51
|
Hade MD, Butsch BL, Palacio PL, Nguyen KT, Shantaram D, Noria S, Brethauer SA, Needleman BJ, Hsueh W, Reategui E, Magana SM. Human differentiated adipocytes can serve as surrogate mature adipocytes for adipocyte-derived extracellular vesicle analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636729. [PMID: 39974962 PMCID: PMC11839020 DOI: 10.1101/2025.02.05.636729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Obesity is a growing global health concern, contributing to diseases such as cancer, autoimmune disorders, and neurodegenerative conditions. Adipose tissue dysfunction, characterized by abnormal adipokine secretion and chronic inflammation, plays a key role in these conditions. Adipose-derived extracellular vesicles (ADEVs) have emerged as critical mediators in obesity-related diseases. However, the study of mature adipocyte-derived EVs (mAdipo-EVs) is limited due to the short lifespan of mature adipocytes in culture, low EV yields, and the low abundance of these EV subpopulations in the circulation. Additionally, most studies rely on rodent models, which have differences in adipose tissue biology compared to humans. To overcome these challenges, we developed a standardized approach for differentiating human preadipocytes (preAdipos) into mature differentiated adipocytes (difAdipos), which produce high-yield, human adipocyte EVs (Adipo-EVs). Using visceral adipose tissue from bariatric surgical patients, we isolated the stromal vascular fraction (SVF) and differentiated preAdipos into difAdipos. Brightfield microscopy revealed that difAdipos exhibited morphological characteristics comparable to mature adipocytes (mAdipos) directly isolated from visceral adipose tissue, confirming their structural similarity. Additionally, qPCR analysis demonstrated decreased preadipocyte markers and increased mature adipocyte markers, further validating successful differentiation. Functionally, difAdipos exhibited lipolytic activity comparable to mAdipos, supporting their functional resemblance to native adipocytes. We then isolated preAdipo-EVs and difAdipo-EVs using tangential flow filtration and characterized them using bulk and single EV analysis. DifAdipo-EVs displayed classical EV and adipocyte-specific markers, with significant differences in biomarker expression compared to preAdipo-EVs. These findings demonstrate that difAdipos serve as a reliable surrogate for mature adipocytes, offering a consistent and scalable source of adipocyte-derived EVs for studying obesity and its associated disorders. Keywords: extracellular vesicles, adipocyte, adipose, adipocyte-derived extracellular vesicles, obesity.
Collapse
|
52
|
Zhao Y, Gao L, Chen J, Wei J, Lin G, Hu K, Zhao W, Wei W, Huang W, Gao L, Yuan A, Qian K, Chen AF, Pu J. Remote limb ischemic conditioning alleviates steatohepatitis via extracellular vesicle-mediated muscle-liver crosstalk. Cell Metab 2025; 37:886-902.e7. [PMID: 40118054 DOI: 10.1016/j.cmet.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 08/30/2024] [Accepted: 02/25/2025] [Indexed: 03/23/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is an advanced form of liver disease with adverse outcomes. Manipulating interorgan communication is considered a promising strategy for managing metabolic disease, including steatohepatitis. Here, we report that remote limb ischemic conditioning (RIC), a clinically validated therapy for distant organ protection by transient muscle ischemia, significantly alleviated steatohepatitis in different mouse models. The beneficial effect of limb ischemic conditioning was mediated by muscle-to-liver transfer of small extracellular vesicles (sEVs) and their cargo microRNAs, leading to elevation of miR-181d-5p in the liver. Hepatic miR-181d-5p overexpression faithfully mirrored the molecular and histological benefits of limb ischemic conditioning by suppressing nuclear receptor 4A3 (NR4A3). Furthermore, circulating EVs from human volunteers undergoing limb ischemic conditioning improved steatohepatitis and transcriptomic perturbations in primary human hepatocytes and animal models. Our data underscore the translational potential of limb ischemic conditioning for steatohepatitis management and extend our understanding of muscle-liver crosstalk.
Collapse
Affiliation(s)
- Yichao Zhao
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Gao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Jianqing Chen
- Graduate School of Bengbu Medical College, Bengbu, Anhui, China
| | - Jingze Wei
- Graduate School of Bengbu Medical College, Bengbu, Anhui, China
| | - Guanqiao Lin
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kewei Hu
- Graduate School of Bengbu Medical College, Bengbu, Anhui, China
| | - Wubin Zhao
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Huang
- Department of Radiation Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingchen Gao
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ancai Yuan
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Qian
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; School of Biomedical Engineering, Institute of Medical Robotics and Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Division of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Graduate School of Bengbu Medical College, Bengbu, Anhui, China.
| |
Collapse
|
53
|
Liu Y, Zhou R, Guo Y, Hu B, Xie L, An Y, Wen J, Liu Z, Zhou M, Kuang W, Xiao Y, Wang M, Xie G, Zhou H, Lu R, Peng H, Huang Y. Muscle-derived small extracellular vesicles induce liver fibrosis during overtraining. Cell Metab 2025; 37:824-841.e8. [PMID: 39879982 DOI: 10.1016/j.cmet.2024.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/24/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025]
Abstract
The benefits of exercise for metabolic health occur in a dose-dependent manner. However, the adverse effects of overtraining and their underlying mechanisms remain unclear. Here, we show that overtraining induces hepatic fibrosis. Mechanistically, we find that excessive lactate accumulation in skeletal muscle leads to the lactylation of SH3 domain-containing 3 (SORBS3), triggering its liquid-liquid phase separation (LLPS). LLPS of SORBS3 enhances its interaction with flotillin 1 and selectively facilitates the sorting of F-box protein 2 (FBXO2) into small extracellular vesicles, referred to as "lactate bodies." Lactate bodies induce hepatocyte apoptosis followed by hepatic stellate cell activation via myeloid cell leukemia sequence 1 (MCL1)-BAX/BAK signaling. Inhibition of SORBS3 lactylation or FBXO2 disrupts lactate bodies formation and alleviates overtraining-triggered liver fibrosis. Likewise, reduction of muscle lactate bodies formation by salidroside attenuates overtraining-induced liver fibrosis. Collectively, we identify a process by which overtraining induces hepatic fibrosis, highlighting a potential therapeutic target for liver health.
Collapse
Affiliation(s)
- Ya Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Rui Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Yifan Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Lingqi Xie
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Yuze An
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Jie Wen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Zheyu Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Min Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Weihong Kuang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Yao Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Min Wang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Genqing Xie
- Department of Endocrinology, The First People's Hospital of Xiangtan City, 411100 Xiangtan, Hunan, China
| | - Haiyan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China
| | - Renbin Lu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China.
| | - Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China.
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 410008 Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 410008 Changsha, Hunan, China; FuRong Laboratory, 410078 Changsha, Hunan, China.
| |
Collapse
|
54
|
Hong Y, Ye M, Wang J, Huang L. Stem Cell-Derived Extracellular Vesicles for Acute Pancreatitis: a Systematic Review and Meta-analysis of Preclinical Studies. Stem Cell Rev Rep 2025; 21:767-778. [PMID: 39964640 PMCID: PMC11965186 DOI: 10.1007/s12015-025-10852-5] [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] [Accepted: 02/06/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Several studies have reported the effectiveness of stem cell-derived extracellular vesicles (SC-EVs) in disease treatment. However, the efficacy of SC-EVs for severe acute pancreatitis (SAP) remains uncertain. This systematic review aimed to analyze and evaluate the effect of SC-EVs in the treatment of SAP in animal models by summarizing data from published studies. METHODS We searched Pubmed, Embase, and Web of Science databases to identify preclinical studies investigating the therapeutic effect of SC-EVs on SAP. The primary outcome was the histopathological scores of pancreatic tissues, including inflammation, edema, and necrosis. Other outcome measures included levels of amylase, IL-6, IL-10, and TNF-α. Eligible studies were selected based on the inclusion and exclusion criteria. SYRCLE checklist was adopted to assess the quality and bias risks of included studies. Mean differences and 95% confidence intervals were calculated using the inverse variance method with a random effects model. All statistical analyses were performed using RevMan 5.3 software. RESULTS A total of 8 studies including 126 animals were included. The results of meta-analysis revealed that SC-EVs treatment significantly reduced pancreatic histopathologic scores (total score: MD = -5.17, 95% CI: -5.79, -4.55; inflammation score: MD = -1.44, 95% CI: -1.70, -1.19; edema score: MD = -1.42, 95% CI: -1.75, -1.09; necrosis score: MD = -1.42, 95% CI: -1.80, -1.04), inhibited pro-inflammatory factor release (IL-6: SMD = -3.20, 95% CI: -4.51, -1.88; TNF-α SMD = -5.18, 95% CI: -6.96, -3.40), and enhancing the release of anti-inflammatory factors (IL-10 SMD = 4.15, 95% CI: 2.49, 5.81). Further subgroup analyses displayed SC-EVs treatment obviously attenuated animal pancreatic pathologic injury in traumatic pancreatitis and drug-induced acute pancreatitis, and the effect of SC-EVs to inhibit TNF-α secretion in the drug-induced SAP model was correlated with the dose of SC-EVs injection. CONCLUSIONS This meta-analysis displayed that SC-EVs were correlated with SAP injury alleviation and pancreas function reservation. Research into the treatment of SAP with SC-EVs is still in its early stage, necessitating further comprehensive investigations in the future to elucidate the therapeutic mechanisms of SC-EVs and their potential application in SAP.
Collapse
Affiliation(s)
- Yinghui Hong
- Department of Intensive Care, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, P. R. China
| | - Mingliang Ye
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Junshi Wang
- Department of Intensive Care, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, P. R. China
| | - Lei Huang
- Department of Intensive Care, Peking University Shenzhen Hospital, Shenzhen, 518036, P. R. China.
- Department of Emergency, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, P. R. China.
| |
Collapse
|
55
|
Li L, Liu Y, Wang K, Mo J, Weng Z, Jiang H, Jin C. Stem cell exosomes: new hope and future potential for relieving liver fibrosis. Clin Mol Hepatol 2025; 31:333-349. [PMID: 39510097 PMCID: PMC12016649 DOI: 10.3350/cmh.2024.0854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024] Open
Abstract
Liver fibrosis is a chronic liver injury resulting from factors like viral hepatitis, autoimmune hepatitis, non-alcoholic steatohepatitis, fatty liver disease, and cholestatic liver disease. Liver transplantation is currently the gold standard for treating severe liver diseases. However, it is limited by a shortage of donor organs and the necessity for lifelong immunosuppressive therapy. Mesenchymal stem cells (MSCs) can differentiate into various liver cells and enhance liver function when transplanted into patients due to their differentiation and proliferation capabilities. Therefore, it can be used as an alternative therapy for treating liver diseases, especially for liver cirrhosis, liver failure, and liver transplant complications. However, due to the potential tumorigenic effects of MSCs, researchers are exploring a new approach to treating liver fibrosis using extracellular vesicles (exosomes) secreted by stem cells. Many studies show that exosomes released by stem cells can promote liver injury repair through various pathways, contributing to the treatment of liver fibrosis. In this review, we focus on the molecular mechanisms by which stem cell exosomes affect liver fibrosis through different pathways and their potential therapeutic targets. Additionally, we discuss the advantages of exosome therapy over stem cell therapy and the possible future directions of exosome research, including the prospects for clinical applications and the challenges to be overcome.
Collapse
Affiliation(s)
- Lihua Li
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Yongjie Liu
- Department of Cell biology, School of Medicine, Taizhou University, Taizhou, Zhejiang Province, P. R. China
- Department of Pathophysiology, School of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning Province, P. R. China
| | - Kunpeng Wang
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Jinggang Mo
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Zhiyong Weng
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Hao Jiang
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Chong Jin
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| |
Collapse
|
56
|
Luo Q, Yang J, Yang M, Wang Y, Liu Y, Liu J, Kalvakolanu DV, Cong X, Zhang J, Zhang L, Guo B, Duo Y. Utilization of nanotechnology to surmount the blood-brain barrier in disorders of the central nervous system. Mater Today Bio 2025; 31:101457. [PMID: 39896289 PMCID: PMC11786670 DOI: 10.1016/j.mtbio.2025.101457] [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: 06/28/2024] [Revised: 10/27/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Central nervous system (CNS) diseases are a major cause of disability and death worldwide. Due to the blood-brain barrier (BBB), drug delivery for CNS diseases is extremely challenging. Nano-delivery systems can overcome the limitations of BBB to deliver drugs to the CNS, improve the ability of drugs to target the brain and provide potential therapeutic methods for CNS diseases. At the same time, the choice of different drug delivery methods (bypassing BBB or crossing BBB) can further optimize the therapeutic effect of the nano-drug delivery system. This article reviews the different methods of nano-delivery systems to overcome the way BBB enters the brain. Different kinds of nanoparticles to overcome BBB were discussed in depth.
Collapse
Affiliation(s)
- Qian Luo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Jiaying Yang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Mei Yang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yingtong Wang
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Yiran Liu
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Jixuan Liu
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Dhan V. Kalvakolanu
- Greenebaum NCI Comprehensive Cancer Center, Department of Microbiology and Immunology University of Maryland School Medicine, Baltimore, MD, USA
| | - Xianling Cong
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Jinnan Zhang
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Ling Zhang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Yanhong Duo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
57
|
Chen Y, Li J, Zhang X, Li S, Cheng Y, Fu X, Li J, Zhu L. Mesenteric adipose-derived exosomal TINAGL1 enhances intestinal fibrosis in Crohn's Disease via SMAD4. J Adv Res 2025; 70:139-158. [PMID: 38750695 PMCID: PMC11976418 DOI: 10.1016/j.jare.2024.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/23/2024] [Accepted: 05/11/2024] [Indexed: 03/23/2025] Open
Abstract
INTRODUCTION Crohn's Disease (CD) is a chronic inflammatory condition characterized by intestinal fibrosis, severely impacting patient quality of life. The molecular mechanisms driving this fibrosis remain inadequately understood. Recent evidence implicates mesenteric adipose tissue (MAT) in CD pathogenesis, particularly through its exosome secretion, which may influence fibrogenic pathways. Understanding the role of MAT-derived exosomes is crucial for unraveling these molecular processes. OBJECTIVES This study aims to elucidate the role of MAT-derived exosomes in CD-related intestinal fibrosis. We focus on investigating their molecular composition and the potential impact on fibrosis progression, with an emphasis on identifying novel therapeutic targets. METHODS We induced chronic intestinal inflammation in mice using dinitrobenzene sulfonic acid (DNBS), simulating CD-like fibrosis. Exosomes were isolated from DNBS-treated mice (MG) and normal controls (NG) for characterization using electron microscopy and proteomic analysis. Additionally, human colonic fibroblasts were exposed to exosomes from CD patients and healthy individuals, with subsequent assessment of fibrogenesis through proteomic and RNA sequencing analyses. RESULTS Proteomic analyses revealed a significant activation of the TGF-β signaling pathway in MG-treated mice compared to controls, correlating with enhanced intestinal fibrosis. In vitro experiments demonstrated that colonic fibroblasts exposed to CD patient-derived exosomes exhibited increased fibrogenic activity. Protein docking and co-immunoprecipitation studies suggested a critical interaction between TINAGL1 and SMAD4, enhancing fibrosis. Importantly, in vivo experiments corroborated that recombinant TINAGL1 protein exacerbated DNBS-induced intestinal fibrosis. CONCLUSION Our findings highlight the pivotal role of MAT-derived exosomes, particularly those carrying TINAGL1, in the progression of intestinal fibrosis in CD. The involvement of the TGF-β signaling pathway, especially the SMAD4 protein, offers new insights into the molecular mechanisms of CD-related fibrosis and presents potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Yidong Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junrong Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaopeng Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuang Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yiyu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoyu Fu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiamin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liangru Zhu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
58
|
Hsu CY, Altalbawy FMA, Oghenemaro EF, Uthirapathy S, Chandra M, Nathiya D, Kaur P, Ravi Kumar M, Kadhim AJ, Kariem M. Exosomal lncRNAs in the Tumor Angiogenesis: As Therapeutic Targets in Cancer Treatment. Arch Pharm (Weinheim) 2025; 358:e202400940. [PMID: 40165644 DOI: 10.1002/ardp.202400940] [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: 12/12/2024] [Revised: 02/12/2025] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
Exosomes, as mediators of intercellular communication, can be released from different types of cells and regulate the function of the target cell by transferring cargo, such as proteins, DNA, and RNA. Recent investigations have revealed a preponderance of long noncoding RNAs (lncRNAs), a subclass of noncoding RNAs, within exosomes, where they exhibit notable stability and are implicated in the development and progression of neoplastic processes, such as tumor angiogenesis. Angiogenesis, as a hallmark of cancer, provides diffusible nutrients and oxygen to the distant cells and guarantees tumorigenesis and metastasis. Exosomal lncRNAs, including MALAT1, OIP5-AS1, PART1, SNHG family, FAM225A, ATB, RAMP2-AS1, UCA1, TRPM2-AS, FGD5-AS1, and LINC0016, could modulate tumor angiogenesis by activating signaling cascades and mediators within the target cells, such as microRNAs (miRNAs). Regulation of tumor angiogenesis through modulation of exosomal lncRNAs could be a reliable strategy for cancer therapy. In this review, we discuss the characteristics and biogenesis of exosomes and lncRNAs and how exosomal lncRNAs are involved in various processes of tumorigenesis. Our primary focus is on exosomal lncRNAs, their impact on tumor angiogenesis, and their potential as novel diagnostic markers and therapeutic targets for various cancers.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, USA
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, Egypt
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Muktesh Chandra
- Marwadi University Research Center, Department of Bioinformatics, Faculty of Engineering and Technology, Marwadi University, Rajkot, Gujarat, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
59
|
Lee EC, Choi D, Lee DH, Oh JS. Engineering Exosomes for CNS Disorders: Advances, Challenges, and Therapeutic Potential. Int J Mol Sci 2025; 26:3137. [PMID: 40243901 PMCID: PMC11989722 DOI: 10.3390/ijms26073137] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
The development of targeted drugs for diseases of the central nervous system (CNS) is a significant challenge due to the structural complexity and functional specificities of these systems. Recently, exosomes have emerged as a promising therapeutic platform, given their unique capacity to traverse the blood-brain barrier and deliver bioactive molecules to target cells. This review examines recent advances in exosome research with a particular focus on CNS diseases, emphasizing their role as carriers of therapeutic cargo, including proteins, RNAs, and lipids. Nevertheless, significant challenges remain before exosome-based therapies can be translated from preclinical research to clinical applications. These include the need for scalable production and standardized isolation methods. Despite these hurdles, ongoing studies continue to shed light on the mechanisms of exosome-mediated neuroprotection and neurodegeneration. This paves the way for innovative therapeutic strategies to address CNS disorders.
Collapse
Affiliation(s)
- Eun Chae Lee
- Department of Medical Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Dongsic Choi
- Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea;
| | - Dong-Hun Lee
- Department of Neurosurgery, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 11765, Republic of Korea
| | - Jae Sang Oh
- Department of Medical Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea;
- Department of Neurosurgery, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 11765, Republic of Korea
| |
Collapse
|
60
|
McMullan E, Joladarashi D, Kishore R. Unpacking Exosomes: A Therapeutic Frontier for Cardiac Repair. Curr Cardiol Rep 2025; 27:73. [PMID: 40111702 PMCID: PMC11925971 DOI: 10.1007/s11886-025-02225-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE OF REVIEW The rising global prevalence of cardiovascular disease is driving the need for innovative biotherapeutics. Recently, exosomes-extracellular vesicles involved in paracrine signaling have shown promise in aiding heart repair associated with cardiovascular conditions. Their therapeutic potential encompasses several beneficial mechanisms, including anti-fibrosis, anti-inflammation, pro-angiogenesis, anti-oxidation, and anti-apoptosis, all contributing to improved cardiac function. This review provides a comprehensive overview of exosomes and highlights the latest research on their effectiveness in addressing current challenges in regenerative cardiac medicine. RECENT FINDINGS Current approaches revolve around elucidating and enhancing how different cell types, cargo, and delivery methods impact healing in a pathological cardiovascular environment. The emerging field of therapeutic exosome research is promising for cardiac regeneration due to the beneficial effects of exosomal cargo. The expansion of mechanistic knowledge and the optimization of techniques are required before standard clinical application.
Collapse
Affiliation(s)
- Elena McMullan
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Darukeshwara Joladarashi
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Raj Kishore
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
61
|
Lu L, Wang L, Wang H, Yang M. Regulatory role of miR-128-2-5p in serum exosomes on COL6A2 expression and postmenopausal osteoporosis. Hum Mol Genet 2025; 34:563-576. [PMID: 39817546 DOI: 10.1093/hmg/ddae147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/18/2024] [Indexed: 01/18/2025] Open
Abstract
This study investigates the influence of miR-128-2-5p within serum-derived exosomes (Exos) on COL6A2 expression and its implications in postmenopausal osteoporosis (POMP). Utilizing bioinformatics analysis, we identified 1317 differentially expressed genes (DEGs), primarily enriched in the focal adhesion pathway-a critical regulator of osteoblast adhesion. A significant gene, COL6A2, emerged as notably downregulated in POMP, possessing potential as a diagnostic marker. Predictive analysis linked the upstream miRNA miR-128-2-5p, highly enriched in Exos, with the regulation of COL6A2. Experimentally, Exos from POMP patients demonstrated elevated miR-128-2-5p levels, which inhibited COL6A2 expression in vitro, reducing osteoblast adhesion and exacerbating osteoporotic conditions. These findings highlight the pivotal role of exosomal miR-128-2-5p in bone metabolism, suggesting a novel molecular mechanism and a potential therapeutic target in POMP.
Collapse
Affiliation(s)
- Liangjie Lu
- Department of Orthopedics, Ningbo Medical Center Li Huili Hospital, Li Huili Hospital Affiliated to Ningbo University, No. 57 Xingning Road, Yinzhou District, Ningbo, Zhejiang 315040, China
| | - Lijun Wang
- Department of Pediatrics, The First Hospital of Jilin University, No. 71, Xinmin Street, Chaoyang District, Changchun City, Jilin Province 130021, China
| | - Huihan Wang
- Department of Orthopaedics, Zhengzhou Central Hospital Affiliated to Zhengzhou University, No. 195, Tongbai Road, Zhongyuan District, Zhengzhou City, South Province 450007, China
| | - Minjie Yang
- Department of Orthopaedics, Jiujiang No.1 People's Hospital, No. 48, Taling South Road, Xunyang District, Jiujiang City, Jiangxi Province 332000, China
| |
Collapse
|
62
|
Qiu D, Yan B, Xue H, Xu Z, Tan G, Liu Y. Perspectives of exosomal ncRNAs in the treatment of bone metabolic diseases: Focusing on osteoporosis, osteoarthritis, and rheumatoid arthritis. Exp Cell Res 2025; 446:114457. [PMID: 39986599 DOI: 10.1016/j.yexcr.2025.114457] [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: 10/23/2024] [Revised: 01/13/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Bone metabolic disorders, constituting a group of prevalent and grave conditions, currently have a scarcity of therapeutic alternatives. Over the recent past, exosomes have been at the forefront of research interest, owing to their nanoparticulate nature and potential for therapeutic intervention. ncRNAs are a class of heterogeneous transcripts that they lack protein-encoding capacity, yet they can modulate the expression of other genes through multiple mechanisms. Mounting evidence underscores the intricate role of exosomes as ncRNAs couriers implicated in the pathogenesis of bone metabolic disorders. In this review, we endeavor to elucidate recent insights into the roles of three ncRNAs - miRNAs, lncRNAs, and circRNAs - in bone metabolic ailments such as osteoporosis, osteoarthritis, and rheumatoid arthritis. Additionally, we examine the viability of exosomal ncRNAs as innovative, cell-free modalities in the diagnosis and therapeutic management of bone metabolic disorders. We aim to uncover the critical function of exosomal ncRNAs within the context of bone metabolic diseases.
Collapse
Affiliation(s)
- Daodi Qiu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Binghan Yan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Haipeng Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Zhanwang Xu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Guoqing Tan
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yajuan Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250300, China.
| |
Collapse
|
63
|
Satao KS, Doshi GM. Intercellular communication via exosomes: A new paradigm in the pathophysiology of neurodegenerative disorders. Life Sci 2025; 365:123468. [PMID: 39954940 DOI: 10.1016/j.lfs.2025.123468] [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: 10/22/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Neurodegenerative disorders are one of the leading causes of death and disability and pose a great economic burden on healthcare systems. Generally, these neurodegenerative disorders have a progressive deterioration in neural function and structure, and deposition of misfolded proteins commonly occurs, such as amyloid-β in AD and α-synuclein in PD. However, there exists a special class of exosomes, which acts like a transmitter and enhances communication between cells. The present review discusses the significant role of exosomes in neurodegenerative diseases, with a focus on Amyotrophic lateral Sclerosis (ALS), AD, PD, and Huntington's disease (HD). In this review, the biogenesis of exosomes is discussed from multivesicular bodies and onwards to their release into the extracellular environment. The present review focuses on recent data concerning the possible use of modified exosomes as ND therapy. Indeed, future work is needed to explain the processes driving exosome biogenesis and cargo selection, while opening new routes by the use of exosome-based therapeutics in neurodegenerative disease diagnosis and treatment.
Collapse
Affiliation(s)
- Kiran S Satao
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Department of Pharmacology, V. M. Road, Vile Parle (W), Mumbai 400 056, Maharashtra, India
| | - Gaurav M Doshi
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Department of Pharmacology, V. M. Road, Vile Parle (W), Mumbai 400 056, Maharashtra, India.
| |
Collapse
|
64
|
Cao C, Wang Y, Deng X, Zhao X, Chen Y, Tan W, Deng F, Li F. Exosomes containing miR-152-3p targeting FGFR3 mediate SLC7A7-induced angiogenesis in bladder cancer. NPJ Precis Oncol 2025; 9:71. [PMID: 40075158 PMCID: PMC11903784 DOI: 10.1038/s41698-025-00859-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Bladder cancer (BCa) is a prevalent malignancy with a poor prognosis. SLC7A7 has been linked to BCa progression and angiogenesis, but its specific role remains unclear. We established a SLC7A7-knockdown BCa cell line to investigate its effects on angiogenesis. In vivo experiments assessed tumor vascularization, while in vitro studies explored exosome involvement. MiRNA sequencing identified miR-152-3p as a key regulator. Further investigation using dual-luciferase reporter assays, qRT-PCR, and Western blot revealed that miR-152-3p inhibits the expression of FGFR3 by binding to its 3' UTR. Meanwhile, functional assays, including angiogenesis assays, Transwell assays, and wound healing assays, were performed to evaluate the effects of miR-152-3p on angiogenesis. We confirmed the significant role of SLC7A7 in BCa progression, specifically in promoting angiogenesis, through the involvement of exosomes and the regulatory axis of miR-152-3p/ FGFR3. Targeting FGFR3 might be a promising strategy to reverse control BCa progression for an improved prognosis.
Collapse
Affiliation(s)
- Chun Cao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yu Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaolin Deng
- Department of Urology, Ganzhou People's Hospital, Ganzhou, People's Republic of China
| | - Xinlei Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yuwen Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China.
| | - Fei Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
65
|
Yaman S, Devoe T, Aygun U, Parlatan U, Bobbili MR, Karim AH, Grillari J, Durmus NG. EV-Lev: extracellular vesicle isolation from human plasma using microfluidic magnetic levitation device. LAB ON A CHIP 2025; 25:1439-1451. [PMID: 39918033 DOI: 10.1039/d4lc00830h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
Biological nanomaterials have unique magnetic and density characteristics that can be employed to isolate them into subpopulations. Extracellular nanovesicles (EVs) are crucial for cellular communication; however, their isolation poses significant challenges due to their diverse sizes and compositions. We present EV-Lev, a microfluidic magnetic levitation technique for high-throughput, selective isolation of small EVs (<200 nm) from human plasma. EV-Lev overcomes the challenges posed by the subtle buoyancy characteristics of EVs, whose small size and varied densities complicate traditional magnetic levitation techniques. It employs antibody-coated polymer beads of varying densities, integrating immuno-affinity and microfluidics to isolate EVs from sub-milliliter plasma volumes efficiently. It facilitates rapid, simultaneous sorting of EV subpopulations based on surface markers, such as CD9, CD63, and CD81, achieving high yield and purity. Subsequent size and morphology analyses confirmed that the isolated EVs maintain their structural integrity. EV-Lev could help uncover the cargo and function of EV subpopulations associated with multiple diseases including cancer, infectious diseases and help to discover potential biomarkers in small volume samples, while offering a portable, cost-effective, and straightforward assay scheme.
Collapse
Affiliation(s)
- Sena Yaman
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305-5281, USA.
| | - Tessa Devoe
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305-5281, USA.
- Brown University, Providence, RI 02912, USA
| | - Ugur Aygun
- Canary Center for Cancer Early Detection, Department of Radiology, Stanford University, Stanford, CA 94304, USA
- Department of Electrical and Electronics Engineering, Koç University, Istanbul, Turkiye
| | - Ugur Parlatan
- Canary Center for Cancer Early Detection, Department of Radiology, Stanford University, Stanford, CA 94304, USA
| | - Madhusudhan Reddy Bobbili
- Canary Center for Cancer Early Detection, Department of Radiology, Stanford University, Stanford, CA 94304, USA
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, 1190 Wien, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Wien, Austria
| | - Asma H Karim
- Canary Center for Cancer Early Detection, Department of Radiology, Stanford University, Stanford, CA 94304, USA
| | - Johannes Grillari
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, 1190 Wien, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Wien, Austria
| | - Naside Gozde Durmus
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305-5281, USA.
| |
Collapse
|
66
|
Wu Q, Wu Q, Lin H, Zhang C, You Z, Kang S, Xu Y, Chen X, Yang C, Song Y, Zhu L. Microfluidic Replication and Phenotypic Profiling of Extracellular Vesicles from the Tumor Microenvironment Using Dual-Switch Aptamer Logic Gates. Anal Chem 2025; 97:5313-5323. [PMID: 40012368 DOI: 10.1021/acs.analchem.5c00234] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The phenotypic profiling of extracellular vesicles (EVs) within the tumor microenvironment (TME) provides critical insights into the intercellular communication mechanisms of EVs underlying tumor physiology. However, conventional methods typically isolate EVs from the extracellular space through tissue fragmentation, which compromises tissue viability, and neglects the spatial organization of the tissue and the dynamic nature of EV secretion. Herein, we introduce an innovative microfluidic platform to cultivate intact tumor tissues while preserving their spatial architecture and facilitating natural EV secretion. This system enables the direct replication of EVs onto the chip for high-fidelity phenotypic analysis. Utilizing a combinatorial-aptamer-induced dual-switch logic gate methodology, this approach allows for the precise subtyping of EVs derived from both tumor cells and immune cells within the TME. Specifically, aptamers targeting EpCAM and PD-L1, along with the connector probe, were employed to induce a dual-switch signal to identify distinct EV populations. This strategy enables noninvasive, real-time capture and phenotypic profiling of EVs directly within the microfluidic environment. Furthermore, our findings indicate that immunotherapy with PD-1 antibodies significantly enhances the secretion of EVs by immune cells within the TME, underscoring the potential role of EVs as mediators of therapeutic responses. Overall, we have developed a robust, noninvasive method for the phenotypic profiling of EVs in the TME, offering a powerful tool for investigating the biological functions and implications of EVs in tumor pathophysiology.
Collapse
Affiliation(s)
- Qiaoyi Wu
- Department of Trauma Center and Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Department of Trauma Center & Emergency Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, P. R. China
| | - Qiuyue Wu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Haoting Lin
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Chi Zhang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Zhenlong You
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Siyin Kang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Yuanfeng Xu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Xiaofeng Chen
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
- School of Environmental Science and Engineering, Hainan University, Haikou 570228, P. R. China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Yanling Song
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Lin Zhu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
- School of Environmental Science and Engineering, Hainan University, Haikou 570228, P. R. China
| |
Collapse
|
67
|
Puagsopa J, Tongviseskul N, Jaroentomeechai T, Meksiriporn B. Recent Progress in Developing Extracellular Vesicles as Nanovehicles to Deliver Carbohydrate-Based Therapeutics and Vaccines. Vaccines (Basel) 2025; 13:285. [PMID: 40266147 PMCID: PMC11946770 DOI: 10.3390/vaccines13030285] [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: 01/31/2025] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
Cell-derived, nanoscale extracellular vesicles (EVs) have emerged as promising tools in diagnostic, therapeutic, and vaccine applications. Their unique properties including the capability to encapsulate diverse molecular cargo as well as the versatility in surface functionalization make them ideal candidates for safe and effective vehicles to deliver a range of biomolecules including gene editing cassettes, therapeutic proteins, glycans, and glycoconjugate vaccines. In this review, we discuss recent advances in the development of EVs derived from mammalian and bacterial cells for use in a delivery of carbohydrate-based protein therapeutics and vaccines. We highlight key innovations in EVs' molecular design, characterization, and deployment for treating diseases including Alzheimer's disease, infectious diseases, and cancers. We discuss challenges for their clinical translation and provide perspectives for future development of EVs within biopharmaceutical research and the clinical translation landscape.
Collapse
Affiliation(s)
- Japigorn Puagsopa
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Niksa Tongviseskul
- Department of Biology, School of Science, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| | - Thapakorn Jaroentomeechai
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Bunyarit Meksiriporn
- Department of Biology, School of Science, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| |
Collapse
|
68
|
Ma S, Wang Y, Fan S, Jiang W, Sun M, Jing M, Bi W, Zhou M, Wu D. TSH-stimulated hepatocyte exosomes modulate liver-adipose triglyceride accumulation via the TGF-β1/ATGL axis in mice. Lipids Health Dis 2025; 24:81. [PMID: 40050912 PMCID: PMC11884018 DOI: 10.1186/s12944-025-02509-6] [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: 01/09/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025] Open
Abstract
Subclinical hypothyroidism (SCH) contributes to obesity, with the liver acting as a crucial metabolic regulator. Thyroid-stimulating hormone (TSH) affects systemic lipid balance, potentially linking SCH to obesity. While the direct impact of TSH on hepatic lipid metabolism has been extensively documented, its role in modulating lipid dynamics in peripheral organs through liver-mediated pathways remains insufficiently understood. This study identifies TSH-stimulated hepatocyte-derived exosomes (exosomesTSH) as key mediators in liver-adipose communication, promoting triglyceride accumulation in adipocytes via the transforming growth factor-beta 1 (TGF-β1)/adipose triglyceride lipase (ATGL) axis. ExosomesTSH enhance lipid storage in adipocytes, significantly increasing triglyceride content and lipid droplet formation while reducing lipolysis, effects that are dependent on TSH receptor (TSHR) activation in hepatocytes. In vivo, exosomesTSH induce weight gain and adipose tissue expansion, impairing glucose metabolism in both chow- and high-fat diet-fed mice. Mechanistically, exosomesTSH upregulate TGF-β1 and downregulate ATGL in adipocytes, establishing the TGF-β1/ATGL pathway as essential for exosome-mediated lipid accumulation. Further, miR-139-5p is identified as a modulator of TGF-β1 expression within this pathway, with overexpression of miR-139-5p alleviating exosomesTSH-induced lipid accumulation in adipocytes. This study elucidates a novel miR-139-5p-dependent mechanism through which TSH modulates lipid metabolism via liver-derived exosomes, highlighting the pivotal role of miR-139-5p in linking SCH to adipose lipid accumulation through the TGF-β1/ATGL signaling axis.
Collapse
Affiliation(s)
- Shizhan Ma
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Yayun Wang
- Department of Neurology, Qingdao Traditional Chinese Medicine Hospital Affiliated of Qingdao University, Qingdao, Shandong, 266033, China
| | - Shuteng Fan
- Department of Nursing, Taishan Vocational College of Nursing, Taian, Shandong, 271000, China
| | - Wanli Jiang
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Mingliang Sun
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Mengzhe Jing
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Wenkai Bi
- Department of Nuclear Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Meng Zhou
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| | - Dongming Wu
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China.
| |
Collapse
|
69
|
Feix AS, Laimer-Digruber A, Cruz-Bustos T, Steiner G, Ruttkowski B, Ehling-Schulz M, Joachim A. Variations in extracellular vesicle shedding of Cystoisospora suis stages (Apicomplexa: Coccidia). Int J Parasitol 2025; 55:197-212. [PMID: 39793881 DOI: 10.1016/j.ijpara.2025.01.001] [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: 06/12/2024] [Revised: 11/21/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025]
Abstract
Cystoisospora suis, a porcine enteral parasite of the order Coccidia, is characterized by a complex life cycle, with asexual and sexual development in the epithelium of the host gut and an environmental phase as an oocyst. All developmental stages vary greatly in their morphology and function, and therefore excrete different bioactive molecules for intercellular communication. Due to their complex development, we hypothesized that the extracellular vesicles (EVs) cargo is highly dependent on the life cycle stages from which they are released. This study aimed to characterize and compare EVs of all developmental stages of C. suis. Nanoparticle tracking analysis and microscopy were used to determine particle numbers and size distributions of stage-specific parasite EVs. Furthermore, Fourier-transform infrared spectral analysis was employed for the metabolic fingerprinting of EVs, and the lipid and protein profiles of all parasite stages were determined. Overall, the study revealed that asexual, sexual and transmissible stages of C. suis release different EVs during the parasite's life cycle. EVs of endogenous asexual and sexual stages were found to be more similar to each other than to those of the transmissible environmental stage, the oocyst. Furthermore, the ratio of fatty acids to polysaccharides and proteins changed during parasite development. In particular, proteins associated with the Apicomplexa and those involved in vesicle shedding showed changes in expression in all parasite stages. Lipid analysis showed that fatty acids were found in the same concentration through all parasite stages, whereas the amount of stereolipids, sphingolipids and glycerolipids changed between the parasite stages. In conclusion, this study, which presents the first known characterization of C. suis EVs, demonstrates a link between EVs and the respective developmental stages of the parasite, and putative functions in the parasite-parasite and host-parasite interplays.
Collapse
Affiliation(s)
- Anna Sophia Feix
- Institute of Parasitology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1 A-1210 Vienna, Austria.
| | - Astrid Laimer-Digruber
- Institute of Microbiology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine, Veterinärplatz 1 A-1210 Vienna, Austria
| | - Teresa Cruz-Bustos
- Institute of Parasitology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1 A-1210 Vienna, Austria
| | - Gerhard Steiner
- Department of Integrative Zoology, Faculty of Life Sciences, University of Vienna, Djerassiplatz 1 1030 Vienna, Austria
| | - Bärbel Ruttkowski
- Institute of Parasitology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1 A-1210 Vienna, Austria
| | - Monika Ehling-Schulz
- Institute of Microbiology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine, Veterinärplatz 1 A-1210 Vienna, Austria
| | - Anja Joachim
- Institute of Parasitology, Department for Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1 A-1210 Vienna, Austria
| |
Collapse
|
70
|
Yan L, Cao Y, Hou L, Luo T, Li M, Gao S, Wang L, Sheng K, Zheng L. Ginger exosome-like nanoparticle-derived miRNA therapeutics: A strategic inhibitor of intestinal inflammation. J Adv Res 2025; 69:1-15. [PMID: 38588850 PMCID: PMC11954804 DOI: 10.1016/j.jare.2024.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024] Open
Abstract
INTRODUCTION MicroRNAs (miRNAs) involve in destabilising messenger RNA or repressing translation of target molecules. Ginger-derived exosome-like nanoparticles (GELNs) play a crucial role in modulating intestinal inflammation. Moreover, GELNs contain highly heterogeneous miRNA. However, the role of miRNAs derived from GELNs in immunomodulation remains unclear. OBJECTIVES This study aimed to elucidate the molecular basis of the unique biological effects mediated by miRNA derived from GELNs on macrophages. METHODS GELNs were isolated using a combination of commercial exosome isolation kits and the differential centrifugation method, and the lipid composition of GELNs was determined using liquid chromatography-mass spectrometry. Subsequently, PKH26 labelled GELNs were taken up by macrophages. Furthermore, the modulation of inflammatory and immune responses by GELNs or osa-miR164d was assessed through the RNA-seq, RT-qPCR, online databases, and dual luciferase reporter assays to explore the underlying mechanisms of osa-miR164d. Biomimetic exosomes loaded with osa-miR164d were prepared using a microfluidic mixing device and systematically characterized. The therapeutic effects of osa-miR164d on relieving colitis were evaluated. RESULTS We report for the first time that GELNs-derived osa-miR164d is a regulatory factor of reprogramming macrophage polarization, thereby inhibiting the intestinal inflammatory response. Mechanistically, osa-miR164d directly targets the 3'-UTRs of TAB1, which regulates macrophage polarization through the downregulation of NF-κB expression. In addition, We have designed a biomimetic exosome mimicking GELNs to deliver osa-miR164d (osa-miR164d-MGELNs). Notably, the osa-miR164d-MGELNs can efficiently reprogram macrophages to alleviate colitis-related symptoms. CONCLUSION Our findings enhance the systematic understanding of how GELNs-derived osa-miR164d mediates cross-kingdom communication and provide an original engineering paradigm for mimicking GELNs to transfer miRNA.
Collapse
Affiliation(s)
- Ling Yan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-Process, Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yaqi Cao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Linhai Hou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Tianyu Luo
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Meiqi Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Shengjie Gao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Lei Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Kangliang Sheng
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China.
| | - Lei Zheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China; Engineering Research Center of Bio-Process, Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China.
| |
Collapse
|
71
|
Thomas MR, Badekila AK, Pai V, S N, Bhandary Y, Rai A, Kini S. Navigating Tumor Microenvironment Barriers with Nanotherapeutic Strategies for Targeting Metastasis. Adv Healthc Mater 2025; 14:e2403107. [PMID: 39840497 DOI: 10.1002/adhm.202403107] [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: 08/19/2024] [Revised: 12/20/2024] [Indexed: 01/23/2025]
Abstract
Therapeutic strategy for efficiently targeting cancer cells needs an in-depth understanding of the cellular and molecular interplay in the tumor microenvironment (TME). TME comprises heterogeneous cells clustered together to translate tumor initiation, migration, and proliferation. The TME mainly comprises proliferating tumor cells, stromal cells, blood vessels, lymphatic vessels, cancer-associated fibroblasts (CAFs), extracellular matrix (ECM), and cancer stem cells (CSC). The heterogeneity and genetic evolution of metastatic tumors can substantially impact the clinical effectiveness of therapeutic agents. Therefore, the therapeutic strategy shall target TME of all metastatic stages. Since the advent of nanotechnology, smart drug delivery strategies are employed to deliver effective drug formulations directly into tumors, ensuring controlled and sustained therapeutic efficacy. The state-of-the-art nano-drug delivery systems are shown to have innocuous modes of action in targeting the metastatic players of TME. Therefore, this review provides insight into the mechanism of cancer metastasis involving invasion, intravasation, systemic transport of circulating tumor cells (CTCs), extravasation, metastatic colonization, and angiogenesis. Further, the novel perspectives associated with current nanotherapeutic strategies are highlighted on different stages of metastasis.
Collapse
Affiliation(s)
- Mahima Rachel Thomas
- Nitte (Deemed to be University), Department of Bio & Nano Technology, Nitte University Centre for Science Education and Research, Mangalore, Karnataka, 575018, India
| | - Anjana Kaveri Badekila
- Nitte (Deemed to be University), Department of Bio & Nano Technology, Nitte University Centre for Science Education and Research, Mangalore, Karnataka, 575018, India
| | - Vishruta Pai
- Nitte (Deemed to be University), Department of Bio & Nano Technology, Nitte University Centre for Science Education and Research, Mangalore, Karnataka, 575018, India
| | - Nijil S
- Nitte (Deemed to be University), Department of Bio & Nano Technology, Nitte University Centre for Science Education and Research, Mangalore, Karnataka, 575018, India
| | - Yashodhar Bhandary
- Cell Biology and Molecular Genetics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka, 575 018, India
| | - Ankit Rai
- Medical Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India
| | - Sudarshan Kini
- Nitte (Deemed to be University), Department of Bio & Nano Technology, Nitte University Centre for Science Education and Research, Mangalore, Karnataka, 575018, India
| |
Collapse
|
72
|
Zhu L, Niu Q, Li D, Li M, Guo W, Han Z, Yang Y. Bone Marrow Mesenchymal Stem Cells-derived Exosomes Promote Survival of Random Flaps in Rats through Nrf2-mediated Antioxidative Stress. J Reconstr Microsurg 2025; 41:177-190. [PMID: 38782030 DOI: 10.1055/a-2331-8046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
BACKGROUND Random flaps are the most used defect repair method for head and neck tumors and trauma plastic surgery. The distal part of the flap often undergoes oxidative stress (OS), ultimately leading to flap necrosis. Stem cells' exosomes exhibit potential effects related to anti-inflammatory, regenerative, and antioxidant properties. Nuclear factor erythroid-2-related factor 2 (Nrf2) is an important factor in regulating oxidative balance. Exosomes have been reported to monitor its transcription to alleviate OS. This study examined the impacts and underlying mechanisms of antioxidant actions of exosomes derived from bone marrow mesenchymal stem cells (BMSCs-Exo) on random flaps. METHODS BMSCs-Exo were injected into the tail veins of rats on days 0, 1, and 2 after surgery of random flaps. The rats were euthanized on day 3 to calculate the survival rate. Immunohistochemical staining, western blotting, dihydroethidium probe, superoxide dismutase, and malondialdehyde assay kits were used to detect the OS level. Human umbilical vein endothelial cells were cocultured with BMSCs-Exo and ML385 (an inhibitor of Nrf2) in vitro. RESULTS BMSCs-Exo may significantly improve the survival rate of the random flaps by reducing apoptosis, inflammation, and OS while increasing angiogenesis. Besides, BMSCs-Exo can also increase mitochondrial membrane potential and reduce reactive oxygen species levels in vitro. These therapeutic effects might stem from the activation of the Kelch-like enyol-CoA hydratase (ECH)-associated protein 1 (Keap1)/Nrf2 signaling pathway. CONCLUSION BMSCs-Exo improved the tissue antioxidant capacity by regulating the Keap1/Nrf2 signaling pathway. BMSCs-Exo may be a new strategy to solve the problem of random flap necrosis.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Qifang Niu
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Delong Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Mozi Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wenwen Guo
- Department of Oral and Maxillofacial Surgery, Beijing Xing Ye Stomatological Hospital, Beijing, People's Republic of China
| | - Zhengxue Han
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yang Yang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
73
|
Yang X, Han H, Wang S, Teng F, Lv X, Zhang M, Zhang Y, Li W, Zhang M, Chen L, Zhang S. Islet-derived exosomal miR-204 accelerates insulin resistance in skeletal muscle by suppressing sirtuin 1: An in vivo study in a mouse model of high-fat diet-induced obesity. Diabetes Obes Metab 2025; 27:1153-1163. [PMID: 39632777 DOI: 10.1111/dom.16102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
AIMS The interaction between pancreatic islets and skeletal muscle plays a pivotal role in the development of insulin resistance. The present study aimed to elucidate the impact of non-hormonal molecules from islets on the insulin sensitivity of skeletal muscle cells. MATERIALS AND METHODS We developed a mouse model of obesity through a high-fat diet, assessing glucose tolerance and conducting miRNA sequencing on skeletal muscle samples. An in vitro model was established by treating cells with palmitic acid, and exosomes in the supernatant were characterized using scanning electron microscopy and CD63 expression analysis. Intracellular miR-204-5p levels were quantified by RT-PCR. RESULTS Our in vivo model demonstrated a robust correlation between miR-204-5p level alterations and obesity-induced insulin resistance. Elevated fatty acid levels were observed to increase miR-204-5p in both skeletal muscle and islets. In cellular studies, palmitic acid increased miR-204-5p in MIN-6 islet β-cells but not in C2C12 skeletal muscle cells. Exosomes containing miR-204-5p, secreted by palmitic acid-treated MIN6 cells, were identified through morphological examination, immunoblotting for the exosomal marker CD63, and intraexosomal miR-204-5p level measurement. C2C12 cells were shown to uptake islet-derived miR-204-5p exosomes, as evidenced by the uptake of Exo-Red labeled exosomes. TargetScan analysis identified a highly conserved binding site for miR-204-5p in the 3' UTR of Sirt mRNA. Functional studies indicated that miR-204-5p overexpression reduced glucose consumption and uptake in C2C12 cells, decreased Sirt expression, and impaired insulin signaling, as evidenced by reduced Akt phosphorylation and membrane Glut4 levels. CONCLUSIONS Our findings reveal that miR-204-5p contributes to the development of insulin resistance in obesity and acts as a signaling molecule in the crosstalk between pancreatic islets and skeletal muscle.
Collapse
Affiliation(s)
- Xuehan Yang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Hao Han
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Sushan Wang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Fei Teng
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xiaoyan Lv
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
- Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Meishuang Zhang
- Department of Pharmacology, School of Nursing, Jilin University, Changchun, Jilin, China
| | - Ying Zhang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Changchun, China
| | - Wei Li
- College of Life Sciences, Jilin Agricultural University, Changchun, Jilin, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
- Department of Pharmacology, School of Nursing, Jilin University, Changchun, Jilin, China
| | - Siwei Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
74
|
Yu Z, Fu J, Mantareva V, Blažević I, Wu Y, Wen D, Battulga T, Wang Y, Zhang J. The role of tumor-derived exosomal LncRNA in tumor metastasis. Cancer Gene Ther 2025; 32:273-285. [PMID: 40011710 DOI: 10.1038/s41417-024-00852-x] [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: 04/24/2024] [Revised: 10/22/2024] [Accepted: 11/05/2024] [Indexed: 02/28/2025]
Abstract
Tumor metastasis regulated by multiple complicated pathways is closely related to variations in the tumor microenvironment. Exosomes can regulate the tumor microenvironment through various mechanisms. Exosomes derived from tumor cells carry a variety of substances, including long non-coding RNAs (lncRNAs), play important roles in intercellular communication and act as critical determinants influencing tumor metastasis. In this review, we elaborate on several pivotal processes through which lncRNAs regulate tumor metastasis, including the regulation of epithelial‒mesenchymal transition, promotion of angiogenesis and lymphangiogenesis, enhancement of the stemness of tumor cells, and evasion of immune clearance. Additionally, we comprehensively summarized a diverse array of potential tumor-derived exosomal lncRNA biomarkers to facilitate accurate diagnosis and prognosis in a clinical setting.
Collapse
Affiliation(s)
- Zhile Yu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Jiali Fu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Vanya Mantareva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Bld. 9, 1113, Sofia, Bulgaria
| | - Ivica Blažević
- Department of Organic Chemistry, Faculty of Chemistry and Technology, University of Split, Ruđera Boškovića 35, 21000, Split, Croatia
| | - Yusong Wu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Dianchang Wen
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Tungalag Battulga
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia.
| | - Yuqing Wang
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China.
- The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 510140, PR China.
| | - Jianye Zhang
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China.
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, 511518, PR China.
| |
Collapse
|
75
|
Wu X, Wu X, Wang Z, Tian X, Zhang C, Cao G, Gu Y, Yan T. Delivery of exogenous miR-19b by Wharton's Jelly Mesenchymal Stem Cells attenuates transplanted kidney ischemia/reperfusion injury by regulating cellular metabolism. Drug Deliv Transl Res 2025; 15:925-938. [PMID: 38918324 DOI: 10.1007/s13346-024-01645-3] [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] [Accepted: 06/03/2024] [Indexed: 06/27/2024]
Abstract
Renal ischemia-reperfusion injury (IRI) frequently occurs following kidney transplantation, and exosomes derived from umbilical cord mesenchymal stem cells (WJ-MSC-Exos) have shown promise in treating IRI in transplanted kidneys. Our study delved into the potential mechanism of WJ-MSC-Exos in ameliorating IRI in transplanted kidneys, revealing that miR-19b is abundantly present in WJ-MSC-Exos. Both in vivo and in vitro experiments demonstrated that the absence of miR-19b abolished the protective effects of WJ-MSC-Exos against renal IRI. Mechanistically, miR-19b suppressed glycogen synthase kinase-3β (GSK3β) expression, thereby stabilizing PDXK protein through direct binding. Treatment with WJ-MSC-Exos led to reduced PDXK levels and enhanced pyridoxine accumulation, ultimately mitigating IRI in transplanted kidneys and I/R-induced HK2 cell apoptosis. These findings elucidate the underlying mechanism of WJ-MSC-Exos in alleviating IRI in transplanted kidneys, unveiling novel therapeutic targets for post-kidney transplantation IRI and providing a solid theoretical foundation for the clinical application of WJ-MSC-Exos in IRI treatment post-transplantation.
Collapse
Affiliation(s)
- Xiaoqiang Wu
- Department of Urology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, China
| | - Xuan Wu
- Department of Urology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, China
| | - Zhiwei Wang
- Department of Urology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, China
| | - Xiangyong Tian
- Department of Urology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, China
| | - Chan Zhang
- Department of Urology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, China
| | - Guanghui Cao
- Department of Urology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, China
| | - Yue Gu
- Department of Nephrology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, China
| | - Tianzhong Yan
- Department of Urology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, China.
| |
Collapse
|
76
|
Aripova A, Kussainova A, Ibragimova M, Bulgakova O, Bersimbaev R. The role of exosomal hsa-miR-125b-5p and hsa-miR-320c as non-invasive biomarkers in high-radon areas of Kazakhstan. Biomarkers 2025; 30:123-130. [PMID: 39819391 DOI: 10.1080/1354750x.2025.2456007] [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: 09/18/2024] [Accepted: 01/12/2025] [Indexed: 01/19/2025]
Abstract
BACKGROUND Radon, a radioactive gas, is a significant risk factor for lung cancer, especially in non-smokers. This study examines the expression of exosomal microRNAs (miRNAs) as potential biomarkers for radon-induced effects. METHODS A total of 109 participants from high- and low-radon areas in Kazakhstan were included. Exosomal hsa-miR-125b-5p and hsa-miR-320c levels were quantified using real-time PCR. RESULTS Results revealed a 25.4-fold increase in hsa-miR-125b-5p and a 12.5-fold decrease in hsa-miR-320c in participants exposed to high-radon levels compared to controls. Bioinformatic analysis identified key target genes, such as PRDM1 and IRF4, which are implicated in cancer development. CONCLUSION These findings suggest that exosomal miRNAs could serve as non-invasive biomarkers for radon exposure, offering potential for early diagnosis and monitoring of radon-induced lung cancer. The study underscores the need for further research to validate these miRNAs as reliable diagnostic tools.
Collapse
Affiliation(s)
- Akmaral Aripova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana, Kazakhstan
| | - Assiya Kussainova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana, Kazakhstan
| | - Milana Ibragimova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana, Kazakhstan
| | - Olga Bulgakova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana, Kazakhstan
| | - Rakhmetkazhi Bersimbaev
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana, Kazakhstan
| |
Collapse
|
77
|
Choi YH, Kim HY, Park JO, Choi E. Enhanced Anti-Tumor Effects of Natural Killer Cell-Derived Exosomes Through Doxorubicin Delivery to Hepatocellular Carcinoma Cells: Cytotoxicity and Apoptosis Study. Int J Mol Sci 2025; 26:2234. [PMID: 40076856 PMCID: PMC11900065 DOI: 10.3390/ijms26052234] [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: 01/17/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Exosomes are nanosized extracellular vesicles secreted by various cells, including natural killer (NK) cells, and are known for their low toxicity, high permeability, biocompatibility, and strong targeting ability. NK cell-derived exosomes (NK-exos) contain cytotoxic proteins that enhance tumor-targeting efficiency, making them suitable for treating solid tumors such as hepatocellular carcinoma (HCC). Despite their potential in drug delivery, the mechanisms of drug-loaded NK-exos, particularly those loaded with doxorubicin (NK-exos-Dox), remain unclear in HCC. This study explored the anti-tumor effects of NK-exos-Dox against Hep3B cells in vitro. NK-exos-Dox expressed exosome markers (CD9 and CD63) and cytotoxic proteins (granzyme B and perforin) and measured 170-220 nm in size. Compared to NK-exos, NK-exos-Dox enhanced cytotoxicity and apoptosis in Hep3B cells by upregulating pro-apoptotic proteins (Bax, cytochrome c, cleaved caspase 3, and cleaved PARP) and inhibiting the anti-apoptotic protein (Bcl-2). These findings suggest that NK-exos-Dox significantly boost anti-tumor effects by activating specific cytotoxic molecules, offering promising therapeutic opportunities for solid tumor treatment, including HCC.
Collapse
Affiliation(s)
- You Hee Choi
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea; (H.Y.K.); (J.-O.P.)
| | - Ho Yong Kim
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea; (H.Y.K.); (J.-O.P.)
| | - Jong-Oh Park
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea; (H.Y.K.); (J.-O.P.)
| | - Eunpyo Choi
- Department of Mechanical Engineering, Sogang University, 35, Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| |
Collapse
|
78
|
Corsi J, Semnani P, Peroni D, Belli R, Morelli A, Lassandro M, Sidarovich V, Adami V, Valentini C, Cavallerio P, Grosskreutz J, Fabbiano F, Grossmann D, Hermann A, Tell G, Basso M, D’Agostino V. Small molecule inhibitors of hnRNPA2B1-RNA interactions reveal a predictable sorting of RNA subsets into extracellular vesicles. Nucleic Acids Res 2025; 53:gkaf176. [PMID: 40103230 PMCID: PMC11915509 DOI: 10.1093/nar/gkaf176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/20/2025] Open
Abstract
Extracellular vesicles (EVs) are cell-secreted membranous particles contributing to intercellular communication. Coding and noncoding RNAs can be detected as EV cargo, and RNA-binding proteins (RBPs), such as hnRNPA2B1, have been circumstantially implicated in EV-RNA sorting mechanisms. However, the contribution of competitive RBP-RNA interactions responsible for RNA-sorting outcomes is still unclear, especially for predicting the EV-RNA content. We designed a reverse proteomic analysis exploiting the EV-RNA to identify intracellular protein binders in vitro. Using cells expressing a recombinant hnRNPA2B1 to normalize competitive interactions, we prioritized a network of heterogeneous nuclear ribonucleoproteins and purine-rich RNA sequences subsequently validated in secreted EV-RNA through short fluorescent RNA oligos. Then, we designed a GGGAG-enriched RNA probe that efficiently interacted with a full-length human hnRNPA2B1 protein. We exploited the interaction to conduct a pharmacological screening and identify inhibitors of the protein-RNA binding. Small molecules were orthogonally validated through biochemical and cell-based approaches. Selected drugs remarkably impacted secreted EV-RNAs and reduced an RNA-dependent, EV-mediated paracrine activation of NF-kB in recipient cells. These results demonstrate the relevance of post-transcriptional mechanisms for EV-RNA sorting and the possibility of predicting the EV-RNA quality for developing innovative strategies targeting discrete paracrine functions.
Collapse
Affiliation(s)
- Jessica Corsi
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Pouriya Sharbatian Semnani
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Daniele Peroni
- MS Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Romina Belli
- MS Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Alessia Morelli
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Michelangelo Lassandro
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Viktoryia Sidarovich
- HTS Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Valentina Adami
- HTS Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Chiara Valentini
- NGS Core Facility, Department of Cellular, Computational and Integrative Biomedicine CIBIO, University of Trento, 38122 Trento, Italy
| | - Paolo Cavallerio
- NGS Core Facility, Department of Cellular, Computational and Integrative Biomedicine CIBIO, University of Trento, 38122 Trento, Italy
| | - Julian Grosskreutz
- Excellence Cluster Precision Medicine in Inflammation, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Fabrizio Fabbiano
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Dajana Grossmann
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, 18147 Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, 18147 Rostock, Germany
| | - Gianluca Tell
- Laboratory of Molecular Biology and DNA repair, Department of Medicine (DMED), University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy
| | - Manuela Basso
- Laboratory of Transcriptional Neurobiology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Vito G D’Agostino
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento, Italy
| |
Collapse
|
79
|
Yang J, Gui Y, Zheng Y, He H, Chen L, Li T, Liu H, Wang D, Yuan D, Yuan C. Total saponins from Panax japonicus reduced lipid deposition and inflammation in hepatocyte via PHD2 and hepatic macrophage-derived exosomal miR-463-5p. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119376. [PMID: 39842748 DOI: 10.1016/j.jep.2025.119376] [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: 12/21/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax japonicus (T. Nees) C.A. Mey. (PJ) is a traditional Chinese herbal medicine revered as the "King of Herbs" in Tujia and Hmong medical practices. Clinically, it is primarily used to treat weakness and fatigue, wound bleeding, arthritis, hyperlipidemia, and fatty liver. It is rich in saponins, and the total saponins from PJ (TSPJ), possess immunomodulatory, antioxidant, and lipid-lowering effects. These properties hold significant potential in managing liver-related metabolic diseases such as non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). AIM OF STUDY Evaluate the therapeutic effects of TSPJ on lipid metabolism disorders in a NASH model and explore the possible underlying mechanisms. MATERIALS AND METHODS To model NASH, C57BL/6J mice were fed a high-fat diet (HFD) and RAW264.7 cells were stimulated with lipopolysaccharide (LPS) and palmitic acid (PA). The animal and cell models were also treated with TSPJ, and the changes in inflammation and lipid metabolism were measured. Additional models were created by transfecting lentiviral vectors to cause miR-463-5p knockdown in the C57BL/6J mouse and the RAW264.7 cells. RESULTS In the HFD-induced mice, TSPJ reduced the body weight and liver weight, lowered the serum levels of TG, T-CHO, ALT, and AST, and reduced the hepatic lipid droplet formation and vacuolization. In the RAW264.7 cells, TSPJ upregulated the M2 markers and downregulated the M1 markers. TSPJ also significantly increased the expression of miR-463-5p in the exosomes derived from the RAW264.7 cells or the primary mouse hepatic macrophages, and miR-463-5p suppressed the expression of PHD2 in hepatocytes to improve lipid metabolism. However, when the exosome secretion inhibitor GW4869 was applied, TSPJ became less effective in alleviating the lipid deposition and inflammation in hepatocytes. CONCLUSIONS TSPJ significantly upregulated the expression of miR-463-5p in the exosomes of hepatic macrophages to thus downregulate PHD2 expression in hepatocytes and improve hepatic lipid metabolism.
Collapse
Affiliation(s)
- Jingjie Yang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| | - Yibei Gui
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| | - Ying Zheng
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| | - Haodong He
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| | - Lihan Chen
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| | - Tongtong Li
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| | - Haoran Liu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| | - Dongshuo Wang
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| | - Ding Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China; College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China.
| | - Chengfu Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China.
| |
Collapse
|
80
|
Cui J, Lin S, Zhang M. Resveratrol loaded microglia-derived exosomes attenuate astrogliasis by restoring mitochondrial function to reduce spinal cord injury. Chem Biol Interact 2025; 408:111407. [PMID: 39884494 DOI: 10.1016/j.cbi.2025.111407] [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: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
The activation of astrocytes in the injured lesion induces the progression of spinal cord injury (SCI). However, adverse side-effects during systemic administration have limited applications. Exosomes (Exos) are an emerging clinical treatment method that exerts anti-inflammatory effects by reducing pro-inflammatory factors and promoting functional recovery. Exosomes exhibit great potential as carriers of traditional Chinese medicine, attributed to their high delivery efficiency to internalized and targeted accumulation in inflammatory tissues. Herein, We synthesized resveratrol loaded microglia-derived exosomes (R-MDEs) for highly efficient accumulation and infiltration in the injured spinal cord. In vitro and in vivo experiments suggested that R-MDEs effectively accumulated in A1 astrocytes, inhibited reactive oxygen species (ROS) and glial scar formation by reprogramming the metabolic astrocytes. R-MDEs achieve a synergistic therapeutic effect of immunomodulation and neuroprotection, thereby shedding new light on the application of Exos and provides great potential for SCI.
Collapse
Affiliation(s)
- Jingwen Cui
- Department of Neurosurgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China; Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, PR China; Key Laboratory of Medical Tissue Engineering of Liaoning Province, Jinzhou Medical University, Jinzhou, PR China
| | - Sen Lin
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, PR China; Key Laboratory of Medical Tissue Engineering of Liaoning Province, Jinzhou Medical University, Jinzhou, PR China.
| | - Mingyuan Zhang
- Department of Neurosurgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China.
| |
Collapse
|
81
|
Yang HB, Lu DC, Shu M, Li J, Ma Z. The roles and therapeutic potential of exosomal non-coding RNAs in microglia-mediated intercellular communication. Int Immunopharmacol 2025; 148:114049. [PMID: 39823800 DOI: 10.1016/j.intimp.2025.114049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/20/2025]
Abstract
Exosomes, which are small extracellular vesicles (sEVs), serve as versatile regulators of intercellular communication in the progression of various diseases, including neurological disorders. Among the diverse array of cargo they carry, non-coding RNAs (ncRNAs) play key regulatory roles in various pathophysiological processes. Exosomal ncRNAs derived from distinct cells modulate their reciprocal crosstalk locally or remotely, thereby mediating neurological diseases. Nevertheless, the emerging role of exosomal ncRNAsin microglia-mediated phenotypes remains largely unexplored. This review aims to summarise the biological functions of exosomal ncRNAs and the molecular mechanisms that underlie their impact on microglia-mediated intercellular communication, modulating neuroinflammation and synaptic functions within the landscape of neurological disorders. Furthermore, this review comprehensively described the potential applications of exosomal ncRNAs as diagnostic and prognostic biomarkers, as well as innovative therapeutic targets for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Hu-Bo Yang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Ding-Ci Lu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Min Shu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Juan Li
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China.
| |
Collapse
|
82
|
Zhao Y, Yu H, Li J, Qian J, Li M, Zhang X, Wang M, Wang Y, Dong Y, You Y, Zhou Q, Gao D, Zhao Y, Liu B, Chen R, Ren Z, Wang Z, Zhang K, Cui J. A glucose-enriched lung pre-metastatic niche triggered by matrix stiffness-tuned exosomal miRNAs in hepatocellular carcinoma. Nat Commun 2025; 16:1736. [PMID: 39966385 PMCID: PMC11836368 DOI: 10.1038/s41467-025-56878-8] [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: 03/05/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
Apart from the classic features, it is almost unknown whether there exist other new pathological features during pre-metastatic niche formation in hepatocellular carcinoma (HCC). Our previous works have highlighted the contribution of increased matrix stiffness to lung pre-metastatic niche formation and metastasis in HCC. However, whether increased matrix stiffness influences glucose metabolism and supply of lung pre-metastatic niche remains largely unclear. Here we uncover the underlying mechanism by which matrix stiffness-tuned exosomal miRNAs as the major contributor modulate glucose enrichment during lung pre-metastatic niche formation through decreasing the glucose uptake and consumption of lung fibroblasts and increasing angiogenesis and vascular permeability. Our findings suggest that glucose enrichment, a new characteristic of the lung pre-metastatic niche triggered by matrix stiffness-tuned exosomal miRNAs, is essential for the colonization and survival of metastatic tumor cells, as well as subsequent metastatic foci growth.
Collapse
Affiliation(s)
- Yingying Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Hongmei Yu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Jiajun Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Jiali Qian
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Miao Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Xi Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Mimi Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Yaohui Wang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, PR China
| | - Yinying Dong
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, PR China
| | - Yang You
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Qiwen Zhou
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Yan Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Binbin Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Rongxin Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Zhenggang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China
| | - Zhiming Wang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | - Kezhi Zhang
- Department of Hepatobiliary Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu Province, PR China.
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, PR China.
| |
Collapse
|
83
|
Chen B, Qiu X. Surface-Enhanced Raman Scattering (SERS) for exosome detection. Clin Chim Acta 2025; 568:120148. [PMID: 39842651 DOI: 10.1016/j.cca.2025.120148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Exosomes, nanoscale extracellular vesicles secreted by various cells, are abundantly present in biological fluids. They have been identified as carriers of specific molecules, suggesting their potential role in early disease detection. However, their clinical application is hindered by several challenges, including the need for large sample volumes for enrichment, limitations of traditional detection methods, and the complexity involved in phenotype analysis and separation. OBJECTIVE This review aims to explore the application of Surface-Enhanced Raman Scattering (SERS) technology in exosome detection. SERS, known for its unique photonic properties and high sensitivity, offers a promising solution for detecting exosomes without the need for large sample volumes or extensive phenotypic analysis. This review focuses on the real-time and non-invasive assessment capabilities of SERS in exosome detection, providing insights into its potential for early disease diagnosis. CONCLUSION The review concludes by emphasizing the potential of SERS-based exosome detection in advancing early disease diagnosis. By overcoming existing challenges, SERS technology offers a promising approach for the development of sensitive and specific diagnostic assays, contributing to better patient outcomes and personalized medicine.
Collapse
Affiliation(s)
- Biqing Chen
- Gynaecology and Obstetrics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Heilongjiang 150081 PR China
| | - Xiaohong Qiu
- Gynaecology and Obstetrics, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Heilongjiang 150081 PR China.
| |
Collapse
|
84
|
Liu AP, Sun TJ, Liu TY, Duan HZ, Jiang XH, Li M, Luo YZ, Feloney MP, Cline M, Zhang YY, Yu AY. Urinary exosomes as promising biomarkers for early kidney disease detection. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2025; 13:1-19. [PMID: 40124571 PMCID: PMC11928825 DOI: 10.62347/dake5842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/21/2025] [Indexed: 03/25/2025]
Abstract
Kidney injury and disease pose a significant global health burden. Despite existing diagnostic methods, early detection remains challenging due to the lack of specific molecular markers to identify and stage various kidney lesions. Urinary exosomes, extracellular vesicles secreted by kidney cells, offer a promising solution. These vesicles contain a variety of biomolecules, such as proteins, RNA, and DNA. These biomolecules can reflect the unique physiological and pathological states of the kidney. This review explores the potential of urinary exosomes as biomarkers for a range of kidney diseases, including renal failure, diabetic nephropathy, and renal tumors. By analyzing specific protein alterations within these exosomes, we aim to develop more precise and tailored diagnostic tools to detect kidney diseases at an early stage and improve patient outcomes. While challenges persist in isolating, characterizing, and extracting reliable information from urinary exosomes, overcoming these hurdles is crucial for advancing their clinical application. The successful implementation of urinary exosome-based diagnostics could revolutionize early kidney disease detection, enabling more targeted treatment and improved patient outcomes.
Collapse
Affiliation(s)
- An-Ping Liu
- Dalian Medical UniversityDalian 116044, Liaoning, China
- Department of Emergency, Affiliated Hospital of Zunyi Medical UniversityZunyi 563003, Guizhou, China
| | - Tian-Jing Sun
- Department of Emergency, Affiliated Hospital of Zunyi Medical UniversityZunyi 563003, Guizhou, China
| | - Tong-Ying Liu
- Department of Emergency, Affiliated Hospital of Zunyi Medical UniversityZunyi 563003, Guizhou, China
| | - Hai-Zhen Duan
- Department of Emergency, Affiliated Hospital of Zunyi Medical UniversityZunyi 563003, Guizhou, China
| | - Xu-Heng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical UniversityZunyi 563003, Guizhou, China
| | - Mo Li
- Department of Emergency, Affiliated Hospital of Zunyi Medical UniversityZunyi 563003, Guizhou, China
| | - Yuan-Ze Luo
- Dejiang County Ethnic Traditional Chinese Medicine HospitalZunyi 563003, Guizhou, China
| | - Michael P Feloney
- Department of Urology, School of Medicine, Creighton University School of MedicineOmaha, NE, USA
| | - Mark Cline
- Department of Pathology, Wake Forest School of MedicineWinston-Salem, NC, USA
| | - Yuan-Yuan Zhang
- Wake Forest Institute of Regenerative Medicine, Wake Forest School of MedicineWinston-Salem, NC, USA
| | - An-Yong Yu
- Dalian Medical UniversityDalian 116044, Liaoning, China
- Department of Emergency, Affiliated Hospital of Zunyi Medical UniversityZunyi 563003, Guizhou, China
| |
Collapse
|
85
|
Li H, Zhang P, Lin M, Li K, Zhang C, He X, Gao K. Pyroptosis: candidate key targets for mesenchymal stem cell-derived exosomes for the treatment of bone-related diseases. Stem Cell Res Ther 2025; 16:68. [PMID: 39940049 PMCID: PMC11816542 DOI: 10.1186/s13287-025-04167-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
Bone-related diseases impact a large portion of the global population and, due to their high disability rates and limited treatment options, pose significant medical and economic challenges. Mesenchymal stem cells (MSCs) can differentiate into multiple cell types and offer strong regenerative potential, making them promising for treating various diseases. However, issues with the immune response and cell survival limit the effectiveness of cell transplantation. This has led to increased interest in cell-free stem cell therapy, particularly the use of exosomes, which is the most studied form of this approach. Exosomes are extracellular vesicles that contain proteins, lipids, and nucleic acids and play a key role in cell communication and material exchange. Pyroptosis, a form of cell death involved in innate immunity, is also associated with many diseases. Studies have shown that MSC-derived exosomes have therapeutic potential for treating a range of conditions by regulating inflammation and pyroptosis. This study explored the role of MSC-derived exosomes in modulating pyroptosis to improve the treatment of bone-related diseases.
Collapse
Affiliation(s)
- Haiming Li
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China
| | - Peng Zhang
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China
| | - Minghui Lin
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China
| | - Kang Li
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China
| | - Cunxin Zhang
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| | - Xiao He
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| | - Kai Gao
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China.
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| |
Collapse
|
86
|
Farzam OR, Eslami S, Jafarizadeh A, Alamdari SG, Dabbaghipour R, Nobari SA, Baradaran B. The significance of exosomal non-coding RNAs (ncRNAs) in the metastasis of colorectal cancer and development of therapy resistance. Gene 2025; 937:149141. [PMID: 39643147 DOI: 10.1016/j.gene.2024.149141] [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: 09/20/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Colorectal cancer (CRC) represents a common type of carcinoma with significant mortality rates globally. A primary factor contributing to the unfavorable treatment outcomes and reduced survival rates in CRC patients is the occurrence of metastasis. Various intricate molecular mechanisms are implicated in the metastatic process, leading to mortality among individuals with CRC. In the realm of intercellular communication, exosomes, which are a form of extracellular vesicle (EV), play an essential role. These vesicles act as conduits for information exchange between cells and originate from multiple sources. By fostering a microenvironment conducive to CRC progression, exosomes and EVs significantly influence the advancement of the disease. They contain a diverse array of molecules, including messenger RNAs (mRNAs), non-coding RNAs (ncRNAs), proteins, lipids, and transcription factors. Notably, ncRNAs, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), are prominently featured within exosomes. These ncRNAs have the capacity to regulate various critical molecules or signaling pathways, particularly those associated with tumor metastasis, thereby playing a crucial role in tumorigenesis. Their presence indicates a substantial potential to affect vital aspects of tumor progression, including proliferation, metastasis, and resistance to treatment. This research aims to categorize exosomal ncRNAs and examine their functions in colorectal cancer. Furthermore, it investigates the clinical applicability of novel biomarkers and therapeutic strategies in CRC. Abbreviations: ncRNAs, non-coding RNAs; CRC, Colorectal cancer; EV, extracellular vesicle; mRNAs, messenger RNAs; miRNAs, microRNAs; lncRNAs, long non-coding RNAs; circRNAs, circular RNAs; HOTTIP, HOXA transcript at the distal tip; NSCLC, non-small cell lung cancer; 5-FU, 5-fluorouracil; OX, Oxaliplatin; PDCD4, programmed cell death factor 4; Tregs, regulatory T cells; EMT, epithelial-mesenchymal transition; PFKFB3, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3; USP2, ubiquitin carboxyl-terminal hydrolase 2; TNM, tumor node metastasis; TAMs, tumor-associated macrophages; RASA1, RAS p21 protein activator 1; PDCD4, programmed cell death 4; ZBTB2, zinc finger and BTB domain containing 2; SOCS1, suppressor of cytokine signaling 1; TUBB3, β-III tubulin; MSCs, mesenchymal stem cells.
Collapse
Affiliation(s)
- Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahand Eslami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Jafarizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Evidence-based Medicine, Iranian EBM Center: A Joana-affiliated Group, Tabriz University of Medicine Science, Tabriz, Iran
| | - Sania Ghobadi Alamdari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Cell and Molecular Biology, Faculty of Basic Sciences, University of Maragheh, Maragheh, Iran
| | - Reza Dabbaghipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Alizadeh Nobari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
87
|
Wang Z, Huang Y, He S, Zhou Y, Zhao L, Wang F. Dynamic and functional analyses of exosomal miRNAs regulating cellular microenvironment of ovarian cancer cells. J Ovarian Res 2025; 18:25. [PMID: 39930447 PMCID: PMC11808964 DOI: 10.1186/s13048-025-01608-3] [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: 06/03/2024] [Accepted: 01/23/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Exosomes, extracellular vesicles with an average diameter of 30 ~ 150 nm, are pivotal in mediating the cellular microenvironment (CM) through their cargo-carrying capability. Despite extensive studies, the dynamic and regulatory mechanisms of exosomal cargoes, including lipids, proteins, nucleic acids, and metabolites, remain poorly understood. METHODS In this study, we collected culture medium of ovarian cancer cells at four different time points (12, 24, 36, 48 h). Exosomes were isolated using ultracentrifugation, and miRNA sequencing was performed for exosomes from each group (T12, T24, T36, and T48). RESULTS A total of 131 miRNAs were identified in all groups. Specifically, 41, 115, 63, and 24 miRNAs were detected in the T12, T24, T36, and T48 groups, respectively. Among these, 15 miRNAs were common to the all groups, while 3, 57, 10, and 3 miRNAs were unique to the T12, T24, T36, and T48 groups, respectively. Functional analyses of the target genes for both common and specific miRNAs indicated that numerous target genes were involved in signaling pathways and cancer-related processes. CONCLUSION It suggested that exosomal miRNAs might be critical in intercellular communication and in dynamically remodeling the tumor microenvironment. These insights could enhance our understanding of the role of exosomal miRNAs in cancer biology and inform the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Zhaoxia Wang
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China.
- First Hospital of Shanxi Medical University, 85 South Jiefang Road, Taiyuan, Shanxi, 030001, P.R. China.
| | - Yanan Huang
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Simin He
- Department of Health Statistics and Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Ying Zhou
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Le Zhao
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Fuyuan Wang
- Department of Gynecology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| |
Collapse
|
88
|
Xu Y, Li J, Qiu Y, Wu F, Xue Z, Liu B, Fan H, Zhou Y, Wu Q. USC-Derived Small Extracellular Vesicles-Functionalized Scaffolds Promote Scarless Vaginal Defect Repair via Delivery of Decorin and DUSP3 Proteins. Int J Nanomedicine 2025; 20:1615-1634. [PMID: 39931530 PMCID: PMC11808217 DOI: 10.2147/ijn.s499856] [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: 10/18/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
Background Scar formation following large-area vaginal defects post-vaginoplasty is a major clinical challenge. Compared to skin scars, vaginal scars can lead to pain during intercourse and urinary difficulties, severely impacting quality of life. Small extracellular vesicles (sEVs) encapsulate diverse bioactive components, making them potential therapeutic agents. Designing functional scaffolds that incorporate sEVs is a promising approach for scarless vaginal defect repair. Methods sEVs-loaded scaffolds were developed through electrostatic interactions between negatively charged sEVs secreted by urine-derived stem cells (USC-sEVs) and positively charged human acellular amniotic membranes. The efficacy of sEVs-loaded scaffolds in the treatment of vaginal defects in rabbits was assessed by histological analysis. Immunofluorescence staining, Western blot, qRT-PCR and collagen gel contraction analyses were conducted to evaluate the antifibrotic effects of USC-sEVs. RNA sequencing was employed to elucidate the underlying mechanisms involved. LC‒MS/MS analysis was used to identify candidate upstream proteins in USC-sEVs. Results In vivo experiments demonstrated that the sEVs-loaded scaffolds promoted scarless healing of vaginal defects in rabbits by modulating collagen deposition, reducing fibrosis, and diminishing inflammation. In vitro experiments revealed that USC-sEVs significantly inhibited the proliferation, collagen production, and activation of fibroblasts with a fibrotic phenotype, indicating the antifibrotic properties of USC-sEVs. Transcriptome and Western blot analyses revealed that USC-sEVs treatment inhibited fibrosis by downregulating the TGF-β and p38 MAPK signaling pathways. LC‒MS/MS analysis identified 2653 proteins encapsulated in USC-sEVs. Western blot analysis revealed that decorin, an inhibitor of the TGF-β signaling pathway, and DUSP3, a negative regulator of p38 phosphorylation, were enriched in USC-sEVs and could be transferred to fibroblasts. Conclusion USC-sEVs inhibited fibrosis and promoted scarless healing by delivering decorin and DUSP3 proteins, which regulate the TGF-β and p38 MAPK signaling pathways, respectively. This study highlights the potential of sEVs-loaded scaffolds as a promising strategy for scarless vaginal repair following vaginoplasty, offering a novel approach for regenerative medicine with significant translational potential for clinical application.
Collapse
Affiliation(s)
- Yiyun Xu
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Jie Li
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Yu Qiu
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Fuyue Wu
- Organoid Regeneration Research Center, ReMed Regenerative Medicine Clinical Application Institute, Shanghai, 201114, People’s Republic of China
| | - Zhuowei Xue
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Bin Liu
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Hongjie Fan
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Yuedi Zhou
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Qingkai Wu
- Department of Obstetrics and Gynecology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| |
Collapse
|
89
|
Zhao X, Yao M, Wang Y, Feng C, Yang Y, Tian L, Bao C, Li X, Zhu X, Zhang X. Neuroregulation during Bone Formation and Regeneration: Mechanisms and Strategies. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7223-7250. [PMID: 39869030 DOI: 10.1021/acsami.4c16786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The skeleton is highly innervated by numerous nerve fibers. These nerve fibers, in addition to transmitting information within the bone and mediating bone sensations, play a crucial role in regulating bone tissue formation and regeneration. Traditional bone tissue engineering (BTE) often fails to achieve satisfactory outcomes when dealing with large-scale bone defects, which is frequently related to the lack of effective reconstruction of the neurovascular network. In recent years, increasing research has revealed the critical role of nerves in bone metabolism. Nerve fibers regulate bone cells through neurotransmitters, neuropeptides, and peripheral glial cells. Furthermore, nerves also coordinate with the vascular and immune systems to jointly construct a microenvironment favorable for bone regeneration. As a signaling driver of bone formation, neuroregulation spans the entire process of bone physiological activities from the embryonic formation to postmaturity remodeling and repair. However, there is currently a lack of comprehensive summaries of these regulatory mechanisms. Therefore, this review sketches out the function of nerves during bone formation and regeneration. Then, we elaborate on the mechanisms of neurovascular coupling and neuromodulation of bone immunity. Finally, we discuss several novel strategies for neuro-bone tissue engineering (NBTE) based on neuroregulation of bone, focusing on the coordinated regeneration of nerve and bone tissue.
Collapse
Affiliation(s)
- Xiangrong Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meilin Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuyi Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cong Feng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuhan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Luoqiang Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
90
|
Hade MD, Greenwald J, Palacio PL, Nguyen KT, Shantaram D, Butsch BL, Kim Y, Noria S, Brethauer SA, Needleman BJ, Hsueh W, Wysocki VH, Reátegui E, Magaña SM. Novel multiparametric bulk and single extracellular vesicle pipeline for adipose cell-specific biomarker discovery in paired human biospecimens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.18.590172. [PMID: 38659953 PMCID: PMC11042368 DOI: 10.1101/2024.04.18.590172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Obesity remains a growing and global public health burden across a broad spectrum of metabolic, systemic, and neurodegenerative diseases. Previously considered merely a fat storage depot, adipose tissue is now recognized as an active endocrine organ crucial for metabolic and systemic regulation of local and distant organs. A burgeoning line of investigation centers on adipose-derived extracellular vesicles (ADEVs) and their pivotal role in obesity-associated pathobiology. However, robust methodologies are lacking for specifically isolating and characterizing human ADEVs. To bridge this gap, we have developed a robust multiparametric framework incorporating bulk and single EV characterization, proteomics, and mRNA phenotyping. EVs from matched human visceral adipose tissue, mature adipocyte-conditioned media, and plasma collected from the same individual bariatric surgical patients were analyzed and subjected to bottom-up proteomics analysis. This framework integrates bulk EV proteomics for cell-specific marker identification and subsequent single EV interrogation with single-particle interferometric reflectance imaging (SP-IRIS) and total internal reflection fluorescence (TIRF) microscopy. Our proteomics analysis revealed 76 unique proteins from adipose tissue-derived EVs (ATEVs), 512 unique proteins from adipocyte EVs (aEVs), and 1003 shared proteins. Prominent pathways enriched in ATEVs included lipid metabolism, extracellular matrix organization, and immune modulation, while aEVs exhibited enhanced roles in chromatin remodeling, oxidative stress responses, and metabolic regulation. Notably, adipose tissue-specific proteins such as adiponectin and perilipin were highly enriched in ADEVs and confirmed in circulating plasma EVs. Colocalization of key EV and adipocyte markers, including CD63 and PPARG, were validated in circulating plasma EVs. In summary, our study paves the way toward a tissue and cell-specific, multiparametric framework for an 'adiposity EV signature' in obesity-driven diseases.
Collapse
|
91
|
Zeng Z, Zhao Z, Yuan Q, Yang S, Wang Z, Wang Z, Zeng S, Li A, Chen Q, Zhu G, Xiao X, Luo G, Luo H, Li J, Zu X, Xie H, Liu J. Hepatic Steatosis Aggravates Vascular Calcification via Extracellular Vesicle-Mediated Osteochondrogenic Switch of Vascular Smooth Muscle Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408660. [PMID: 39680681 PMCID: PMC11791995 DOI: 10.1002/advs.202408660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/17/2024] [Indexed: 12/18/2024]
Abstract
The global incidence of metabolic dysfunction-associated fatty liver disease (MAFLD) has risen sharply. This condition is strongly associated with the risk of cardiovascular disease (CVD), but how MAFLD affects the development and progression of CVD, particularly concerning vascular calcification, remains unclear. Herein, extracellular vesicles (EVs) are identified from steatotic hepatocytes as a trigger that accelerated the progression of both vascular intimal and medial calcification. Steatotic hepatocytes are found to release more EVs, which are able to reach the vascular tissue, be taken up by vascular smooth muscle cells (VSMCs), and promote their osteogenic differentiation. Within these toxic vesicles, a protein cargo is identified called lectin galactoside-binding soluble 3 binding protein (Lgals3bp) that acted as a potent inducer of osteochondrogenic transformation in VSMCs. Both the inhibition of EV release and the liver-specific knockdown of Lgals3bp profoundly attenuated vascular calcification. This work partially explains the reason for the high incidence of vascular calcification in MAFLD and unveils a novel mechanism that may be used to prevent or treat cardiovascular complications in patients with MAFLD.
Collapse
Affiliation(s)
- Zhao‐Lin Zeng
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Institute of Cardiovascular DiseaseKey Lab for Arteriosclerology of Hunan ProvinceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Zhi‐Bo Zhao
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Qing Yuan
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Shi‐Qi Yang
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Zhen‐Xing Wang
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zuo Wang
- Institute of Cardiovascular DiseaseKey Lab for Arteriosclerology of Hunan ProvinceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Shi‐Yu Zeng
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - An‐Qi Li
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Qian Chen
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Guo‐Qiang Zhu
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Xin‐Hua Xiao
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Guang‐Hua Luo
- Department of RadiologyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Hai‐Yan Luo
- Department of GastroenterologyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Jiao‐Yang Li
- Department of Occupational and Environmental HealthSchool of Public HealthWuhan UniversityWuhan430071P. R. China
| | - Xu‐Yu Zu
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Hui Xie
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jiang‐Hua Liu
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| |
Collapse
|
92
|
Wu X, Yuan P, Wei N, Ma C, Fu M, Wu W. Extracellular vesicles derived from "serum and glucose" deprived HUCMSCs promoted skin wound healing through enhanced angiogenesis. Mol Cell Biochem 2025; 480:1255-1273. [PMID: 38967721 DOI: 10.1007/s11010-024-05058-1] [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: 12/28/2023] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
Extracellular vesicles (EVs) produced from MSCs were currently considered as a novel therapeutic agent for skin tissue regeneration and repair. Preconditioning stem cells may activate more molecular pathways and release more bioactive agents. In this study, we obtained EVs from normal (N-EVs) and serum- and glucose-deprived (SGD-EVs) human umbilical cord mesenchymal stem cells (HUCMSCs), and showed that SGD-EVs promoted the migration, proliferation, and tube formation of HUVECs in vitro. In vivo experiments utilizing a rat model show that both N-EVs and SGD-EVs boosted angiogenesis of skin defects and accelerated skin wound healing, while treating wounds with SGD-EVs led to faster skin healing and enhanced angiogenesis. miRNA sequencing showed that miR-29a-3p was abundant in SGD-EVs, and overexpressing miR-29a-3p enhanced the angiogenic ability of HUVECs, while inhibiting miR-29a-3p presented the opposite effect. Further studies demonstrated that miR-29a-3p directly targeted CTNNBIP1, which mediated angiogenesis of HUCMSCs-derived EVs through inhibiting CTNNBIP1 to activate Wnt/β-catenin signaling pathway. Taken together, these findings suggested that SGD-EVs promote angiogenesis via transferring miR-29a-3p, and activation of Wnt/β-catenin signaling pathway played a crucial role in SGD-EVs-induced VEGFA production during wound angiogenesis. Our results offered a new avenue for modifying EVs to enhance tissue angiogenesis and augment its role in skin repair.
Collapse
Affiliation(s)
- Xiaopeng Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
- College of Life Science, Northwest University, Xi'an, 710069, China
| | - Pingping Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Na Wei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
- College of Life Science, Northwest University, Xi'an, 710069, China
| | - Chaoqun Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Mingdi Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Wei Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, 145 Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
93
|
Chen M, Huang B, Su X. Mesenchymal stem cell-derived extracellular vesicles in periodontal bone repair. J Mol Med (Berl) 2025; 103:137-156. [PMID: 39821702 DOI: 10.1007/s00109-025-02513-4] [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: 08/08/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
Periodontitis is a chronic inflammatory disease that destroys tooth-supporting structures and poses significant public health challenges due to its high prevalence and links to systemic health conditions. Traditional treatments are effective in reducing the inflammatory response and improving the clinical symptoms of periodontitis. However, these methods are challenging to achieve an ideal treatment effect in alveolar bone repair. Mesenchymal stem cells (MSCs) represent a potential alternative for the treatment of periodontal bone defects due to their self-renewal and differentiation capabilities. Recent research indicates that MSCs exert their effects primarily through paracrine mechanisms. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) serve as pivotal mediators in intercellular communication, transferring microRNAs (miRNAs), messenger RNAs (mRNAs), proteins, and cytokines to recipient cells, thereby emulating the therapeutic effects of MSCs. In periodontitis, MSC-EVs play a pivotal role in immunomodulation and bone remodeling, thereby facilitating periodontal bone repair. As a cell-free therapy, MSC-EVs demonstrate considerable clinical potential due to their specialized membrane structure, minimal immunogenicity, low toxicity, high biocompatibility, and nanoscale size. This review indicates that MSC-EVs represent a promising approach for periodontitis treatment, with the potential to overcome the limitations of traditional therapies and offer a more effective solution for bone repair in periodontal disease. In this review, we introduce MSC-EVs, emphasizing their mechanisms and clinical applications in periodontal bone repair. It synthesizes recent advances, existing challenges, and future prospects to present up-to-date information and novel techniques for periodontal regeneration and to guide the improvement of MSC-EV therapy in clinical practice.
Collapse
Affiliation(s)
- Mengbing Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases &, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Bo Huang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases &, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaoxia Su
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases &, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
94
|
Rohm TV, Cunha E Rocha K, Olefsky JM. Metabolic Messengers: small extracellular vesicles. Nat Metab 2025; 7:253-262. [PMID: 39920357 DOI: 10.1038/s42255-024-01214-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 12/19/2024] [Indexed: 02/09/2025]
Abstract
Small extracellular vesicles (sEVs) are signalling molecules and biomarkers of cell status that govern a complex intraorgan and interorgan communication system through their cargo. Initially recognized as a waste disposal mechanism, they have emerged as important metabolic regulators. They transfer biological signals to recipient cells through their cargo content, and microRNAs (miRNAs) often mediate their metabolic effects. This review provides a concise overview of sEVs, specifically in the context of obesity-associated chronic inflammation and related metabolic disorders, describing their role as metabolic messengers, identifying their key sites of action and elucidating their mechanisms. We highlight studies that have shaped our understanding of sEV metabolism, address critical questions for future exploration, discuss the use of miRNAs as disease biomarkers and provide insights into the therapeutic potential of sEVs or specific miRNAs for treating metabolic diseases and related disorders in the future.
Collapse
Affiliation(s)
- Theresa V Rohm
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Karina Cunha E Rocha
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jerrold M Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
95
|
Sen MG, Chooi R, McMullen JR. Heart-derived factors and organ cross-talk in settings of health and disease: new knowledge and clinical opportunities for multimorbidity. J Physiol 2025. [PMID: 39888058 DOI: 10.1113/jp287400] [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: 09/26/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Cardiovascular disease affects millions of people worldwide and often presents with other conditions including metabolic, renal and neurological disorders. A variety of secreted factors from multiple organs/tissues (proteins, nucleic acids and lipids) have been implicated in facilitating organ cross-talk that may contribute to the development of multimorbidity. Secreted proteins have received the most attention, with the greatest body of research related to factors released from adipose tissue (adipokines), followed by skeletal muscle (myokines). To date, there have been fewer studies on proteins released from the heart (cardiokines) implicated with organ cross-talk. Early evidence for the secretion of cardiac-specific factors facilitating organ cross-talk came in the form of natriuretic peptides which are secreted via the classical endoplasmic reticulum-Golgi pathway. More recently, studies in cardiomyocyte-specific genetic mouse models have revealed cardiac-initiated organ cross-talk. Cardiomyocyte-specific modulation of microRNAs (miR-208a and miR-23-27-24 cluster) and proteins such as the mediator complex subunit 13 (MED13), G-protein-coupled receptor kinase 2 (GRK2), mutant α-myosin heavy-chain (αMHC), ubiquitin-like modifier-activating enzyme (ATG7), oestrogen receptor alpha (ERα) and fibroblast growth factor 21 (FGF21) have resulted in metabolic and renal phenotypes. These studies have implicated a variety of factors which can be secreted via the classical pathway or via non-classical mechanisms including the release of extracellular vesicles. Cross-talk between the heart and the brain has also been described (e.g. via miR-1 and an emerging concept, interoception: detection of internal neural signals). Here we summarize these studies taking into consideration that factors may be secreted in both settings of health and in disease.
Collapse
Affiliation(s)
- Melodi G Sen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Roger Chooi
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Heart Research Institute, Newtown, New South Wales, Australia
- Monash Alfred Baker Centre for Cardiovascular Research, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
96
|
Li H, Liu G, Wang B, Momeni MR. Exosomes and microRNAs as mediators of the exercise. Eur J Med Res 2025; 30:38. [PMID: 39828711 PMCID: PMC11742998 DOI: 10.1186/s40001-025-02273-4] [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] [Received: 04/03/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
MicroRNAs (miRNAs), also known as microribonucleic acids, are small molecules found in specific tissues that are essential for maintaining proper control of genes and cellular processes. Environmental factors, such as physical exercise, can modulate miRNA expression and induce targeted changes in gene transcription. This article presents an overview of the present knowledge on the principal miRNAs influenced by physical activity in different tissues and bodily fluids. Numerous research projects have emphasized the significant impact of miRNAs on controlling biological changes brought about by physical activity. These molecules play main roles in important processes such as the growth of skeletal muscle and heart muscle cells, the creation of mitochondria, the development of the vascular system, and the regulation of metabolism. Studies have shown that physical exertion utilizes the contributions of miR-1, miR-133, miR-206, miR-208, and miR-486 to revitalize and restore skeletal muscle tissue. Moreover, detecting alterations in miRNA levels and connecting them to the specific outcomes of various exercise routines and intensities can act as indicators for physical adaptation and the reaction to physical activity in long-term diseases. Numerous studies have confirmed that extracellular vesicles (EVs) which composed of different members such as exosomes have the ability to reduce inflammation through the activation of the nuclear factor kappa B (NF-κB pathway. Furthermore, physical activity greatly affects the levels of specific miRNAs present in exosomes derived from skeletal muscle. Therefore, exosomal miRNAs target some pathways that are related to growth and development, such asWnt/β-catenin, PI3K/AKT, and insulin-like growth factor 1 (IGF1). Exercise-induced exosomes have also been identified as important mediators in promoting beneficial effects throughout the body. The aim of this review is to summarize the effect of exercise on the function of miRNAs and exosomes.
Collapse
Affiliation(s)
- Haoyuan Li
- Department of Sport Leisure, Sungshin Women's University, Seoul, 02844, Korea
| | - Guifang Liu
- Department of Physical Education, Zhengzhou Normal University, Zhengzhou, 450044, Henan, China.
| | - Bing Wang
- School of Physical Education, Zhengzhou Normal University, Zhengzhou, 450044, Henan, China
| | | |
Collapse
|
97
|
Yuan S, Zhang P, Zhang F, Yan S, Dong R, Wu C, Deng J. Profiling signaling mediators for cell-cell interactions and communications with microfluidics-based single-cell analysis tools. iScience 2025; 28:111663. [PMID: 39868039 PMCID: PMC11763584 DOI: 10.1016/j.isci.2024.111663] [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] [Indexed: 01/28/2025] Open
Abstract
Cell-cell interactions and communication represent the fundamental cornerstone of cells' collaborative efforts in executing diverse biological processes. A profound understanding of how cells interface through various mediators is pivotal across a spectrum of biological systems. Recent strides in microfluidic technologies have significantly bolstered the precision and prowess in capturing and manipulating cells with exceptional spatial and temporal resolution. These advanced methodologies converge with multi-signal mediator detection systems, furnishing potent, high-throughput platforms for dissecting cell-cell interactions at the single-cell level. This approach empowers researchers to delve into intricate cellular dynamics with unprecedented accuracy and efficiency. Here, we present a critical evaluation of the latest advancements in microfluidics-driven techniques for detecting signal mediators involved in cell-cell interactions and communication at the single-cell level. We underscore notable biological applications that have benefited from these technologies and identify pressing challenges that must be addressed in future endeavors leveraging microfluidic tools for single-cell interaction studies.
Collapse
Affiliation(s)
- Shuai Yuan
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Peng Zhang
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Saudi Arabia
| | - Feng Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Shiqiang Yan
- Center of Cancer Immunology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ruihua Dong
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Chengjun Wu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Jiu Deng
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| |
Collapse
|
98
|
Chen G, Pei Y, Jiang P, Ye Q, Xie Z, Gyawali L. Exosomal NEDD4L derived from HG+oxLDL-induced vascular endothelial cells accelerates macrophage M1 polarization and oxLDL uptake by ubiquitinating IκBα and PPARγ. Cell Biol Toxicol 2025; 41:23. [PMID: 39775116 PMCID: PMC11706887 DOI: 10.1007/s10565-024-09973-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Vascular endothelial cell-derived exosomes are thought to mediate disease progression by regulating macrophage polarization. However, its mechanism in diabetes mellitus (DM)-related atherosclerosis (AS) progress is unclear. METHODS High-glucose (HG) and oxLDL were used to induce human cardiac microvascular endothelial cells (HCMECs) to mimic DM-related AS model. The conditioned medium (CM) from HG+oxLDL-induced HCMECs was incubated with THP1-M0 monocytes treated with LPS or oxLDL. The mRNA levels of macrophage M1/M2 polarization markers, NEDD4L, IκBα and PPARγ were determined by qRT-PCR. Flow cytometry was used to analyze macrophage marker. Dil-labeled oxLDL and oil red O staining were performed to assess oxLDL uptake by THP1-M0 cells. The levels of inflammatory factors were examined using ELISA. Transmission electron microscope was used for observing foam cell formation and exosome morphology. The protein levels of p-Smad1/Smad1, p-Smad2/Smad2, p-IκBα/IκBα, p-P65/P65, anti-lipid metabolism-related markers, and NEDD4L were tested by western blot. The interaction between NEDD4L and IκBα or PPARγ was assessed by Co-IP assay. RESULTS The CM of HG+oxLDL-induced HCMECs could promote macrophage M1 polarization, oxLDL uptake and foam cell formation, and exosome inhibiter GW4869 eliminated these effects. NEDD4L was overexpressed in exosomes from HG+oxLDL-induced HCMECs, which could be taken up by THP1-M0 cells. Exosomal NEDD4L knockdown inhibited macrophage M1 polarization, oxLDL uptake and foam cell formation by reducing the protein levels of p-Smad1/Smad1, p-Smad2/Smad2, p-IκBα/IκBα and p-P65/P65. NEDD4L could reduce IκBα and PPARγ expression through ubiquitination. CONCLUSION HG+oxLDL-induced HCMECs-derived exosomal NEDD4L could enhance the ubiquitination of IκBα and PPARγ to facilitate macrophage M1 polarization and oxLDL uptake, thus accelerating DM-related AS.
Collapse
Affiliation(s)
- Guozhu Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400030, China.
- Department of Cardiology, The Beibei Affiliated Hospital of Chongqing Medical University, No. 69, Jialing Village, Beibei District, Chongqing, 400030, China.
| | - Yisong Pei
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400030, China
| | - Peng Jiang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400030, China
| | - Qiaoling Ye
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400030, China
| | - Zulong Xie
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400030, China
| | | |
Collapse
|
99
|
Yang X, Liu J, Yin Y, Yang L, Gao M, Wu Z, Lu B, Luo S, Wang W, Li R. MSC-EXs inhibits uranium nephrotoxicity by competitively binding key proteins and inhibiting ROS production. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117654. [PMID: 39793287 DOI: 10.1016/j.ecoenv.2024.117654] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
Uranium poisoning, particularly from exposure to Depleted Uranium (DU), occurs when uranyl ions enter the bloodstream and bind primarily to transferrin, osteopontin, and albumin before entering cells via corresponding receptors on renal tubular membranes, leading to cellular damage. Uranium poisoning remains a significant clinical challenge, with no ideal treatment currently available. In this study, we investigate the therapeutic potential of human umbilical cord-derived mesenchymal stem cell exosomes (MSC-EXs) in mice exposed to DU. Our results showed that MSC-EXs could ameliorate renal damage and enhance kidney and bone marrow morphology but also effectively promote uranium excretion while reducing internal retention. Notably, the protective effects of MSC-EXs exceed those of MSCs and are comparable to those of sodium bicarbonate, as confirmed by various analytical techniques. Proteomic studies have shown that MSC-EXs reduce uranyl ion deposition in renal tubule cells through competitive binding with transferrin, osteopontin, and albumin. They also enhance oxidative stress resistance via modulation of glutathione metabolism, Cysteine and Methionine metabolism signaling pathways. This regulation leads to a reduction in mitochondrial ROS production, alleviates lipid peroxidation, and consequently decreases cellular apoptosis and ferroptosis. This study identifies MSC-EXs as a novel therapeutic strategy against depleted uranium poisoning, presenting potential advancements in treatment methodologies.
Collapse
Affiliation(s)
- Xinrui Yang
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Jing Liu
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Yaru Yin
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Luxun Yang
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Mingquan Gao
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Zifei Wu
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Binghui Lu
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Shenglin Luo
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Weidong Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu 610041, China.
| | - Rong Li
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
100
|
Guo Q, Liao H, Hao S, Hou D, Liu R, Zhang Y, Tang X, Song R, Tan X, Luo Z, Huang H, Duan C. TSPO exacerbates sepsis-induced cardiac dysfunction by inhibiting p62-Mediated autophagic flux via the ROS-RIP1/RIP3-exosome axis. Free Radic Biol Med 2025; 226:56-69. [PMID: 39542185 DOI: 10.1016/j.freeradbiomed.2024.11.018] [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: 09/14/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Septic cardiomyopathy (SCM) is a critical complication of sepsis, primarily attributed to mitochondrial dysfunction and impaired autophagic flux. This study explores the role of translocator protein (TSPO) in SCM pathogenesis and assesses its potential as a therapeutic target. We identified increased TSPO expression in plasma samples from sepsis patients, with further validation in septic rats and LPS-stimulated H9C2 cardiomyocytes. Elevated TSPO disrupted mitochondrial function, leading to increased reactive oxygen species (ROS) production and activation of the RIP1/RIP3 pathway, which hindered p62-positive autophagosome degradation and promoted inflammation. Moreover, exosome release containing TSPO-positive autophagosomes into plasma may exacerbate systemic inflammation. NADH, identified as a TSPO-binding molecule, restored autophagic flux, improved mitochondrial function, and enhanced cardiac performance and survival in septic rats. These findings suggest that targeting TSPO with NADH could alleviate mitochondrial dysfunction and inflammatory responses in SCM, providing a promising therapeutic strategy for sepsis-induced cardiac injury.
Collapse
Affiliation(s)
- Qiao Guo
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Haitang Liao
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Intensive Care Unit, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, PR China
| | - Shuai Hao
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China
| | - Dongyao Hou
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, PR China
| | - Ruixue Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yunting Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Xinhao Tang
- Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Rui Song
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Xuxin Tan
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Zhenchun Luo
- Intensive Care Unit, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, PR China.
| | - He Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| | - Chenyang Duan
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| |
Collapse
|