collapse
|
31
|
Liu S, Li F, Cai Y, Sun L, Ren L, Yin M, Cui H, Pan Y, Gang X, Wang G. Gout drives metabolic dysfunction-associated steatotic liver disease through gut microbiota and inflammatory mediators. Sci Rep 2025; 15:9395. [PMID: 40102566 PMCID: PMC11920238 DOI: 10.1038/s41598-025-94118-7] [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: 11/25/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
This study explores the relationship between gout and metabolic dysfunction-associated steatotic liver disease (MASLD), two metabolic conditions linked to worsening health outcomes. While hyperuricemia's association with MASLD is established, the specific connection between gout and MASLD remains less explored. Using data from the UK Biobank, the study employs COX proportional hazard models, multi-state survival analysis, and Mendelian randomization to assess the independent and mutual risks of gout and MASLD. Findings indicate a mutual risk increase: male gout patients, those younger than 60, and those with high BMI are particularly susceptible to MASLD, while female MASLD patients are at heightened risk for gout. Shared risk factors for both conditions include high BMI, hypertension, diabetes, and hyperuricemia. The study further identifies a bidirectional causal link, with gout leading to MASLD, mediated by gut microbiota Ruminococcaceae and proteins like IL-2 and GDF11, implicating specific metabolic pathways. The findings highlight a clinical and mechanistic correlation, emphasizing the need for targeted interventions to address these overlapping metabolic pathways in future treatments.
Collapse
Affiliation(s)
- Siyuan Liu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Fan Li
- Department of Hepatobiliary and Pancreatic Medicine, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Yunjia Cai
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Linan Ren
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Mengsha Yin
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Huijuan Cui
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Yujie Pan
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
32
|
Kim J, Moon SY, Kang HG, Kim HJ, Choi JS, Lee SHS, Park K, Won SY. Therapeutic potential of AAV2-shmTOR gene therapy in reducing retinal inflammation and preserving endothelial Integrity in age-related macular degeneration. Sci Rep 2025; 15:9517. [PMID: 40108376 PMCID: PMC11923296 DOI: 10.1038/s41598-025-93993-4] [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: 09/25/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Age-related macular degeneration (AMD) is a prevalent retinal disorder that leads to central vision loss, mainly due to chronic inflammation. Tumor necrosis factor-alpha (TNF-α) is a critical mediator of inflammatory responses within the retinal environment. This study has investigated TNF-α's influence on inflammatory cytokine production and endothelial barrier integrity in human microglial (HMC3) and endothelial (HUVEC) cells. We found that TNF-α significantly elevated the expression and secretion of interleukin-6 (IL-6) and interleukin-1β (IL-1β) in HMC3 cells and disrupted endothelial tight junctions in HUVECs, as evidenced by weakened ZO-1 staining and compromised barrier function. To mitigate these effects and further investigate the in vitro mechanism of actions in CRG-01's in vivo therapeutic efficacy of anti-inflammation, we employed AAV2-shmTOR, CRG-01, as the candidate for therapeutic vector targeting the mammalian target of the rapamycin (mTOR) pathway. TNF-α-induced IL-6, IL-1β, and NF-κB signaling in HMC3 cells were significantly reduced by AAV2-shmTOR treatment, which may present a promising avenue for the fight against AMD. It also effectively preserved endothelial tight junction integrity in TNF-α-treated HUVECs, providing reassurance about its effectiveness. Furthermore, the supernatant medium collected from AAV2-shmTOR-treated HMC3 cells decreased oxidative stress, protein oxidation, and cytotoxicity in ARPE retinal pigment epithelial cells. These results strongly suggested that CRG-01, the candidate therapeutic vector of AAV2-shmTOR, may have a therapeutic potential to treat AMD-related retinal inflammation.
Collapse
Affiliation(s)
- Jin Kim
- Institute of New Drug Development Research, CdmoGen Co., Ltd, Seoul, 05855, Republic of Korea
- CdmoGen Co., Ltd, Cheongju, 28577, Republic of Korea
| | - Seo Yun Moon
- Institute of New Drug Development Research, CdmoGen Co., Ltd, Seoul, 05855, Republic of Korea
- CdmoGen Co., Ltd, Cheongju, 28577, Republic of Korea
| | - Ho Geun Kang
- Institute of New Drug Development Research, CdmoGen Co., Ltd, Seoul, 05855, Republic of Korea
- CdmoGen Co., Ltd, Cheongju, 28577, Republic of Korea
| | - Hee Jong Kim
- Institute of New Drug Development Research, CdmoGen Co., Ltd, Seoul, 05855, Republic of Korea
- CdmoGen Co., Ltd, Cheongju, 28577, Republic of Korea
| | - Jun Sub Choi
- Institute of New Drug Development Research, CdmoGen Co., Ltd, Seoul, 05855, Republic of Korea
- CdmoGen Co., Ltd, Cheongju, 28577, Republic of Korea
| | - Steven Hyun Seung Lee
- Institute of New Drug Development Research, CdmoGen Co., Ltd, Seoul, 05855, Republic of Korea
- CdmoGen Co., Ltd, Cheongju, 28577, Republic of Korea
| | - Keerang Park
- Institute of New Drug Development Research, CdmoGen Co., Ltd, Seoul, 05855, Republic of Korea.
- CdmoGen Co., Ltd, Cheongju, 28577, Republic of Korea.
| | - So-Yoon Won
- Institute of New Drug Development Research, CdmoGen Co., Ltd, Seoul, 05855, Republic of Korea.
- CdmoGen Co., Ltd, Cheongju, 28577, Republic of Korea.
| |
Collapse
|
33
|
Li L, Hammerlindl H, Shen SQ, Bao F, Hammerlindl S, Altschuler SJ, Wu LF. A phenopushing platform to identify compounds that alleviate acute hypoxic stress by fast-tracking cellular adaptation. Nat Commun 2025; 16:2684. [PMID: 40102413 PMCID: PMC11920246 DOI: 10.1038/s41467-025-57754-1] [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: 06/25/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
Severe acute hypoxic stress is a major contributor to the pathology of human diseases, including ischemic disorders. Current treatments focus on managing consequences of hypoxia, with few addressing cellular adaptation to low-oxygen environments. Here, we investigate whether accelerating hypoxia adaptation could provide a strategy to alleviate acute hypoxic stress. We develop a high-content phenotypic screening platform to identify compounds that fast-track adaptation to hypoxic stress. Our platform captures a high-dimensional phenotypic hypoxia response trajectory consisting of normoxic, acutely stressed, and chronically adapted cell states. Leveraging this trajectory, we identify compounds that phenotypically shift cells from the acutely stressed state towards the adapted state, revealing mTOR/PI3K or BET inhibition as strategies to induce this phenotypic shift. Importantly, our compound hits promote the survival of liver cells exposed to ischemia-like stress, and rescue cardiomyocytes from hypoxic stress. Our "phenopushing" platform offers a general, target-agnostic approach to identify compounds and targets that accelerate cellular adaptation, applicable across various stress conditions.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Heinz Hammerlindl
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Susan Q Shen
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Feng Bao
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Sabrina Hammerlindl
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Steven J Altschuler
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.
| | - Lani F Wu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
34
|
Dartora VFC, Carney R, Wang A, Qiu P, Panitch A. Extracellular matrix ligands modulate the endothelial progenitor cell secretome for enhanced angiogenesis. Acta Biomater 2025; 195:240-255. [PMID: 39954753 DOI: 10.1016/j.actbio.2025.02.028] [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: 09/28/2024] [Revised: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Wound healing is a complex physiological process fundamentally dependent on angiogenesis for effective tissue repair. Endothelial progenitor cells (EPCs) have shown significant potential in promoting angiogenesis, primarily through their secretome, rich in proteins and extracellular vesicles (EVs) essential for cell signaling and tissue regeneration. This study investigates the effect of a collagen-bound proteoglycan mimetic (LXW7-DS-SILY or LDS), that binds to the αvβ3 integrin receptor, on the EPC secretome, with a dual focus on the proteomic content and the functional properties of EVs. Utilizing high-resolution two-dimensional liquid chromatography-tandem mass spectrometry (LC-MS/MS) alongside bioinformatic analysis, we identified significant alterations in protein expression profiles, particularly in angiogenesis and wound healing pathways. The functional impact of these changes was validated through biological assays, where the whole secretome and the EV fraction from EPCs seeded on collagen-bound LDS markedly enhanced vascular network formation, driven by the increase of growth factors and angiogenic regulators such as FGFR1, NRP1, and ANGPT2 within the EV fraction. Gene Ontology (GO) enrichment analysis further highlighted the enrichment of proteins within the EVs driving biological processes, including 'response to wounding' and 'positive regulation of cell motility'. These results underscore that LDS modulates the EPC secretome and significantly enhances its angiogenic potential, offering a promising therapeutic strategy for non-healing and ischemic wounds and suggesting that biomaterials can be modified to control the EV secretome to enhance tissue repair. Functional assays validating the omics data highlight the robustness of LDS as a targeted therapeutic for enhancing angiogenesis and tissue repair in clinical settings. Moreover, the pivotal role of EVs in mediating pro-angiogenic effects offers insights into developing biomaterial therapies that exploit key regulators within the EPC secretome for wound healing. STATEMENT OF SIGNIFICANCE: This manuscript explores how a proteoglycan mimetic that binds to both collagen and the αvβ3 integrin receptor affects the proteome component of the secretome from endothelial progenitor cells (EPCs). It presents functional biological data, analytical data, and proteomic analysis of the soluble and extracellular vesical (EV) components of the secratome. The proteomic data maps to the observed enhanced angiogenic potential of the EVs. These results suggest that by controlling the cellular environment and judicially engineering how cells interact with a biomaterial can influence the proteomic composition of EVs to enhance tissue regeneration. This is the foundation of future work aimed at engineering biomaterial cell systems to influence the proteomic component of EVs for therapeutic use.
Collapse
Affiliation(s)
- Vanessa F C Dartora
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Randy Carney
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA
| | - Aijun Wang
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Peng Qiu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Alyssa Panitch
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Department of Surgery, University of California Davis, Sacramento, CA 95817, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA.
| |
Collapse
|
35
|
Pooraskari Z, Barri Ghazani H, Piri R, Habibi S, Shahidi M. Evaluation of the antiangiogenic effect of AMG232 in multiple myeloma coculture systems. Med Oncol 2025; 42:107. [PMID: 40082344 DOI: 10.1007/s12032-025-02659-8] [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: 01/03/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
This study explored the efficacy of AMG232, a potent and selective MDM2 inhibitor, as an antiangiogenic agent in a multiple myeloma (MM) cell line (AMO-1) cocultured with endothelial cells (HUVECs) in vitro. HUVECs and AMO-1 cells were cocultured in transwell systems. Cell viability was assessed through an MTT assay after exposure to various concentrations of AMG232. Following treatment, gene expression changes were analyzed via quantitative real-time PCR. Wound healing and tube formation assays were also conducted to quantify the effects on cell migration and angiogenesis. AMG232 showed dose-dependent cytotoxicity in AMO-1 cells (IC50 = 386.1 nM), whereas HUVECs were moderately sensitive (IC50= 942.1 nM). In coculture, both cell types displayed increased resistance to AMG232, indicating a protective cell-cell interaction. Treatment with 250-nM AMG232 significantly downregulated the mRNA expression of angiogenic factors-including VEGF-A, VEGFR-2, MMP-2, IL-6, and HIF-1α-in both AMO-1 cells and HUVECs (P < 0.05). Wound healing assays revealed that AMG232 markedly inhibited HUVEC migration, with significantly reduced wound closure rates at 24 and 48 h compared with the controls (P < 0.01). Tube formation assays further revealed that AMG232 substantially decreased angiogenesis in HUVECs, as evidenced by reductions in junction number, mesh number, and total tube length (P < 0.01). Our research revealed that AMG232 effectively inhibited angiogenesis and exhibited cytotoxic effects on MM cells by downregulating key angiogenic factors and impairing endothelial cell functions. These results suggest that AMG232 has significant potential as a therapeutic agent for targeting angiogenesis in MM treatment.
Collapse
Affiliation(s)
- Zahra Pooraskari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Barri Ghazani
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reyhane Piri
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sina Habibi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Minoo Shahidi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
36
|
Pushpakumar S, Juin SK, Almarshood H, Gondim DD, Ouseph R, Sen U. Diallyl Trisulfide Attenuates Ischemia-Reperfusion-Induced ER Stress and Kidney Dysfunction in Aged Female Mice. Cells 2025; 14:420. [PMID: 40136669 PMCID: PMC11941362 DOI: 10.3390/cells14060420] [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: 12/11/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Ischemia-reperfusion injury (IRI) is a common cause of acute kidney injury (AKI) in the aging population. Gender studies show that aging is associated with loss of protection from AKI in the female population. While ER stress contributes to IRI-induced AKI in the young, ER regulation during IR in the aged kidney is unclear. Because current evidence suggests hydrogen sulfide (H2S) modulates ER stress, we investigated whether exogenous supplementation of diallyl trisulfide (DATS), an H2S donor, mitigates AKI in aged female kidneys. Wild-type (WT, C57BL/6J) mice aged 75-78 weeks were treated with or without DATS before and after renal IRI. IRI increased ER stress proteins, inflammation, and fibrosis markers in the IRI kidney compared to the control. DATS mitigated ER stress, and reduced inflammation and fibrosis markers in the IRI kidney. Further, IRI kidneys demonstrated reduced blood flow, vascularity, angiogenesis, increased resistive index (RI), and reduced function. DATS treatment upregulated PI3K, AKT, p-mTOR, and pMAPK signaling to stimulate angiogenesis, which improved vascular density, blood flow, and renal function. Together, our results suggest that DATS rescues the aged female kidney IRI by modulating ER stress and upregulation of angiogenesis.
Collapse
Affiliation(s)
- Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Subir Kumar Juin
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| | - Hebah Almarshood
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Dibson Dibe Gondim
- Department of Pathology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rosemary Ouseph
- Division of Nephrology & Hypertension, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
37
|
Zhou L, Liu B, Jian X, Jiang L, Liu K. Effect of dietary patterns and nutritional supplementation in the management of endometriosis: a review. Front Nutr 2025; 12:1539665. [PMID: 40144566 PMCID: PMC11937854 DOI: 10.3389/fnut.2025.1539665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Endometriosis is an estrogen-dependent chronic inflammatory disease which causes dysmenorrhea, chronic pelvic pain, and infertility in women of childbearing age, significantly impacting their quality of life and physical and mental health. The etiology of endometriosis remains unclear, with oxidative stress and inflammation currently thought to play pivotal roles in its pathophysiology. Epidemiological studies and clinical trials indicate that varying dietary patterns and specific nutrient supplementation can influence oxidative stress markers and levels of inflammatory factors and related pathways, potentially impacting the progression of endometriosis. In this review, we summarize the roles of oxidative stress and inflammation in endometriosis and thoroughly examine the current understanding of the effect of dietary patterns and nutrient supplementation in treating endometriosis. This study suggests that nutrients may prevent the occurrence of endometriosis by modulating levels of inflammatory factors, regulating angiogenesis, and influencing the metabolism of estrogen pathways. The findings might provide new insights into the treatment of endometriosis patients and the potential benefits of dietary patterns and nutrient supplementation in patients with endometriosis.
Collapse
Affiliation(s)
| | | | | | - Lili Jiang
- *Correspondence: Lili Jiang, ; Kuiran Liu,
| | - Kuiran Liu
- *Correspondence: Lili Jiang, ; Kuiran Liu,
| |
Collapse
|
38
|
Long S, Wang Y. Association of TAB2 gene polymorphism with endometrial cancer susceptibility and clinical analysis. Turk J Obstet Gynecol 2025; 22:1-12. [PMID: 40062608 PMCID: PMC11894771 DOI: 10.4274/tjod.galenos.2025.24983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/06/2025] [Indexed: 03/14/2025] Open
Abstract
Objective Transforming growth factor-β-activated kinase 1 binding protein 2 (TAB2) plays a vital role in inflammatory pathways. It has also been considered a potential target for the enhancement of the the antiestrogen effects. Previous evidence has indicated that TAB2 gene variants are associated with several diseases, whereas their potential correlation with endometrial cancer (EC) is unclear. This study aims to initially explore the association between TAB2 gene polymorphisms (rs237028 /AG, rs521845 T/G, and rs652921 T/C) and EC. Materials and Methods Polymerase chain reaction-restriction fragment length polymorphism was applied to determine the genotype composition and the allele frequencies of TAB2 gene variant polymorphisms in 270 EC patients and 294 healthy controls. Results The G allele of rs521845 was related to the increase of EC risk [p=0.08, odds ratio (OR): 0.72, 95% confidence interval (CI): 0.56-0.91]. Moreover, EC risk was associated with rs521845 in different genetic models (p=0.017, OR: 0.63, 95% CI: 0.44-0.91 in the codominant model; p=0.0051, OR: 0.61, 95% CI: 0.43-0.87 in the dominant model). For rs237028, the percentage of AG genotype in patients with highly differentiated tumours (G1) was significantly higher than that in moderately, poorly differentiated patients (G2/G3) (p=0.031, OR: 0.77, 95% CI: 0.45-1.30). Conclusion Our results showed that the rs521845 polymorphism of TAB2, was associated with EC risk, suggesting that TAB2 may play a crucial role in EC prognosis.
Collapse
Affiliation(s)
- Siyu Long
- Sichuan University, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Center for Translational Medicine, Laboratory of Molecular Translational Medicine, Sichuan, China
- Sichuan University West China Second University Hospital, Clinic of Andrology/Sichuan Human Sperm Bank, Chengdu, China
| | - Yanyun Wang
- Sichuan University, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Center for Translational Medicine, Laboratory of Molecular Translational Medicine, Sichuan, China
| |
Collapse
|
39
|
Wang Y, Rozen V, Zhao Y, Wang Z. Oncogenic activation of PI K3 CA in cancers: Emerging targeted therapies in precision oncology. Genes Dis 2025; 12:101430. [PMID: 39717717 PMCID: PMC11665392 DOI: 10.1016/j.gendis.2024.101430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/04/2024] [Accepted: 08/25/2024] [Indexed: 12/25/2024] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) are heterodimers consisting of a p110 catalytic subunit and a p85 regulatory subunit. The PIK3CA gene, which encodes the p110α, is the most frequently mutated oncogene in cancer. Oncogenic PIK3CA mutations activate the PI3K pathway, promote tumor initiation and development, and mediate resistance to anti-tumor treatments, making the mutant p110α an excellent target for cancer therapy. PIK3CA mutations occur in two hotspot regions: one in the helical domain and the other in the kinase domain. The PIK3CA helical and kinase domain mutations exert their oncogenic function through distinct mechanisms. For example, helical domain mutations of p110α gained direct interaction with insulin receptor substrate 1 (IRS-1) to activate the downstream signaling pathways. Moreover, p85β proteins disassociate from helical domain mutant p110α, translocate into the nucleus, and stabilize enhancer of zeste homolog 1/2 (EZH1/2). Due to the fundamental role of PI3Kα in tumor initiation and development, PI3Kα-specific inhibitors, represented by FDA-approved alpelisib, have developed rapidly in recent decades. However, side effects, including on-target side effects such as hyperglycemia, restrict the maximum dose and thus clinical efficacy of alpelisib. Therefore, developing p110α mutant-specific inhibitors to circumvent on-target side effects becomes a new direction for targeting PIK3CA mutant cancers. In this review, we briefly introduce the function of the PI3K pathway and discuss how PIK3CA mutations rewire cell signaling, metabolism, and tumor microenvironment, as well as therapeutic strategies under development to treat patients with tumors harboring a PIK3CA mutation.
Collapse
Affiliation(s)
- Yuxiang Wang
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Valery Rozen
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Yiqing Zhao
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zhenghe Wang
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
40
|
Chen H, Peng C, Fang F, Li Y, Liu X, Hu Y, Wang G, Liu X, Shen Y. Angiogenesis within atherosclerotic plaques: Mechanical regulation, molecular mechanism and clinical diagnosis. MECHANOBIOLOGY IN MEDICINE 2025; 3:100114. [PMID: 40396135 PMCID: PMC12082165 DOI: 10.1016/j.mbm.2025.100114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/12/2024] [Accepted: 01/05/2025] [Indexed: 05/22/2025]
Abstract
Atherosclerosis (AS) is a disease characterized by focal cholesterol accumulation and insoluble inflammation in arterial intima, leading to the formation of an atherosclerotic plaque consisting of lipids, cells, and fibrous matrix. The presence of plaque can restrict or obstruct blood flow, resulting in arterial stenosis and local mechanical microenvironment changes including flow shear stress, vascular matrix stiffness, and plaque structural stress. Neovascularization within the atherosclerotic plaque plays a crucial role in both plaque growth and destabilization, potentially leading to plaque rupture and fatal embolism. However, the exact interactions between neovessels and plaque remain unclear. In this review, we provide a comprehensive analysis of the origin of intraplaque neovessels, the contributing factors, underlying molecular mechanisms, and associated signaling pathways. We specifically emphasize the role of mechanical factors contributing to angiogenesis in atherosclerotic plaques. Additionally, we summarize the imaging techniques and therapeutic strategies for intraplaque neovessels to enhance our understanding of this field.
Collapse
Affiliation(s)
- Hanxiao Chen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Chengxiu Peng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Fei Fang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yuhao Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiaran Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Ying Hu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Guixue Wang
- Jinfeng Laboratory, Chongqing 401329, China
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- Jinfeng Laboratory, Chongqing 401329, China
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
41
|
Dai Q, Peng Y, He P, Wu X. Interactions and communications in the prostate tumour microenvironment: evolving towards effective cancer therapy. J Drug Target 2025; 33:295-315. [PMID: 39445641 DOI: 10.1080/1061186x.2024.2418344] [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: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Prostate cancer is one of the most common malignancies in men. The tumour microenvironment (TME) has a critical role in the initiation, progression, and metastasis of prostate cancer. TME contains various cell types, including cancer-associated fibroblasts (CAFs), endothelial cells, immune cells such as macrophages, lymphocytes B and T, natural killer (NK) cells, and other proteins such as extracellular matrix (ECM) components. The interactions and communications between these cells within the TME are crucial for the growth and response of various solid tumours, such as prostate cancer to different anticancer modalities. In this review article, we exemplify the various mechanisms by which the TME influences prostate cancer progression. The roles of different cells, cytokines, chemokines, and growth factors in modulating the immune response and prostate tumour growth will be discussed. The impact of these cells and factors and other ECM components on tumour cell invasion and metastasis will also be discussed. We explain how these interactions in TME can affect the response of prostate cancer to therapy. We also highlight the importance of understanding these interactions to develop novel therapeutic approaches for prostate cancer.
Collapse
Affiliation(s)
- Qiang Dai
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yanling Peng
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Peng He
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaojun Wu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
42
|
Li D, Jiang M, Song Y, Liang X, Lv J, Zhang L, Li Z, Fan L, Du H. Preventive vs. therapeutic effects of Shoutai Wan: Maintaining an acidic microenvironment at the maternal-fetal interface to promote angiogenesis and minimize pregnancy loss in RSA mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119345. [PMID: 39824268 DOI: 10.1016/j.jep.2025.119345] [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: 10/13/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The classic TCM prescription, Shoutai Wan (STW), is extensively used in clinical settings to manage threatened miscarriage and Recurrent spontaneous abortion (RSA). The complexity of pregnancy physiology, coupled with diverse etiologies, and the specificity of energy metabolism for normal embryo attachment and development, pose challenges to clinical diagnosis and treatment. The specific molecular mechanisms of how STW regulates these biological processes and contributes to the treatment of RSA remain to be elucidated. AIM OF THE STUDY This study aims to investigate the causes of early pregnancy loss in RSA mice and explore how STW mitigates this loss. MATERIALS AND METHODS An RSA mouse model will be established and treated with STW and Dydrogesterone (DYD). Embryo loss will be quantified on the 14th day of pregnancy, and embryos will be collected on the 6th and 10th days to observe the embryonic condition and assess pathological changes. The study will analyze aerobic glycolysis and angiogenesis at the maternal-fetal interface (MFI). Additionally, STW on a knockdown LDHA mouse model and Human Endometrial Microvascular Endothelial Cells (HEMECs) in vitro will also be examined to verify the mechanism. RESULTS Compared with the control group, the RSA group exhibited significant embryo loss, and reduced levels of aerobic glycolysis at the MFI, the precarious acidic microenvironment (AME), and the PI3K/AKT/mTOR signaling axis downregulated, leading to impaired angiogenesis, which ameliorated following STW treatment. STW treatment enhanced key aerobic glycolysis enzymes-HK2, PKM2, LDHA-and lactate levels, thereby maintaining the AME and upregulating the PI3K/AKT/mTOR axis. This, in turn, promoted the expression of angiogenesis-related factors (VEGFA and VEGFR2) at the MFI, thereby improving angiogenesis, and the same was seen in sh-LDHA mice. In vitro studies confirmed that STW could counteract the glycolysis decline caused by increased oxygen levels, a recovery that was impaired after LDHA knockdown or PI3K inhibition. CONCLUSIONS In RSA mice, disturbances in aerobic glycolysis at the MFI prevent the maintenance of a stable AME, thus impairing angiogenesis and leading to embryo loss, and STW effectively improve early pregnancy outcomes, and laying the foundation for uterine spiral artery remodeling.
Collapse
Affiliation(s)
- Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China
| | - Min Jiang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China
| | - Yajing Song
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China
| | - Xiao Liang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China
| | - Jingfang Lv
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China
| | - Li Zhang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; First College of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Lijie Fan
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China; Hebei Key Laboratory of Integrative Medicine on Liver-kidney Patterns, Shijiazhuang, 050091, China
| | - Huilan Du
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China; Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, 050091, China; Hebei Key Laboratory of Integrative Medicine on Liver-kidney Patterns, Shijiazhuang, 050091, China.
| |
Collapse
|
43
|
Zhu Y, Chen P, Zhang Z, He X, Wang R, Fang Q, Xu Z, He W. aFGF gene-modified adipose-derived mesenchymal stem cells promote healing of full-thickness skin defects in diabetic rats. Stem Cell Res Ther 2025; 16:93. [PMID: 40001190 PMCID: PMC11863861 DOI: 10.1186/s13287-025-04241-5] [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: 12/04/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Chronic diabetic wounds pose a significant clinical challenge due to the limited efficacy of current treatments. This study aimed to investigate the role and potential mechanisms of adipose-derived mesenchymal stem cells (ADSCs) overexpressing acidic fibroblast growth factor (aFGF) in diabetic wound healing in a rat model. METHODS ADSCs were genetically modified to achieve stable overexpression of aFGF. Varying doses of aFGF-ADSCs (1 × 106, 2 × 106, 3 × 106, 4 × 106) were injected into the muscular tissue surrounding diabetic rat wounds. We assessed aFGF expression and its impact on various stages of wound healing, including angiogenesis, inflammatory response, epithelialization, and collagen deposition. Transcriptomic sequencing was performed to explore the underlying mechanisms driving enhanced wound healing. RESULTS Lentiviral transduction successfully induced stable aFGF overexpression in ADSCs. In vivo experiments revealed that varying doses of aFGF-ADSCs markedly enhanced wound healing in diabetic rats in a dose-dependent manner. The dose of 3 × 10⁶ aFGF-ADSCs demonstrated the most significant effect. In the 3 × 106 aFGF-ADSCs group, expression levels of aFGF, CD31, and CD163 were significantly higher than in other groups (p < 0.05), while CD86 expression was significantly lower (p < 0.05). CONCLUSION Single doses of aFGF-ADSCs comprehensively improved various aspects of wound repair in diabetic rats, offering a potential new approach for treating chronic diabetic wounds. The mechanism of action involves promoting angiogenesis, modulating inflammatory responses, accelerating epithelialization, and optimizing collagen deposition.
Collapse
Affiliation(s)
- Yiren Zhu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, 350001, China
| | - Pinhua Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, 350001, China
- Department of Emergency Trauma Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fujian Trauma Medicine Center, Fuzhou, Fujian, 350001, China
- Fujian Key Laboratory of Emergency Medicine, Fuzhou, Fujian, 350001, China
| | - Zhengchao Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, 350001, China
- Department of Emergency Trauma Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fujian Trauma Medicine Center, Fuzhou, Fujian, 350001, China
- Fujian Key Laboratory of Emergency Medicine, Fuzhou, Fujian, 350001, China
| | - XueYi He
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, 350001, China
- Department of Emergency Trauma Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fujian Trauma Medicine Center, Fuzhou, Fujian, 350001, China
- Fujian Key Laboratory of Emergency Medicine, Fuzhou, Fujian, 350001, China
| | - Ruoli Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, 350001, China
- Department of Emergency Trauma Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fujian Trauma Medicine Center, Fuzhou, Fujian, 350001, China
- Fujian Key Laboratory of Emergency Medicine, Fuzhou, Fujian, 350001, China
| | - Qi Fang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, 350001, China
- Department of Emergency Trauma Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fujian Trauma Medicine Center, Fuzhou, Fujian, 350001, China
- Fujian Key Laboratory of Emergency Medicine, Fuzhou, Fujian, 350001, China
| | - Zhixian Xu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, 350001, China
- Department of Emergency Trauma Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, 350001, China
- Fujian Trauma Medicine Center, Fuzhou, Fujian, 350001, China
- Fujian Key Laboratory of Emergency Medicine, Fuzhou, Fujian, 350001, China
| | - Wubing He
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, 350001, China.
- Department of Emergency Trauma Surgery, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China.
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, 350001, China.
- Fujian Trauma Medicine Center, Fuzhou, Fujian, 350001, China.
- Fujian Key Laboratory of Emergency Medicine, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
44
|
Guo K, Ma P, Yang Q, Xu L, Zhang B, Zhang H, Zheng Z, Zhuo Z. Activation of RHO-GTPase gene pattern correlates with adverse clinical outcome and immune microenvironment in clear cell renal cell carcinoma. Clin Exp Med 2025; 25:67. [PMID: 39998699 PMCID: PMC11861022 DOI: 10.1007/s10238-025-01593-3] [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: 01/11/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC), the most prevalent renal cancer subtype, is frequently associated with poor prognosis. RHO-GTPase signaling genes have been implicated in tumor aggressiveness and unfavorable survival, but their potential in risk stratification and therapeutic guidance for ccRCC patients remains unexplored. Univariate regression identified prognostically relevant RHO-GTPase signaling genes, followed by consensus clustering for ccRCC subtype classification. LASSO regression selected key genes to construct a six-gene risk model. The model was evaluated for prognostic stratification, immune status, immunotherapy response, and chemotherapy sensitivity. Key genes were analyzed at the genomic, single-cell, and protein levels to explore underlying mechanisms. Among 62 prognostically relevant RHO-GTPase signaling genes, six (ARHGAP11B, NUF2, PLK1, CYFIP2, IQGAP2, and VAV3) were identified to form a robust prognostic signature. This model stratified patients into high- and low-risk groups, with high-risk patients demonstrating significantly worse outcomes. The model exhibited excellent predictive accuracy (AUC > 0.7 in training and validation cohorts). High-risk patients were characterized by an immunosuppressive microenvironment and reduced sensitivity to immunotherapy. Drug sensitivity analysis revealed 107 agents correlated with the risk score, underscoring therapeutic relevance. Mechanistically, the six key genes showed distinct expression patterns, cellular distribution, and positive correlation with VHL mutations, highlighting their potential as actionable drug targets. This study established a novel six-gene RHO-GTPase signaling model for predicting prognosis, immune status, and therapeutic responses in ccRCC, which offers potential for improving patient stratification and guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Kehang Guo
- Department of Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Pengyue Ma
- Department of Nephrology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Yang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Lingli Xu
- Dadong Street Community Health Service Center, Guangzhou, 510080, China
| | - Biixiong Zhang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Hong Zhang
- Department of Lymphoma, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China.
| | - Zhongwen Zheng
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Heyuan People's Hospital, Heyuan, 517001, Guangdong, China.
| | - Zewei Zhuo
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
45
|
Deng Q, Du F, Pan S, Xia Y, Zhu Y, Zhang J, Li C, Yu S. Activation of angiopoietin-1 signaling with engineering mesenchymal stem cells promoted efficient angiogenesis in diabetic wound healing. Stem Cell Res Ther 2025; 16:75. [PMID: 39985096 PMCID: PMC11846275 DOI: 10.1186/s13287-025-04207-7] [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: 11/13/2024] [Accepted: 01/29/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Vascular insufficiency is associated with the pathogenesis and therapeutic outcomes of diabetic foot ulcers (DFU). While mesenchymal stem cells (MSCs) hold potential for DFU treatment, further enhancement in promoting angiogenesis in the challenging DFU wounds is imperative. METHODS The differential expression of pro- and anti-angiogenic factors during both normal and diabetic wound healing was compared using quantitative PCR. MSCs derived from the umbilical cord was prepared, and the engineered MSC (MSCANG1) overexpressing both the candidate pro-angiogenic gene, angiopoietin-1 (ANG1), and green fluorescent protein (GFP) was constructed using a lentiviral system. The pro-vascular stabilizing effects of MSCANG1 were assessed in primary endothelial cell cultures. Subsequently, MSCANG1 was transplanted into streptozotocin (STZ)-induced diabetic wound models to evaluate therapeutic effects on angiogenesis and wound healing. The underlying mechanisms were further examined both in vitro and in vivo. RESULTS The comprehensive analysis of the temporal expression of pro- and anti-angiogenic factors revealed a consistent impairment in ANG1 expression throughout diabetic wound healing. MSCANG1 exhibited robust EGFP expression in 80% of cells, with overexpression and secretion of the ANG1 protein. MSCANG1 notably enhanced the survival and tubulogenesis of endothelial cells and promoted the expression of junction proteins, facilitating the establishment of functional vasculature with improved vascular leakage. Although MSCANG1 did not enhance the survival of engrafted MSCs in diabetic wounds, it significantly promoted angiogenesis in diabetic wound healing, fostering the establishment of stable vasculature during the healing process. Activation of the protein kinase B (Akt) pathway and suppression of proto-oncogene tyrosine kinase Src (Src) activity in MSCANG1-treated diabetic wounds confirmed efficient angiogenesis process. Consequently, epidermal and dermal reconstruction, as well as skin appendage regeneration were markedly accelerated in MSCANG1-treated diabetic wounds compared to MSC-treated wounds. CONCLUSION Treatment with MSCs alone promotes angiogenesis and DFU healing, while the engineering of MSCs with ANG1 provides substantial additional benefits to this therapeutic process. The engineering of MSCs with ANG1 presents a promising avenue for developing innovative strategies in managing DFU.
Collapse
Affiliation(s)
- Qiong Deng
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Fangzhou Du
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Shenzhen Pan
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yuchen Xia
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yuxin Zhu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jingzhong Zhang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China.
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Chenglong Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Shuang Yu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China.
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
46
|
Wu B, Wang X, Zhang W, Wang X, Wang QM, Pang YJ, Kong YC. Lycium barbarum polysaccharide inhibits retinal neovascularization and inflammation in vitro and in vivo. Int J Ophthalmol 2025; 18:216-221. [PMID: 39967970 PMCID: PMC11754022 DOI: 10.18240/ijo.2025.02.03] [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: 10/09/2024] [Accepted: 11/25/2024] [Indexed: 02/20/2025] Open
Abstract
AIM To explore the effect and mechanism of Lycium barbarum polysaccharide (LBP) inhibiting retinal neovascularization. METHODS In vitro tests were performed on human retinal microvascular endothelial cells (HRECs) from three groups, including control group (normal oxygen), hypoxic group (hypoxia at 37°C, 1% O2, 5% CO2, and 94% N2), and LBP group (hypoxic group with LBP 100 µg/mL). In vivo experiments, C57 mice were divided into three groups: control group (normal rearing group), the oxygen-induced ischemic retinopathy (OIR) group, and the OIR with 50 mg/kg LBP group. Retinal neovascularization was observed by fluorescein angiography and quantified. Retinal thickness was evaluated by Hematoxylin and eosin (HE) stain. The expression of epidermal growth factor receptor (EGFR), phosphatidylinositol 3-kinase (PI3K), mammalian target of rapamycin (mTOR), phosphorylated mammalian target of rapamycin (p-mTOR), protein kinase B (AKT), phosphorylated protein kinase B (p-AKT), interleukin-1β (IL-1β), inducible nitric oxide synthase (iNOS), and tumor necrosis factor-α (TNF-α) in each group were analyzed by Western blot. IL-1β level in retina was analyzed using immunohistochemical staining. RESULTS The increased area of neovascular clusters in OIR mice was significantly decreased by LBP. Retinal thickness of OIR mice was significantly thinner compared with normal oxygenated mice and was increased in LBP group. Compared with those in the hypoxic groups, Western blotting of HRECs and retinal tissues revealed that the expression of EGFR, PI3K, p-mTOR, p-AKT, IL-1β, iNOS, and TNF-α decreased in the LBP group but was still greater than that in control group. Moreover, IL-1β was reduced in retinal sections treated with LBP. In the scratch test, the cell migration of the hypoxic group was significantly greater than that of the control group, while LBP treatment attenuated this increase in migration. CONCLUSION LBP reduces retinal neovascularization and inflammation in vivo and inhibits the migration of HRECs in vitro by regulating the EGFR/PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Bin Wu
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Clinical College of Ophthalmology Tianjin Medical University, Tianjin 300102, China
| | - Xue Wang
- Department of Ophthalmology, Tianjin Hospital of Integrated Traditional Chinese and Western Medicine, Tianjin 300100, China
| | - Wei Zhang
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Clinical College of Ophthalmology Tianjin Medical University, Tianjin 300102, China
| | - Xin Wang
- Department of Ophthalmology, Tianjin Hospital of Integrated Traditional Chinese and Western Medicine, Tianjin 300100, China
| | - Qi-Miao Wang
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Clinical College of Ophthalmology Tianjin Medical University, Tianjin 300102, China
| | - Ya-Ju Pang
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Clinical College of Ophthalmology Tianjin Medical University, Tianjin 300102, China
| | - Yi-Chun Kong
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Clinical College of Ophthalmology Tianjin Medical University, Tianjin 300102, China
| |
Collapse
|
47
|
Li S, Wang L, Bendersky VA, Gao Q, Wang J, Xu H, Kirk AD. Immunomodulation of T cell-mediated alloimmunity by proximity to endothelial cells under the mammalian target of rapamycin blockade. Am J Transplant 2025; 25:284-301. [PMID: 39426498 DOI: 10.1016/j.ajt.2024.10.008] [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: 03/02/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Endothelial cells (ECs) are an initial barrier between vascularized organ allografts and the host immune system and are thus well positioned to initiate and influence alloimmune rejection. The mammalian target of rapamycin inhibitor rapamycin is known to inhibit T cell activation and attenuate acute allograft rejection. It also has numerous effects on ECs. We hypothesized that A mammalian target of rapamycin blockade might directly alter EC alloimmunogenicity and reduce alloimmune responses independent of its effects on T cell function. Here we report that rapamycin treatment modulates EC coinhibitory ligand expression and alters cytokine/chemokine production. It alters the EC transcriptome broadly associated with negative regulation of immune responses. Rapamycin-treated ECs suppress EC-specific T cell proliferation independent of programmed cell death 1/programmed death-ligand interaction and inhibit T cells responding to adjacent allogeneic cells in a contact-independent manner via secreted inhibitory mediators above 10 kDa. The T cell hyporesponsiveness induced by rapamycin-pretreated ECs was rescued by exogenous interleukin 2. Preexposing donor hearts to rapamycin improves the effect of B7 costimulation blockade in prolonging heart allograft survival in a major histocompatibility complex-mismatched mouse model. Our results indicate that rapamycin-treated ECs have reduced alloimmunogenicity and created a local, contact-independent environment that limits T cell alloreactivity via anergy induction and improves the efficacy of B7 costimulation blockade.
Collapse
Affiliation(s)
- Shu Li
- Division of Transplant and Immunobiology Research, Departments of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Liuyang Wang
- Departments of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Victoria A Bendersky
- Division of Transplant and Immunobiology Research, Departments of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Qimeng Gao
- Division of Transplant and Immunobiology Research, Departments of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jun Wang
- Division of Transplant and Immunobiology Research, Departments of Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - He Xu
- Division of Transplant and Immunobiology Research, Departments of Surgery, Duke University School of Medicine, Durham, North Carolina, USA.
| | - Allan D Kirk
- Division of Transplant and Immunobiology Research, Departments of Surgery, Duke University School of Medicine, Durham, North Carolina, USA; Departments of Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
48
|
Lee D, Kozurek EC, Abdullah M, Wong EJ, Li R, Liu ZS, Nguyen HD, Dickerson EB, Kim JH. PIK3CA mutation fortifies molecular determinants for immune signaling in vascular cancers. Cancer Gene Ther 2025; 32:254-267. [PMID: 39709507 PMCID: PMC11839470 DOI: 10.1038/s41417-024-00867-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
Angiosarcomas are a group of vascular cancers that form malignant blood vessels. These malignancies are seemingly inflamed primarily due to their pathognomonic nature, which consists of irregular endothelium and tortuous blood channels. PIK3CA mutations are oncogenic and disrupt the PI3K pathway. In this study, we aimed to define the molecular and functional consequences of oncogenic PIK3CA mutations in angiosarcoma. We first generated two isogenic hemangiosarcoma cell lines harboring the H1047R hotspot mutations in PIK3CA gene using CRISPR/Cas9. We found PIK3CA-mutant cells established distinct molecular signatures in global gene expression and chromatin accessibility, which were associated with enrichment of immune cytokine signaling, including IL-6, IL-8, and MCP-1. These molecular processes were disrupted by the PI3K-α specific inhibitor, alpelisib. We also observed that the molecular distinctions in PIK3CA-mutant cells were linked to metabolic reprogramming in glycolytic activity and mitochondrial respiration. Our multi-omics analysis revealed that activating PIK3CA mutations regulate molecular machinery that contributes to phenotypic alterations and resistance to alpelisib. Furthermore, we identified potential therapeutic vulnerabilities of PIK3CA mutations in response to PI3K-α inhibition mediated by MAPK signaling. In summary, we demonstrate that PIK3CA mutations perpetuate PI3K activation and reinforce immune enrichment to promote drug resistance in vascular cancers.
Collapse
Affiliation(s)
- Donghee Lee
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Emma C Kozurek
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN, USA
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Md Abdullah
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Ethan J Wong
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN, USA
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Rong Li
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Zhiyan Silvia Liu
- Department of Pharmacology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Hai Dang Nguyen
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Pharmacology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Erin B Dickerson
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, MN, USA
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Jong Hyuk Kim
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- Artificial Intelligence Academic Initiative (AI2) Center, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
49
|
Apsel Winger B, Dowd CF, Shimano KA, Devine WP, Mathes E, Frieden I, Schaefer C, Kothari A. Effective Use of ALK Inhibitors in EML4::ALK-Positive Lymphatic Malformations. Pediatr Blood Cancer 2025; 72:e31441. [PMID: 39529238 DOI: 10.1002/pbc.31441] [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: 08/01/2024] [Revised: 10/08/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Genetically targeted medications are emerging as important therapies for lymphatic malformations (LMs) unresponsive to sirolimus. We describe two patients with EML4::ALK-positive LMs, one with Gorham Stout disease and one with a large genitourinary (GU) LM, who were successfully treated with ALK inhibitors. This report adds ALK inhibitors to the growing toolbox of molecularly targeted therapies for LMs.
Collapse
Affiliation(s)
- Beth Apsel Winger
- Department of Clinical Pharmacy, UCSF, San Francisco, California, USA
- Division of Pediatric Hematology, UCSF, San Francisco, California, USA
| | - Christopher F Dowd
- Division of Interventional Neuroradiology, UCSF, San Francisco, California, USA
| | - Kristin A Shimano
- Division of Pediatric Hematology, UCSF, San Francisco, California, USA
| | - W Patrick Devine
- Department of Anatomic Pathology, UCSF, San Francisco, California, USA
| | - Erin Mathes
- Department of Dermatology, UCSF, San Francisco, California, USA
| | - Ilona Frieden
- Department of Dermatology, UCSF, San Francisco, California, USA
| | - Carrie Schaefer
- Department of Radiology, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Alok Kothari
- Division of Pediatric Hematology/Oncology, Phoenix Children's Hospital, Phoenix, Arizona, USA
| |
Collapse
|
50
|
Ashayeri Ahmadabad H, Mohammadi Panah S, Ghasemnejad-Berenji H, Ghojavand S, Ghasemnejad-Berenji M, Khezri MR. Metformin and the PI3K/AKT signaling pathway: implications for cancer, cardiovascular, and central nervous system diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1035-1055. [PMID: 39225830 DOI: 10.1007/s00210-024-03358-3] [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: 04/15/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Recent findings have brought our understanding of diseases at the molecular level, highlighting upstream intracellular pathways as potential therapeutic targets. The PI3K/AKT pathway, a key regulator of cellular responses to environmental changes, is frequently altered in various diseases, making it a promising target for intervention. Metformin is the most known anti-diabetic agent that is known due to its effects on cancer, inflammatory-related diseases, oxidative stress, and other human diseases. It is clearly understood that metformin modulates the activity of the PI3K/AKT pathway leading to a wide variety of outcomes. This interaction has been well-studied in various diseases. Therefore, this review aims to examine PI3K/AKT-modulating properties of metformin in cancer, cardiovascular, and central nervous system diseases. Our findings indicate that metformin is effective in treating cancer and CNS diseases, and plays a role in both the prevention and treatment of cardiovascular diseases. These insights support the potential of metformin in comprehensive strategies for disease management.
Collapse
Affiliation(s)
| | | | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Shabnam Ghojavand
- Faculty of Pharmacy, Islamic Azad University of Tehran, Tehran, Iran
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.
- Research Center for Experimental and Applied Pharmaceutical Sciences, Urmia University of Medical Sciences, Urmia, Iran.
| | - Mohammad Rafi Khezri
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|