1
|
Tian M, Zhou Y, Guo Y, Xia Q, Wang Z, Zheng X, Shen J, Guo J, Duan S, Wang L. MicroRNAs in adipose tissue fibrosis: Mechanisms and therapeutic potential. Genes Dis 2025; 12:101287. [PMID: 40242037 PMCID: PMC12002615 DOI: 10.1016/j.gendis.2024.101287] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/07/2024] [Indexed: 04/18/2025] Open
Abstract
Adipose tissue fibrosis, characterized by abnormal extracellular matrix deposition within adipose tissue, signifies a crucial indicator of adipose tissue malfunction, potentially leading to organ tissue dysfunction. Various factors, including a high-fat diet, non-alcoholic fatty liver disease, and insulin resistance, coincide with adipose tissue fibrosis. MicroRNAs (miRNAs) represent a class of small non-coding RNAs with significant influence on tissue fibrosis through diverse signaling pathways. For instance, in response to a high-fat diet, miRNAs can modulate signaling pathways such as TGF-β/Smad, PI3K/AKT, and PPAR-γ to impact adipose tissue fibrosis. Furthermore, miRNAs play roles in inhibiting fibrosis in different contexts: suppressing corneal fibrosis via the TGF-β/Smad pathway, mitigating cardiac fibrosis through the VEGF signaling pathway, reducing wound fibrosis via regulation of the MAPK signaling pathway, and diminishing fibrosis post-fat transplantation via involvement in the PDGFR-β signaling pathway. Notably, the secretome released by miRNA-transfected adipose-derived stem cells facilitates targeted delivery of miRNAs to evade host immune rejection, enhancing their anti-fibrotic efficacy. Hence, this study endeavors to elucidate the role and mechanism of miRNAs in adipose tissue fibrosis and explore the mechanisms and advantages of the secretome released by miRNA-transfected adipose-derived stem cells in combating fibrotic diseases.
Collapse
Affiliation(s)
- Mei Tian
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Yang Zhou
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Yitong Guo
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Qing Xia
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Zehua Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Xinying Zheng
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Jinze Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Junping Guo
- Rainbowfish Rehabilitation and Nursing School, Hangzhou Vocational & Technical College, Hangzhou, Zhejiang 310018, China
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Lijun Wang
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
2
|
Phan N, Li Y, Yang M, Liu F. Tear Fluid Derived Extracellular Vesicles for New Biomarker Discovery. Ocul Surf 2025:S1542-0124(25)00062-X. [PMID: 40368029 DOI: 10.1016/j.jtos.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 04/23/2025] [Accepted: 05/05/2025] [Indexed: 05/16/2025]
Abstract
Various cell types release extracellular vesicles (EVs) containing proteins, DNA, and RNA essential for intercellular communication. The bioactive molecules from EVs can reflect disease status and monitor progression, while their communication abilities suggest therapeutic potential. We will review various EV isolation methods, EV-enriched fluids, and studies analyzing differential mi-RNA and protein levels extracted from EVs. Specifically, tear-derived EVs, which protect their molecular content and allow for real-time monitoring of ocular conditions such as Dry Eye Disease (DED), Sjögren's disease (SJD), Ocular graft-versus-host disease (oGVHD), and Diabetic Retinopathy (DR), which all currently remain undiagnosed in patients. EVs also provide potential as carriers for gene transfer, and mesenchymal stem cell (MSCs)-derived EVs are shown to be immunomodulatory, demonstrating promise for autoimmune ocular diseases. Through the multi-omic analysis of tear-fluid content, EVs are promising biomarkers and therapeutic agents in ocular diseases.
Collapse
Affiliation(s)
- Natalie Phan
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, 94720, USA
| | - Yi Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Menglu Yang
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.
| | - Fei Liu
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
3
|
Cao H, Li Z, Ye J, Lv Y, Zhang C, Liang T, Wang Y. Emerging roles of exosomes in the diagnosis and treatment of kidney diseases. Front Pharmacol 2025; 16:1525314. [PMID: 40308771 PMCID: PMC12041035 DOI: 10.3389/fphar.2025.1525314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
The complex etiology and spectrum of kidney diseases necessitate vigilant attention; the focus on early diagnosis and intervention in kidney diseases remains a critical issue in medical research. Recently, with the expanding studies on extracellular vesicles, exosomes have garnered increasing interest as a promising tool for the diagnosis and treatment of kidney diseases. Exosomes are nano-sized extracellular vesicles that transport a diverse array of bioactive substances, which can influence various pathological processes associated with kidney diseases and exhibit detrimental or beneficial effects. Within the kidney, exosomes derived from the glomeruli and renal tubules possess the ability to enter systemic circulation or urine. The biomarkers they carry can reflect alterations in the pathological state of the kidneys, thereby offering novel avenues for early diagnosis. Furthermore, research studies have confirmed that exosomes originating from multiple cell types exhibit therapeutic potential in treating kidney disease; notably, those derived from mesenchymal stem cells (MSCs) have shown significant treatment efficacy. This comprehensive review summarizes the contributions of exosomes from different cell types within the kidneys while exploring their physiological and pathological roles therein. Additionally, we emphasize recent advancements in exosome applications for the diagnosis and treatment of various forms of kidney diseases over the past decades. We not only introduce the urinary and blood biomarkers linked to kidney diseases found within exosomes but also explore their therapeutic effects. Finally, we discuss existing challenges and future directions concerning the clinical applications of exosomes for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Huanhuan Cao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zixi Li
- Department of Clinical Laboratory, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiajia Ye
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Lv
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Liang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yumei Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Liu Z, Liu X, Zhou Y, Wen X, Xu J, He M, Chen J, Jia N, Liu Y. Extracellular vesicles play a central role in linking podocyte injury to mesangial activation in glomerular disease. Theranostics 2025; 15:5121-5137. [PMID: 40303345 PMCID: PMC12036872 DOI: 10.7150/thno.110034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Podocyte injury leading to proteinuria is the primary feature of a vast majority of glomerular diseases, while mesangial cell activation is the hallmark of glomerulosclerosis. Whether and how these two events are connected remains elusive. In this study, we investigated the role of extracellular vesicles (EVs) in linking podocyte injury to mesangial activation in glomerular disease. Methods: EVs were characterized by nanoparticle tracking analysis and electron microscopy. Differentially expressed proteins from podocyte-derived EVs were analyzed by protein microarray. The role and mechanism by which EVs-packaged sonic hedgehog (Shh) mediates mesangial cell activation were investigated in vitro and in vivo. Results: An increased production of EVs in mouse podocytes (MPC5) was observed after injury induced by angiotensin II (Ang II). Shh and N-Shh were identified as major constituents of the proteins encapsulated in EVs isolated from Ang II-treated MPC5 cells (Ang II-EVs). In vitro, Ang II-EVs induced the activation and proliferation of rat mesangial cells (HBZY-1), whereas inhibition of EV secretion with dimethyl amiloride, depletion of EVs from conditioned media or knockdown of Shh expression abolished the ability of Ang II-EVs to induce HBZY-1 activation. In vivo, intravenous injection of Ang II-EVs exacerbated glomerulosclerosis, which was negated by hedgehog inhibitor. Furthermore, blocking EV secretion also ameliorated glomerulosclerosis in mouse model of glomerular disease. Conclusions: These findings suggest that podocyte injury can cause mesangial cell activation and glomerulosclerosis by releasing Shh-enriched EVs. Therefore, strategies targeting EVs may be a novel way to ameliorate proteinuric kidney disease.
Collapse
Affiliation(s)
- Zhao Liu
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xi Liu
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Zhou
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Wen
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Xu
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meizhi He
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiongcheng Chen
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Nan Jia
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
5
|
Niazi V, Ghafouri-Fard S. Effect of hypoxia on extracellular vesicles in malignant and non-malignant conditions. Cancer Treat Res Commun 2025; 43:100924. [PMID: 40209539 DOI: 10.1016/j.ctarc.2025.100924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025]
Abstract
Extracellular vesicles (EVs) are produced by virtually all types of cells and can be detected in nearly all extracellular places. These particles mediate intercellular communication and transfer their cargo to the recipient cells, inducing a variety of processes in these cells through transmission of several biomolecules such as miRNAs, lncRNAs, other transcripts and a variety of proteins. It has been documented that size, quantity, and expression of biomolecules in the EVs are influenced by the level of oxygen. In fact, hypoxia can affect several cellular processes through modulation of the cargo of these vesicles. Hypoxic exosomes derived from tumor cells have several protumoral effects on the recipient cells, including enhancement of proliferation, migration, and invasion in other tumoral cells, induction of metastasis in distant organs, stimulation of angiogenesis in the endothelial cells, and modulation of macrophage polarization. Hypoxic EVs also contribute to several non-malignant diseases. This review summarizes the effect of hypoxia on EVs cargo in malignant and nonmalignant diseases of different organs.
Collapse
Affiliation(s)
- Vahid Niazi
- Stem Cell Research Center, Golestan University of Medical Science, Gorgan, Iran; School of Advanced Technologies in Medicine, Golestan University of Medical Science, Gorgan, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Klymiuk MC, Speer J, Marco ID, Elashry MI, Heimann M, Wenisch S, Arnhold S. Determination of the miRNA profile of extracellular vesicles from equine mesenchymal stem cells after different treatments. Stem Cell Res Ther 2025; 16:162. [PMID: 40188160 PMCID: PMC11972531 DOI: 10.1186/s13287-025-04287-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/19/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common and incurable disease in humans and animals. To gain a better understanding of the pathogenesis and identify potential treatments, miRNAs will be extracted and analysed from extracellular vesicles (EVs) of equine adipose derived mesenchymal stem cells (AdMSCs). METHODS For this purpose we cultivated and pretreated AdMSCs under different conditions: interleukin 1β, shock wave, chondrogenic differentiation, chondrogenic differentiation under hypoxia, or after senescence. After treatment, EVs were harvested from the cell culture supernatants. Next-generation sequencing (NGS) was used to sequence the miRNAs from the EVs. RESULTS A total of 89 miRNAs whose expression was significantly altered compared with that of an untreated negative control were identified. On average, 53 miRNAs were upregulated and 6 miRNAs were downregulated. Among others, the miRNAs eca-miR-101, eca-miR-143, eca-miR-145, eca-miR-146a, eca-miR-27a, eca-miR-29b, eca-miR-93, eca-miR-98, and eca-miR-221 were significantly increased after the stimulations, which, as known anti-inflammatory miRNAs, could be candidates for therapeutic use in the treatment of OA. CONCLUSION These results lay the foundation for further research into the significance and efficacy of these miRNAs so that this knowledge can be improved in further experiments and, ideally, translated into therapeutic use.
Collapse
Affiliation(s)
- Michele C Klymiuk
- Institute of Veterinary-Anatomy, -Histology and -Embryology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 98, 35392, Giessen, Germany.
| | - Julia Speer
- Institute of Veterinary-Anatomy, -Histology and -Embryology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 98, 35392, Giessen, Germany
| | - Isabelle De Marco
- Clinic of Small Animals, c/o Institute of Veterinary-Anatomy, -Histology and -Embryology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 98, 35392, Giessen, Germany
| | - Mohamed I Elashry
- Institute of Veterinary-Anatomy, -Histology and -Embryology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 98, 35392, Giessen, Germany
| | - Manuela Heimann
- Institute of Veterinary-Anatomy, -Histology and -Embryology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 98, 35392, Giessen, Germany
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary-Anatomy, -Histology and -Embryology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 98, 35392, Giessen, Germany
| | - Stefan Arnhold
- Institute of Veterinary-Anatomy, -Histology and -Embryology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 98, 35392, Giessen, Germany
| |
Collapse
|
7
|
Gao Y, Zhang Y, Mi N, Miao W, Zhang J, Liu Y, Li Z, Song J, Li X, Guan W, Bai C. Exploring the link between M1 macrophages and EMT of amniotic epithelial cells: implications for premature rupture of membranes. J Nanobiotechnology 2025; 23:163. [PMID: 40033278 PMCID: PMC11877754 DOI: 10.1186/s12951-025-03192-6] [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: 01/23/2024] [Accepted: 02/01/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Despite increasing evidence supporting the role of an amniotic epithelial-mesenchymal transition (EMT) in the premature rupture of membranes (PROMs), it remains unclear if extracellular vesicle (EV) derived from M1 macrophages play a critical role in triggering the EMT of amniotic epithelial cells (AECs). RESULTS This study revealed that under inflammatory conditions, EV-miR-146a/155 from M1 macrophages could trigger EMTs and MMP-9 transcription in AECs, elevating the risk of PROM in both mice and humans. Introduction of EV-miR-155 led to inhibition of Ehf expression and reduced E-cadherin transcription in AECs. Meanwhile, EV-miR-146a activated the β-catenin/Tcf7 complex to promote the transcription of Snail, MMP-9, and miR-146a/155, inducing EMTs. Subsequently, EMT induction in AECs is associated with a loss of epithelial characteristics, disruption of cellular junctions, widening of intercellular spaces, and diminished biomechanical properties of the amniotic membrane. CONCLUSION Inflammatory stimulation prompts the polarization of macrophages in amniotic fluid into the M1 type, which subsequently secrete EVs laden with inflammatory miRNAs. These EVs trigger the EMT of AECs, causing the loss of their epithelial phenotype. Consequently, the biomechanical properties of the amnion deteriorate, ultimately leading to its rupture, posing risks relevant to pregnancy complications such as premature rupture of membranes. The results of this study provide insights into the pathogenesis of PROM and will aid in treatment development.
Collapse
Affiliation(s)
- Yuhua Gao
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No.2 Yuanmingyuan West Street, Haidian District, Beijing, 100193, P. R. China
| | - Yanan Zhang
- Department of Obstetrics, Affiliated Hospital of Jining Medical University, Jining, 272000, P. R. China
| | - Ningning Mi
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, No. 666, Wusu Road, Lin'an, 311300, P. R. China
| | - Wang Miao
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
| | - Jingmiao Zhang
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
| | - Yize Liu
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
| | - Zhikun Li
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
| | - Jiaxun Song
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China
| | - Xiangchen Li
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, No. 666, Wusu Road, Lin'an, 311300, P. R. China.
| | - Weijun Guan
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No.2 Yuanmingyuan West Street, Haidian District, Beijing, 100193, P. R. China.
| | - Chunyu Bai
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, No.133 Hehua Road, Jining, Shandong, 272067, P. R. China.
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, No.2 Yuanmingyuan West Street, Haidian District, Beijing, 100193, P. R. China.
| |
Collapse
|
8
|
Lu D, Aji G, Li G, Li Y, Fang W, Zhang S, Yu R, Jiang S, Gao X, Jiang Y, Wang Q. ZDHHC18 promotes renal fibrosis development by regulating HRAS palmitoylation. J Clin Invest 2025; 135:e180242. [PMID: 39913299 PMCID: PMC11910235 DOI: 10.1172/jci180242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 01/24/2025] [Indexed: 03/18/2025] Open
Abstract
Fibrosis is the final common pathway leading to end-stage chronic kidney disease (CKD). However, the function of protein palmitoylation in renal fibrosis and the underlying mechanisms remain unclear. In this study, we observed that expression of the palmitoyltransferase ZDHHC18 was significantly elevated in unilateral ureteral obstruction (UUO) and folic acid-induced (FA-induced) renal fibrosis mouse models and was significantly upregulated in fibrotic kidneys of patients with CKD. Functionally, tubule-specific deletion of ZDHHC18 attenuated tubular epithelial cells' partial epithelial-mesenchymal transition (EMT) and then reduced the production of profibrotic cytokines and alleviated tubulointerstitial fibrosis. In contrast, ZDHHC18 overexpression exacerbated progressive renal fibrosis. Mechanistically, ZDHHC18 catalyzed the palmitoylation of HRAS, which was pivotal for its translocation to the plasma membrane and subsequent activation. HRAS palmitoylation promoted downstream phosphorylation of MEK/ERK and further activated Ras-responsive element-binding protein 1 (RREB1), enhancing SMAD binding to the Snai1 cis-regulatory regions. Taken together, our findings suggest that ZDHHC18 plays a crucial role in renal fibrogenesis and represents a potential therapeutic target for combating kidney fibrosis.
Collapse
Affiliation(s)
- Di Lu
- Nephrology Department, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Gulibositan Aji
- Department of Endocrinology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
| | - Guanyu Li
- Nephrology Department, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yue Li
- Nephrology Department, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wenlin Fang
- Department of Pediatrics, Jiangxi Children’s Medical Center, Nanchang, China
| | - Shuai Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ruiqi Yu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Sheng Jiang
- Department of Endocrinology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
| | - Xia Gao
- Nephrology Department, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yuhang Jiang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Qi Wang
- Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
9
|
Peppicelli S, Calorini L, Bianchini F, Papucci L, Magnelli L, Andreucci E. Acidity and hypoxia of tumor microenvironment, a positive interplay in extracellular vesicle release by tumor cells. Cell Oncol (Dordr) 2025; 48:27-41. [PMID: 39023664 PMCID: PMC11850579 DOI: 10.1007/s13402-024-00969-z] [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: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
The complex and continuously evolving features of the tumor microenvironment, varying between tumor histotypes, are characterized by the presence of host cells and tumor cells embedded in a milieu shaped by hypoxia and low pH, resulting from the frequent imbalance between vascularity and tumor cell proliferation. These microenvironmental metabolic stressors play a crucial role in remodeling host cells and tumor cells, contributing to the stimulation of cancer cell heterogeneity, clonal evolution, and multidrug resistance, ultimately leading to progression and metastasis. The extracellular vesicles (EVs), membrane-enclosed structures released into the extracellular milieu by tumor/host cells, are now recognized as critical drivers in the complex intercellular communication between tumor cells and the local cellular components in a hypoxic/acidic microenvironment. Understanding the intricate molecular mechanisms governing the interactions between tumor and host cells within a hypoxic and acidic microenvironment, triggered by the release of EVs, could pave the way for innovative strategies to disrupt the complex interplay of cancer cells with their microenvironment. This approach may contribute to the development of an efficient and safe therapeutic strategy to combat cancer progression. Therefore, we review the major findings on the release of EVs in a hypoxic/acidic tumor microenvironment to appreciate their role in tumor progression toward metastatic disease.
Collapse
Affiliation(s)
- Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy.
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| |
Collapse
|
10
|
Renu K. Exosomes derived from human adipose mesenchymal stem cells act as a therapeutic target for oral submucous fibrosis. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2025:102224. [PMID: 39765310 DOI: 10.1016/j.jormas.2025.102224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
Oral submucosal fibrosis is a highly malignant oral condition that necessitates the use of sophisticated therapeutic procedures. OSF is a multifactorial precancerous condition induced by areca nut chewing, deficiencies in vitamins and trace minerals, immunological aspects, and hereditary factors. Adipose tissue-derived mesenchymal stem cells possess the capability for multidirectional activation and are extensively distributed throughout the body. They have minimal immunogenicity and are extensively utilized in cancer treatment. Exosomes are extracellular vesicles produced by the intracellular route. They are biological carriers comprising microRNA, messenger RNA, lipids and proteins crucial for intercellular communication. ADSC exosomes, serving as a vehicle for miRNA, possess accessibility and little immunogenicity. They can significantly contribute to adipose tissue regrowth, angiogenesis, immunological modulation, and tissue repair. ADSC-Exo exhibits antifibrotic properties and may serve as a potential treatment for OSF. This review presents a novel therapeutic approach and clarifies the precise mechanisms involved in the clinical management of OSF using ADSC-Exo.
Collapse
Affiliation(s)
- Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600077, Tamil Nadu, India.
| |
Collapse
|
11
|
Janosevic D, De Luca T, Eadon MT. The Kidney Precision Medicine Project and Single-Cell Biology of the Injured Proximal Tubule. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:7-22. [PMID: 39332674 PMCID: PMC11686451 DOI: 10.1016/j.ajpath.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) has led to major advances in our understanding of proximal tubule subtypes in health and disease. The proximal tubule serves essential functions in overall homeostasis, but pathologic or physiological perturbations can affect its transcriptomic signature and corresponding tasks. These alterations in proximal tubular cells are often described within a scRNA-seq atlas as cell states, which are pathophysiological subclassifications based on molecular and morphologic changes in a cell's response to that injury compared with its native state. This review describes the major cell states defined in the Kidney Precision Medicine Project's scRNA-seq atlas. It then identifies the overlap between the Kidney Precision Medicine Project and other seminal works that may use different nomenclature or cluster proximal tubule cells at different resolutions to define cell state subtypes. The goal is for the reader to understand the key transcriptomic markers of important cellular injury and regeneration processes across this highly dynamic and evolving field.
Collapse
Affiliation(s)
- Danielle Janosevic
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Thomas De Luca
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Michael T Eadon
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
12
|
Lawson JS, Williams TL. Extracellular vesicles in kidney disease - A veterinary perspective. Vet J 2024; 308:106247. [PMID: 39276847 DOI: 10.1016/j.tvjl.2024.106247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/21/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Extracellular vesicles (EVs) are membrane bound vesicles secreted from cells into the extracellular space which have an emerging role in both normal kidney physiology and the pathophysiology of kidney injury, predominantly as mediators of intercellular communication. EVs contain proteins and RNA cargo which reflect their cell of origin and can be isolated from the urine of cats and dogs. The majority of urinary EVs (uEVs) originate from the kidney, and both the uEV proteome and transcriptome have been investigated as sources of biomarkers of kidney disease. In addition to their possible diagnostic role, EVs may also have therapeutic potential, and veterinary species have been used as models to demonstrate the efficacy of exogenous EVs derived from mesenchymal stromal cells in the treatment of acute kidney injury. Furthermore, bioengineered EVs may represent a novel vehicle for the administration of drugs or therapeutic nucleic acids in kidney disease. This article reviews the biological functions of EVs within the kidney, techniques for their isolation, and their potential use as biomarkers and therapeutic agents, with particular focus on the potential significance to veterinary patients.
Collapse
Affiliation(s)
- Jack S Lawson
- The Royal Veterinary College, Hawkshead Ln, Brookmans Park, Hatfield AL9 7TA, UK.
| | - Timothy L Williams
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| |
Collapse
|
13
|
Li C, Wang X, Tian M, Zhang M, Zhang X, Fu Q, Liu L, Zhang L, Wang H. The JNK-associated leucine zipper protein exerts a protective effect on renal parenchymal injury by limiting the inflammatory secretome in tubular cells. Cell Signal 2024; 124:111428. [PMID: 39307375 DOI: 10.1016/j.cellsig.2024.111428] [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: 06/14/2024] [Revised: 09/01/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
JNK-associated leucine zipper protein (JLP) is a newly identified renal endogenous anti-fibrotic factor that is selectively enriched in renal tubular epithelial cells (TECs). The loss of JLP by TECs is a landmark event that heralds the progression of kidney fibrosis. JLP deficiency ensues a series of pathogenetic cellular processes that are conducive to fibrotic injury. Inflammatory injury is functionally relevant in fibrotic kidneys, and TECs play an essential role in fueling inflammation through aberrant secretions. It is speculated that the loss of JLP in TECs is associated with the relentless inflammation during the development of kidney fibrosis. This study examined the alteration of a panel of inflammatory signatures in TECs under kidney fibrotic circumstances using a Jlp gene-modified unilateral ureteral obstruction (UUO) mouse model or cultured HK-2 cells. It was found that a deficiency of JLP in TECs led to a significant increase in the secretion of inflammatory cytokines including interleukin-1β (IL-1β), tumor necrosis factor (TNF-α), and monocyte chemotactic protein-1 (MCP-1), overactivation of the nuclear factor (NF)-κB/c-Jun N-terminal kinase (JNK) pathway, as well as nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome-mediated pyroptosis in response to pro-fibrotic damage. Additionally, the absence of JLP resulted in enhanced macrophage migration and fibroblast activation as paracrine effects elicited by injured TECs. In conclusion, the loss of JLP in TECs catalyses inflammatory injuries in the development of kidney fibrosis.
Collapse
Affiliation(s)
- Chen Li
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaofei Wang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Maoqing Tian
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Meng Zhang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Zhang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiang Fu
- Paediatric Department, Central Hospital of Jingzhou City, Jingzhou, China
| | - Lunzhi Liu
- Hubei Provincial Clinical Medical Research Center for Nephropathy, Minda Hospital of Hubei Minzu University, Enshi, China.
| | - Lu Zhang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Huiming Wang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
14
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Extracellular Vesicles in Viral Liver Diseases. Viruses 2024; 16:1785. [PMID: 39599900 PMCID: PMC11598962 DOI: 10.3390/v16111785] [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: 09/30/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Extracellular vesicles (EVs) are bilayer vesicles released by cells in the microenvironment of the liver including parenchymal and non-parenchymal cells. They are the third important mechanism in the communications between cells, besides the secretion of cytokines and chemokines and the direct cell-to-cell contact. The aim of this review is to discuss the important role of EVs in viral liver disease, as there is increasing evidence that the transportation of viral proteins, all types of RNA, and viral particles including complete virions is implicated in the pathogenesis of both viral cirrhosis and viral-related hepatocellular carcinoma. The biogenesis of EVs is discussed and their role in the pathogenesis of viral liver diseases is presented. Their use as diagnostic and prognostic biomarkers is also analyzed. Most importantly, the significance of possible novel treatment strategies for liver fibrosis and hepatocellular carcinoma is presented, although available data are based on experimental evidence and clinical trials have not been reported.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece;
| |
Collapse
|
15
|
Sunartvanichkul T, Chaweewannakorn C, Tabtimmai L, Chiangjong W, Iwasaki K, Pattanapanyasat K, Sritanaudomchai H. Apoptosis-induced exosomes from human exfoliated deciduous teeth enhance angiogenesis in human umbilical vein endothelial cells. Sci Rep 2024; 14:27921. [PMID: 39537956 PMCID: PMC11561270 DOI: 10.1038/s41598-024-79692-6] [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: 05/13/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
Exosomes derived from the stem cells of human exfoliated deciduous teeth (SHED) hold promise for tissue regeneration. Apoptotic cells release a variety of extracellular vesicles that affect intercellular communication. This study aimed to investigate the angiogenic effects of SHED-derived exosomes modified via apoptosis induction on human umbilical vein endothelial cells (HUVECs). Apoptosis was induced in SHED via serum starvation for 3 weeks and confirmed by the upregulation of the apoptotic genes, caspase 3 and 9, and via annexin V staining. The apoptotic SHED-derived exosomes were isolated, characterized, and subjected to proteomic analysis. In vitro experiments were performed to assess the effects of apoptotic SHED exosomes on the proliferation, migration, and tube formation of HUVECs. The apoptosis-induced SHED showed increased cell viability and decreased numbers of dead cells compared with those of conventional cultures while retaining their identity as mesenchymal stem cells positive for CD44, CD73, and CD90. The apoptotic SHED-derived exosomes exhibited characteristic features, such as standard size, cup-shaped morphology, and positive staining, for exosomal markers CD9, CD63, and CD81. Proteins associated with apoptosis, programmed cell death, and cellular senescence were downregulated in the apoptotic SHED exosomes, whereas those associated with extracellular matrix organization were upregulated, indicating positive angiogenesis. HUVECs treated with apoptotic SHED exosomes exhibited significantly enhanced proliferation and migration compared with those treated with normal SHED exosomes. The mesh-like structures in the apoptotic SHED exosomes exhibited significantly increased signs of angiogenesis. The findings of this study provide new insights into the potential use of apoptotic SHED-derived exosomes in regenerative medicine.
Collapse
Affiliation(s)
| | | | - Lueacha Tabtimmai
- Department of Biotechnology, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Wararat Chiangjong
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Kengo Iwasaki
- Divison of Creative and Integrated Medicine, Advanced Medicine Research Center, Translation Research Institute for Medical Innovation, Osaka Dental University, Osaka, 573-1121, Japan
| | - Kovit Pattanapanyasat
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Hathaitip Sritanaudomchai
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, 6 Yothi Road, Ratchathewi, Bangkok, 10400, Thailand.
| |
Collapse
|
16
|
Liu C, Li Q, Ma JX, Lu B, Criswell T, Zhang Y. Exosome-mediated renal protection: Halting the progression of fibrosis. Genes Dis 2024; 11:101117. [PMID: 39263535 PMCID: PMC11388648 DOI: 10.1016/j.gendis.2023.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 09/13/2024] Open
Abstract
Renal fibrosis is a complex and multifactorial process that involves inflammation, cell proliferation, collagen, and fibronectin deposition in the kidney, ultimately leading to chronic kidney disease and even end-stage renal disease. The main goal of treatment is to slow down or halt the progression of fibrosis and to improve or preserve kidney function. Despite significant progress made in understanding the underlying mechanisms of renal fibrosis, current therapies have limited renal protection as the disease progresses. Exosomes derived from stem cells are a newer area of research for the treatment of renal fibrosis. Exosomes as nano-sized extracellular vesicles carry proteins, lipids, and nucleic acids, which can be taken up by local or distant cells, serving as mediators of intercellular communication and as drug delivery vehicles. Exosomes deliver molecules that reduce inflammation, renal fibrosis and extracellular matrix protein production, and promote tissue regeneration in animal models of kidney disease. Additionally, they have several advantages over stem cells, such as being non-immunogenic, having low risk of tumor formation, and being easier to produce and store. This review describes the use of natural and engineered exosomes containing therapeutic agents capable of mediating anti-inflammatory and anti-fibrotic processes during both acute kidney injury and chronic kidney disease. Exosome-based therapies will be compared with stem cell-based treatments for tissue regeneration, with a focus on renal protection. Finally, future directions and strategies for improving the therapeutic efficacy of exosomes are discussed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Baisong Lu
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Tracy Criswell
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
17
|
Cortinovis M, Perico N, Remuzzi G. Tubulointerstitial injury in proteinuric chronic kidney diseases. Front Med (Lausanne) 2024; 11:1478697. [PMID: 39529801 PMCID: PMC11550959 DOI: 10.3389/fmed.2024.1478697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Proteinuria is an independent risk factor for chronic kidney disease progression and cardiovascular diseases. Apart from its prognostic role, the load of proteins that pass across the disrupted glomerular capillary wall trigger multiple pathophysiologic processes. These include, among others, intratubular complement activation and excessive proximal tubular reabsorption of filtered proteins, especially albumin and albumin-bound free fatty acids, which can set off several pathways of cellular damage. The activation of these pathways can cause apoptosis of proximal tubular cells and paracrine effects that incite the development of interstitial inflammation and fibrosis, ultimately leading to irreversible kidney injury. In this review, we provide a comprehensive overview of the current understanding on the mechanisms underlying the tubular toxicity of ultrafiltered proteins in the setting of proteinuric chronic kidney diseases. The acquired knowledge is expected to be instrumental for the development of novel therapeutic classes of medications to be tested on top of standard of care with optimized renin-angiotensin-aldosterone blockade and sodium-glucose cotransporter-2 inhibition, in order to further improve the clinical outcomes of patients with proteinuric chronic kidney diseases.
Collapse
Affiliation(s)
- Monica Cortinovis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | | | | |
Collapse
|
18
|
Zhang Y, Wu D, Zhou C, Bai M, Wan Y, Zheng Q, Fan Z, Wang X, Yang C. Engineered extracellular vesicles for tissue repair and regeneration. BURNS & TRAUMA 2024; 12:tkae062. [PMID: 39439545 PMCID: PMC11495891 DOI: 10.1093/burnst/tkae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) are heterogeneous membrane-like vesicles secreted by living cells that are involved in many physiological and pathological processes and act as intermediaries of intercellular communication and molecular transfer. Recent studies have shown that EVs from specific sources regulate tissue repair and regeneration by delivering proteins, lipids, and nucleic acids to target cells as signaling molecules. Nanotechnology breakthroughs have facilitated the development and exploration of engineered EVs for tissue repair. Enhancements through gene editing, surface modification, and content modification have further improved their therapeutic efficacy. This review summarizes the potential of EVs in tissue repair and regeneration, their mechanisms of action, and their research progress in regenerative medicine. This review highlights their design logic through typical examples and explores the development prospects of EVs in tissue repair. The aim of this review is to provide new insights into the design of EVs for tissue repair and regeneration applications, thereby expanding their use in regenerative medicine.
Collapse
Affiliation(s)
- Yan Zhang
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
- School of Public Health, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Dan Wu
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Chen Zhou
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-Sen University, No. 3025 Shennan Middle Road, Futian District, Shenzhen, China
| | - Muran Bai
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Yucheng Wan
- Hospital of Stomatology, Zunyi Medical University, No. 89, Wujiang East Road, Xinpu New District, Zunyi City, Guizhou Province, China
| | - Qing Zheng
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| | - Zhijin Fan
- Institute for Engineering Medicine, Kunming Medical University, No. 1168 Chunrong West Road, Yuhua Street, Chenggong District, Kunming City, Yunnan Province China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, No.81 Meishan Road, Shushan District, Hefei 230032, China
| | - Chun Yang
- College of Basic Medicin, Beihua University, No. 3999 Binjiang East Road, Fengman District, Jilin City, Jilin Province, China
| |
Collapse
|
19
|
Ao R, Liang W, Wang Z, Li Q, Pan X, Zhen Y, An Y. Delivery Strategies of Growth Factors in Cartilage Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39345121 DOI: 10.1089/ten.teb.2024.0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Cartilage plays an important role in supporting soft tissues, reducing joint friction, and distributing pressure. However, its self-repair capacity is limited due to the lack of blood vessels, nerves, and lymphatic systems. Tissue engineering offers a potential solution to promote cartilage regeneration by combining scaffolds, seed cells, and growth factors. Among these, growth factors play a critical role in regulating cell proliferation, differentiation, and extracellular matrix remodeling. However, their instability, susceptibility to degradation and potential side effects limit their effectiveness. This article reviews the main growth factors used in cartilage tissue engineering and their delivery strategies, including affinity-based delivery, carrier-assisted delivery, stimuli-responsive delivery, spatial structure-based delivery, and cell system-based delivery. Each method shows unique advantages in enhancing the delivery efficiency and specificity of growth factors but also faces challenges such as cost, biocompatibility, and safety. Future research needs to further optimize these strategies to achieve more efficient, safe, and economical delivery of growth factors, thereby advancing the clinical application of cartilage tissue engineering.
Collapse
Affiliation(s)
- Rigele Ao
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Wei Liang
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Zimo Wang
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Qiaoyu Li
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Xingyi Pan
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191China
| |
Collapse
|
20
|
Chen W, Gao Y, Liu Y, Luo Y, Xue X, Xiao C, Wei K. Tanshinone IIA Loaded Inhaled Polymer Nanoparticles Alleviate Established Pulmonary Fibrosis. ACS Biomater Sci Eng 2024; 10:6250-6262. [PMID: 39288315 DOI: 10.1021/acsbiomaterials.4c00532] [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] [Indexed: 09/19/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal respiratory disease characterized by chronic, progressive scarring of the lung parenchyma, leading to an irreversible decline in lung function. Apart from supportive care, there is currently no specific treatment available to reverse the disease. Based on the fact that tanshinone IIA (TAN) had an effect on protecting against TGF-β1-induced fibrosis through the inhibition of Smad and non-Smad signal pathways to avoid myofibroblasts activation, this study reported the development of the inhalable tanshinone IIA-loaded chitosan-oligosaccharides-coated poly(lactic-co-glycolic acid) (PLGA) nanoparticles (CPN@TAN) for enhancing the pulmonary delivery of tanshinone IIA to treat pulmonary fibrosis. The CPN@TAN with a size of 206.5 nm exhibited excellent in vitro aerosol delivery characteristics, featuring a mass median aerodynamic diameter (MMAD) of 3.967 ± 0.025 μm and a fine particle fraction (FPF) of 70.516 ± 0.929%. Moreover, the nanoparticles showed good stability during atomization and enhanced the mucosal penetration capabilities. The results of confocal spectroscopy confirmed the potential of the nanoparticles as carriers that facilitated the uptake of drugs by NIH3T3, A549, and MH-S cells. Additionally, the nanoparticles demonstrated good in vitro biocompatibility. In a mouse model of bleomycin-induced pulmonary fibrosis, noninvasive inhalation of aerosol CPN@TAN greatly suppressed collagen formation and facilitated re-epithelialization of the destroyed alveolar epithelium without causing systemic toxicity compared with intravenous administration. Consequently, our noninvasive inhalation drug delivery technology based on polymers may represent a promising paradigm and open the door to overcoming the difficulties associated with managing pulmonary fibrosis.
Collapse
Affiliation(s)
- Wenyu Chen
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yuanyuan Gao
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yuanqi Liu
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yujia Luo
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xinrui Xue
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Chujie Xiao
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| | - Kun Wei
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
21
|
Lumpuy-Castillo J, Amador-Martínez I, Díaz-Rojas M, Lorenzo O, Pedraza-Chaverri J, Sánchez-Lozada LG, Aparicio-Trejo OE. Role of mitochondria in reno-cardiac diseases: A study of bioenergetics, biogenesis, and GSH signaling in disease transition. Redox Biol 2024; 76:103340. [PMID: 39250857 PMCID: PMC11407069 DOI: 10.1016/j.redox.2024.103340] [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: 07/18/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are global health burdens with rising prevalence. Their bidirectional relationship with cardiovascular dysfunction, manifesting as cardio-renal syndromes (CRS) types 3 and 4, underscores the interconnectedness and interdependence of these vital organ systems. Both the kidney and the heart are critically reliant on mitochondrial function. This organelle is currently recognized as a hub in signaling pathways, with emphasis on the redox regulation mediated by glutathione (GSH). Mitochondrial dysfunction, including impaired bioenergetics, redox, and biogenesis pathways, are central to the progression of AKI to CKD and the development of CRS type 3 and 4. This review delves into the metabolic reprogramming and mitochondrial redox signaling and biogenesis alterations in AKI, CKD, and CRS. We examine the pathophysiological mechanisms involving GSH redox signaling and the AMP-activated protein kinase (AMPK)-sirtuin (SIRT)1/3-peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α) axis in these conditions. Additionally, we explore the therapeutic potential of GSH synthesis inducers in mitigating these mitochondrial dysfunctions, as well as their effects on inflammation and the progression of CKD and CRS types 3 and 4.
Collapse
Affiliation(s)
- Jairo Lumpuy-Castillo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz-Ciberdem, Medicine Department, Autonomous University, 28040, Madrid, Spain.
| | - Isabel Amador-Martínez
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico; Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04510, Mexico City, Mexico.
| | - Miriam Díaz-Rojas
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, 43210, Columbus, Ohio, USA.
| | - Oscar Lorenzo
- Laboratory of Diabetes and Vascular Pathology, IIS-Fundación Jiménez Díaz-Ciberdem, Medicine Department, Autonomous University, 28040, Madrid, Spain.
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, 04510, Mexico City, Mexico.
| | - Laura Gabriela Sánchez-Lozada
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico.
| | - Omar Emiliano Aparicio-Trejo
- Department of Cardio-Renal Physiopathology, National Institute of Cardiology Ignacio Chávez, 14080, Mexico City, Mexico.
| |
Collapse
|
22
|
Cerrotti G, Buratta S, Latella R, Calzoni E, Cusumano G, Bertoldi A, Porcellati S, Emiliani C, Urbanelli L. Hitting the target: cell signaling pathways modulation by extracellular vesicles. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:527-552. [PMID: 39697631 PMCID: PMC11648414 DOI: 10.20517/evcna.2024.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/10/2024] [Accepted: 09/18/2024] [Indexed: 12/20/2024]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed nanoparticles released outside the cell. EVs have drawn attention not only for their role in cell waste disposal, but also as additional tools for cell-to-cell communication. Their complex contents include not only lipids, but also proteins, nucleic acids (RNA, DNA), and metabolites. A large part of these molecules are involved in mediating or influencing signal transduction in target cells. In multicellular organisms, EVs have been suggested to modulate signals in cells localized either in the neighboring tissue or in distant regions of the body by interacting with the cell surface or by entering the cells via endocytosis or membrane fusion. Most of the EV-modulated cell signaling pathways have drawn considerable attention because they affect morphogenetic signaling pathways, as well as pathways activated by cytokines and growth factors. Therefore, they are implicated in relevant biological processes, such as embryonic development, cancer initiation and spreading, tissue differentiation and repair, and immune response. Furthermore, it has recently emerged that multicellular organisms interact with and receive signals through EVs released by their microbiota as well as by edible plants. This review reports studies investigating EV-mediated signaling in target mammalian cells, with a focus on key pathways for organism development, organ homeostasis, cell differentiation and immune response.
Collapse
Affiliation(s)
- Giada Cerrotti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Raffaella Latella
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Eleonora Calzoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Gaia Cusumano
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Agnese Bertoldi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Serena Porcellati
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Perugia 06123, Italy
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Perugia 06123, Italy
| |
Collapse
|
23
|
Song A, Yan R, Xiong W, Xiang H, Huang J, Jiang A, Zhang C. Early growth response protein 2 promotes partial epithelial-mesenchymal transition by phosphorylating Smad3 during renal fibrosis. Transl Res 2024; 271:13-25. [PMID: 38679230 DOI: 10.1016/j.trsl.2024.04.005] [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: 02/06/2024] [Revised: 03/30/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
Chronic kidney disease (CKD) is a serious health problem worldwide, which ultimately leads to end-stage renal disease (ESRD). Renal fibrosis is the common pathway and major pathological manifestation for various CKD proceeding to ESRD. However, the underlying mechanisms and effective therapies are still ambiguous. Early growth response 2 (EGR2) is reportedly involved in organ formation and cell differentiation. To determine the role of EGR2 in renal fibrosis, we respectively confirmed the increased expression of EGR2 in kidney specimens from both CKD patients and mice with location in proximal tubules. Genetic deletion of EGR2 attenuated obstructive nephropathy while EGR2 overexpression further promoted renal fibrosis in mice subjected to unilateral ureteral obstruction (UUO) due to extracellular matrix (ECM) deposition mediating by partial epithelial-mesenchymal transition (EMT) as well as imbalance between matrix metalloproteinases (MMPs) and tissue inhibitor of MMPs (TIMPs). We found that EGR2 played a critical role in Smad3 phosphorylation, and inhibition of EGR2 reduced partial EMT leading to blockade of ECM accumulation in cultured human kidney 2 cells (HK2) treated with transforming growth factor β1 (TGF-β1). In addition, the transcription co-stimulator signal transducer and activator of transcription 3 (STAT3) phosphorylation was confirmed to regulate the transcription level of EGR2 in TGF-β1-induced HK2 cells. In conclusion, this study demonstrated that EGR2 played a pathogenic role in renal fibrosis by a p-STAT3-EGR2-p-Smad3 axis. Thus, targeting EGR2 could be a promising strategy for CKD treatment.
Collapse
Affiliation(s)
- Anni Song
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruiwei Yan
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Xiong
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huiling Xiang
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Huang
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anni Jiang
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Zhang
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
24
|
Youssef KK, Nieto MA. Epithelial-mesenchymal transition in tissue repair and degeneration. Nat Rev Mol Cell Biol 2024; 25:720-739. [PMID: 38684869 DOI: 10.1038/s41580-024-00733-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/02/2024]
Abstract
Epithelial-mesenchymal transitions (EMTs) are the epitome of cell plasticity in embryonic development and cancer; during EMT, epithelial cells undergo dramatic phenotypic changes and become able to migrate to form different tissues or give rise to metastases, respectively. The importance of EMTs in other contexts, such as tissue repair and fibrosis in the adult, has become increasingly recognized and studied. In this Review, we discuss the function of EMT in the adult after tissue damage and compare features of embryonic and adult EMT. Whereas sustained EMT leads to adult tissue degeneration, fibrosis and organ failure, its transient activation, which confers phenotypic and functional plasticity on somatic cells, promotes tissue repair after damage. Understanding the mechanisms and temporal regulation of different EMTs provides insight into how some tissues heal and has the potential to open new therapeutic avenues to promote repair or regeneration of tissue damage that is currently irreversible. We also discuss therapeutic strategies that modulate EMT that hold clinical promise in ameliorating fibrosis, and how precise EMT activation could be harnessed to enhance tissue repair.
Collapse
Affiliation(s)
| | - M Angela Nieto
- Instituto de Neurociencias (CSIC-UMH), Sant Joan d'Alacant, Spain.
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain.
| |
Collapse
|
25
|
Burgy O, Mayr CH, Schenesse D, Fousekis Papakonstantinou E, Ballester B, Sengupta A, She Y, Hu Q, Melo-Narvaéz MC, Jain E, Pestoni JC, Mozurak M, Estrada-Bernal A, Onwuka U, Coughlan C, Parimon T, Chen P, Heimerl T, Bange G, Schmeck BT, Lindner M, Hilgendorff A, Ruppert C, Güenther A, Mann M, Yildirim AÖ, Eickelberg O, Jung AL, Schiller HB, Lehmann M, Burgstaller G, Königshoff M. Fibroblast-derived extracellular vesicles contain SFRP1 and mediate pulmonary fibrosis. JCI Insight 2024; 9:e168889. [PMID: 39315549 PMCID: PMC11457858 DOI: 10.1172/jci.insight.168889] [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: 02/17/2023] [Accepted: 08/07/2024] [Indexed: 09/25/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal chronic lung disease characterized by aberrant intercellular communication, extracellular matrix deposition, and destruction of functional lung tissue. While extracellular vesicles (EVs) accumulate in the IPF lung, their cargo and biological effects remain unclear. We interrogated the proteome of EV and non-EV fractions during pulmonary fibrosis and characterized their contribution to fibrosis. EVs accumulated 14 days after bleomycin challenge, correlating with decreased lung function and initiated fibrogenesis in healthy precision-cut lung slices. Label-free proteomics of bronchoalveolar lavage fluid EVs (BALF-EVs) collected from mice challenged with bleomycin or control identified 107 proteins enriched in fibrotic vesicles. Multiomic analysis revealed fibroblasts as a major cellular source of BALF-EV cargo, which was enriched in secreted frizzled related protein 1 (SFRP1). Sfrp1 deficiency inhibited the activity of fibroblast-derived EVs to potentiate lung fibrosis in vivo. SFRP1 led to increased transitional cell markers, such as keratin 8, and WNT/β-catenin signaling in primary alveolar type 2 cells. SFRP1 was expressed within the IPF lung and localized at the surface of EVs from patient-derived fibroblasts and BALF. Our work reveals altered EV protein cargo in fibrotic EVs promoting fibrogenesis and identifies fibroblast-derived vesicular SFRP1 as a fibrotic mediator and potential therapeutic target for IPF.
Collapse
Affiliation(s)
- Olivier Burgy
- INSERM U1231 Center for Translational and Molecular Medicine (CTM), Faculty of Health Sciences, Université de Bourgogne, Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital Dijon-Bourgogne, Dijon, France
| | - Christoph H. Mayr
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Déborah Schenesse
- INSERM U1231 Center for Translational and Molecular Medicine (CTM), Faculty of Health Sciences, Université de Bourgogne, Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital Dijon-Bourgogne, Dijon, France
- Department of Pulmonary Medicine and Intensive Care Unit, University Hospital Dijon-Bourgogne, Dijon, France
| | | | - Beatriz Ballester
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
- Faculty of Health Sciences, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Arunima Sengupta
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Yixin She
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qianjiang Hu
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Maria Camila Melo-Narvaéz
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Eshita Jain
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Jeanine C. Pestoni
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Molly Mozurak
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adriana Estrada-Bernal
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ugochi Onwuka
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christina Coughlan
- Division of Neurology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Tanyalak Parimon
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Peter Chen
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | - Gert Bange
- Center for Synthetic Microbiology (SYNMIKRO) and
| | - Bernd T. Schmeck
- Department of Pulmonary Medicine and Intensive Care Unit, University Hospital Dijon-Bourgogne, Dijon, France
- Center for Synthetic Microbiology (SYNMIKRO) and
- Core Facility Flow Cytometry – Bacterial Vesicles, Philipps-University Marburg, Marburg, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC) Giessen Biobank, Justus-Liebig-University Giessen, DZL, Giessen, Germany
| | - Michael Lindner
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
- Paracelsus Medical Private University, Salzburg, Austria
| | - Anne Hilgendorff
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Clemens Ruppert
- Universities of Giessen and Marburg Lung Center (UGMLC) Giessen Biobank, Justus-Liebig-University Giessen, DZL, Giessen, Germany
| | - Andreas Güenther
- Universities of Giessen and Marburg Lung Center (UGMLC) Giessen Biobank, Justus-Liebig-University Giessen, DZL, Giessen, Germany
- European IPF Registry (eurIPFreg), Center for Interstitial and Rare Lung Diseases, UGMLC, Justus-Liebig University Giessen, DZL, Giessen, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Oliver Eickelberg
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anna Lena Jung
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Core Facility Flow Cytometry – Bacterial Vesicles, Philipps-University Marburg, Marburg, Germany
| | - Herbert B. Schiller
- Institute of Experimental Pneumology, LMU University Hospital, Ludwig-Maximilians University, Munich, Germany
- Research Unit for Precision Regenerative Medicine, Helmholtz Munich, Munich, Germany
| | - Mareike Lehmann
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Gerald Burgstaller
- Comprehensive Pneumology Center (CPC) with the CPC-M BioArchive and Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Member of the DZL, Munich, Germany
| | - Melanie Königshoff
- Center for Lung Aging and Regeneration (CLAR), Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Geriatric Research Education and Clinical Center (GRECC) at the VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
26
|
Wang L, Wang J, Xu A, Wei L, Pei M, Shen T, Xian X, Yang K, Fei L, Pan Y, Yang H, Wang X. Future embracing: exosomes driving a revolutionary approach to the diagnosis and treatment of idiopathic membranous nephropathy. J Nanobiotechnology 2024; 22:472. [PMID: 39118155 PMCID: PMC11312222 DOI: 10.1186/s12951-024-02633-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: 03/09/2024] [Accepted: 06/13/2024] [Indexed: 08/10/2024] Open
Abstract
Membranous nephropathy (MN) is a leading cause of nephrotic syndrome in adults and is associated with high rates of end-stage renal disease. Early detection and precise interventions are crucial for improving patient prognosis and quality of life. However, the current diagnosis primarily relies on renal biopsies and traditional biomarkers, which have limitations. Additionally, targeted therapeutic strategies are lacking. Exosomes, small vesicles that facilitate intercellular communication, have emerged as potential noninvasive diagnostic markers due to their stability, diverse cargo, and rapid detectability. They also hold promise as carriers for gene and drug delivery, presenting innovative opportunities in renal disease prognosis and treatment. However, research on exosomes in the context of idiopathic membranous nephropathy (IMN) remains limited, with a focus on exploring urinary exosomes as IMN markers. In this review, we summarize the current status of MN diagnosis and treatment, highlight the fundamental characteristics of exosomes, and discuss recent advancements in their application to IMN diagnosis and therapy. We provide insights into the clinical prospects of exosomes in IMN and acknowledge potential challenges. This article aims to offer forward-looking insights into the future of exosome-mediated IMN diagnosis and treatment, indicating a revolutionary transformation in this field.
Collapse
Affiliation(s)
- Lin Wang
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jinxiang Wang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Guangdong, 518107, China
| | - Ao Xu
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lijuan Wei
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
| | - Ming Pei
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
| | - Tuwei Shen
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xian Xian
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Kang Yang
- Nephrology Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Henan, 450099, China
| | - Lingyan Fei
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Yihang Pan
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Guangdong, 518107, China.
| | - Hongtao Yang
- Nephrology Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300381, China.
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, People's Republic of China.
| |
Collapse
|
27
|
Yuan N, Xiao L, Chen J, Liu B, Ren S, Sheng X, Qi X, Wang Y, Chen C, Guo K, Yang X, Yang L, Wang X. CREG1 promotes bovine placental trophoblast cells exosome release by targeting IGF2R and participates in regulating organoid differentiation via exosomes transport. Int J Biol Macromol 2024; 274:133298. [PMID: 38917918 DOI: 10.1016/j.ijbiomac.2024.133298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/18/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Placental exosomes are a kind of intercellular communication media secreted by placental cells during pregnancy, exosomogenesis and release are regulated by many secretory glycoproteins. CREG1 is a kind of secreted glycoprotein widely expressed in various organs and tissues of the body, which inhibits cell proliferation and enhances cell differentiation. The aim of this study was to explore the role of CREG1 in regulating exosomogenesis during the proliferation and differentiation of placental trophoblast cells in early pregnant dairy cows by targeting IGF2R and participating in regulating organoid differentiation via exosomes transport. METHODS Molecular biological methods were firstly used to investigate the expression patterns of CREG1, IGF2R and exosomal marker proteins in early placental development of pregnant dairy cows. Subsequently, the effects of CREG1 on the formation and release of bovine placental trophoblast (BTCs) derived exosomes by targeting IGF2R were investigated. Further, the effects of CREG1 on the change of gene expression patterns along with the transport of exosomes to recipient cells and participate in regulating the differentiation of organoids were explored. RESULTS The expression of CREG1, IGF2R and exosomal marker proteins increased with the increase of pregnancy months during the early evolution of placental trophoblast cells in dairy cows. Overexpression of Creg1 enhanced the genesis and release of exosomes derived from BTCs, while knocking down the expression of Igf2r gene not only inhibited the genesis of exosomes, but also inhibited the genesis and release of exosomes induced by overexpression of CREG1 protein. Interestingly, IGF2R can regulate the expression of CREG1 through reverse secretion. What's more, the occurrence and release of trophoblast-derived exosomes are regulated by CREG1 binding to IGF2R, which subsequently binds to Rab11. CREG1 can not only promote the formation and release of exosomes in donor cells, but also regulate the change of gene expression patterns along with the transport of exosomes to recipient cells and participate in regulating the early development of placenta. CONCLUSIONS Our study confirmed that CREG1 is involved in the exosomogenesis and release of exosomes during the proliferation and differentiation of placental trophoblast cells in early pregnant dairy cows by targeting IGF2R, and is involved in the regulation of organoid differentiation through exosome transport.
Collapse
Affiliation(s)
- Naihan Yuan
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Longfei Xiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Jiaxi Chen
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Bingying Liu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Siqi Ren
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xihui Sheng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xiaolong Qi
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Yingqiu Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Chang Chen
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Kaijun Guo
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xiaowen Yang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lin Yang
- Animal Epidemic prevention and Quarantine center, Huimin District, Hohhot, Inner Mongolia Autonomous Region 010030, China
| | - Xiangguo Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China.
| |
Collapse
|
28
|
Long Z, Luo Y, Yu M, Wang X, Zeng L, Yang K. Targeting ferroptosis: a new therapeutic opportunity for kidney diseases. Front Immunol 2024; 15:1435139. [PMID: 39021564 PMCID: PMC11251909 DOI: 10.3389/fimmu.2024.1435139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Ferroptosis is a form of non-apoptotic regulated cell death (RCD) that depends on iron and is characterized by the accumulation of lipid peroxides to lethal levels. Ferroptosis involves multiple pathways including redox balance, iron regulation, mitochondrial function, and amino acid, lipid, and glycometabolism. Furthermore, various disease-related signaling pathways also play a role in regulating the process of iron oxidation. In recent years, with the emergence of the concept of ferroptosis and the in-depth study of its mechanisms, ferroptosis is closely associated with various biological conditions related to kidney diseases, including kidney organ development, aging, immunity, and cancer. This article reviews the development of the concept of ferroptosis, the mechanisms of ferroptosis (including GSH-GPX4, FSP1-CoQ1, DHODH-CoQ10, GCH1-BH4, and MBOAT1/2 pathways), and the latest research progress on its involvement in kidney diseases. It summarizes research on ferroptosis in kidney diseases within the frameworks of metabolism, reactive oxygen biology, and iron biology. The article introduces key regulatory factors and mechanisms of ferroptosis in kidney diseases, as well as important concepts and major open questions in ferroptosis and related natural compounds. It is hoped that in future research, further breakthroughs can be made in understanding the regulation mechanism of ferroptosis and utilizing ferroptosis to promote treatments for kidney diseases, such as acute kidney injury(AKI), chronic kidney disease (CKD), diabetic nephropathy(DN), and renal cell carcinoma. This paves the way for a new approach to research, prevent, and treat clinical kidney diseases.
Collapse
Affiliation(s)
- Zhiyong Long
- Department of Physical Medicine and Rehabilitation, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanfang Luo
- Department of Nephrology, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Min Yu
- Department of Physical Medicine and Rehabilitation, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyan Wang
- Department of Nephrology, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Liuting Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
29
|
Komatsu S, Kato N, Kitai H, Funahashi Y, Noda Y, Tsubota S, Tanaka A, Sato Y, Maeda K, Saito S, Furuhashi K, Ishimoto T, Kosugi T, Maruyama S, Kadomatsu K. Detecting and exploring kidney-derived extracellular vesicles in plasma. Clin Exp Nephrol 2024; 28:617-628. [PMID: 38436899 PMCID: PMC11190017 DOI: 10.1007/s10157-024-02464-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/13/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Extracellular vesicles (EVs) have received considerable attention as ideal biomarkers for kidney diseases. Most reports have focused on urinary EVs, that are mainly derived from the cells in the urinary tract. However, the detection and the application of kidney-derived EVs in plasma remains uncertain. METHODS We examined the kidney-derived small EVs (sEVs) in plasma that were supposedly released from renal mesangial and glomerular endothelial cells, using clinical samples from healthy controls and patients with kidney transplants. Plasma from healthy controls underwent ultracentrifugation, followed by on-bead flow cytometry, targeting α8 integrin, an antigen-specific to mesangial cells. To confirm the presence of kidney-derived sEVs in peripheral blood, plasma from ABO-incompatible kidney transplant recipients was ultracentrifuged, followed by western blotting for donor blood type antigens. RESULTS Immunohistochemistry and immunoelectron microscopy confirmed α8 integrin expression in kidney mesangial cells and their sEVs. The CD9-α8 integrin double-positive sEVs were successfully detected using on-bead flow cytometry. Western blot analysis further revealed transplanted kidney-derived sEVs containing blood type B antigens in non-blood type B recipients, who had received kidneys from blood type B donors. Notably, a patient experiencing graft kidney loss exhibited diminished signals of sEVs containing donor blood type antigens. CONCLUSION Our findings demonstrate the potential usefulness of kidney-derived sEVs in plasma in future research for kidney diseases.
Collapse
Affiliation(s)
- Shintaro Komatsu
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Division of Molecular Oncology, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Noritoshi Kato
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
| | - Hiroki Kitai
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yoshio Funahashi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuhei Noda
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shoma Tsubota
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Akihito Tanaka
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuka Sato
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kayaho Maeda
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shoji Saito
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kazuhiro Furuhashi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Takuji Ishimoto
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Tomoki Kosugi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
30
|
Cui HS, Joo SY, Cho YS, Lee YR, Ro YM, Kwak IS, Hur GY, Seo CH. Exosomes Derived from Hypertrophic Scar Fibroblasts Suppress Melanogenesis in Normal Human Epidermal Melanocytes. Int J Mol Sci 2024; 25:7236. [PMID: 39000342 PMCID: PMC11241421 DOI: 10.3390/ijms25137236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Post-burn hypertrophic scars often exhibit abnormal pigmentation. Exosomes play important roles in maintaining normal physiological homeostasis and in the pathological development of diseases. This study investigated the effects of the exosomes derived from hypertrophic scar fibroblasts (HTSFs) on melanocytes, which are pigment-producing cells. Normal fibroblasts (NFs) and HTSFs were isolated and cultured from normal skin and hypertrophic scar (HTS) tissue. Both the NF- and HTSF-exosomes were isolated from a cell culture medium and purified using a column-based technique. The normal human epidermal melanocytes were treated with both exosomes at a concentration of 100 μg/mL at different times. The cell proliferation, melanin content in the medium, apoptotic factors, transcription factors, melanin synthesis enzymes, signaling, signal transduction pathways, and activators of transcription factors (STAT) 1, 3, 5, and 6 were investigated. Compared with the Dulbecco's phosphate-buffered saline (DPBS)-treated controls and NF-exosomes, the HTSF-exosomes decreased the melanocyte proliferation and melanin secretion. The molecular patterns of apoptosis, proliferation, melanin synthesis, Smad and non-Smad signaling, and STATs were altered by the treatment with the HTSF-exosomes. No significant differences were observed between the DPBS-treated control and NF-exosome-treated cells. HTSF-derived exosomes may play a role in the pathological epidermal hypopigmentation observed in patients with HTS.
Collapse
Affiliation(s)
- Hui Song Cui
- Burn Institute, Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul 07247, Republic of Korea; (H.S.C.); (Y.R.L.); (Y.M.R.)
| | - So Young Joo
- Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul 07247, Republic of Korea; (S.Y.J.); (Y.S.C.)
| | - Yoon Soo Cho
- Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul 07247, Republic of Korea; (S.Y.J.); (Y.S.C.)
| | - You Ra Lee
- Burn Institute, Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul 07247, Republic of Korea; (H.S.C.); (Y.R.L.); (Y.M.R.)
| | - Yu Mi Ro
- Burn Institute, Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul 07247, Republic of Korea; (H.S.C.); (Y.R.L.); (Y.M.R.)
| | - In Suk Kwak
- Department of Anesthesiology and Pain Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul 07247, Republic of Korea;
| | - Gi Yeun Hur
- Department of Plastic and Reconstructive Surgery, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul 07247, Republic of Korea
| | - Cheong Hoon Seo
- Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul 07247, Republic of Korea; (S.Y.J.); (Y.S.C.)
| |
Collapse
|
31
|
Wang R, Shi Y, Lv Y, Xie C, Hu Y. The novel insights of epithelial-derived exosomes in various fibrotic diseases. Biomed Pharmacother 2024; 174:116591. [PMID: 38631144 DOI: 10.1016/j.biopha.2024.116591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
The characteristics of fibrosis include the abnormal accumulation of extracellular matrix proteins and abnormal tissue repair caused by injury, infection, and inflammation, leading to a significant increase in organ failure and mortality. Effective and precise treatments are urgently needed to halt and reverse the progression of fibrotic diseases. Exosomes are tiny vesicles derived from endosomes, spanning from 40 to 160 nanometers in diameter, which are expelled into the extracellular matrix environment by various cell types. They play a crucial role in facilitating cell-to-cell communication by transporting a variety of cargoes, including proteins, RNA, and DNA. Epithelial cells serve as the primary barrier against diverse external stimuli that precipitate fibrotic diseases. Numerous research suggests that exosomes from epithelial cells have a significant impact on several fibrotic diseases. An in-depth comprehension of the cellular and molecular mechanisms of epithelial cell-derived exosomes in fibrosis holds promise for advancing the exploration of novel diagnostic biomarkers and clinical drug targets. In this review, we expand upon the pathogenic mechanisms of epithelium-derived exosomes and highlight their role in the fibrotic process by inducing inflammation and activating fibroblasts. In addition, we are particularly interested in the bioactive molecules carried by epithelial-derived exosomes and their potential value in the diagnosis and treatment of fibrosis and delineate the clinical utility of exosomes as an emerging therapeutic modality, highlighting their potential application in addressing various medical conditions.
Collapse
Affiliation(s)
- Rifu Wang
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yuxin Shi
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yonglin Lv
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Changqing Xie
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, China.
| | - Yanjia Hu
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China.
| |
Collapse
|
32
|
Li X, Gao T, Ma X, Zhong J, Qin L, Nian Y, Wang X, Luo Y. Extraction and identification of exosomes from three different sources of human ovarian granulosa cells and analysis of their differential miRNA expression profiles. J Assist Reprod Genet 2024; 41:1371-1385. [PMID: 38492155 PMCID: PMC11143209 DOI: 10.1007/s10815-024-03086-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/05/2024] [Indexed: 03/18/2024] Open
Abstract
OBJECTIVE As important functional cells in the ovary, ovarian granulosa cells are involved in the regulation of oocyte growth and development and play an important role in the study of female fertility preservation. Based on the importance of granulosa cell functionalism, in this study, we analyzed the exosome secretion capacity of human ovarian granulosa cells (SVOG/KGN-cell line, PGC-primary cells) and the differences in their miRNA expression. METHODS Cells were identified by hematoxylin-eosin staining (HE) and FSHR immunofluorescence staining; CCK8 and colony-forming assay were performed to compare cell proliferation capacity; exosomes were extracted and identified by ultra-high speed centrifugation, transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and western blot analysis (WB), and the expression profile of each cellular exosomal miRNA was analyzed by miRNA high-throughput sequencing. RESULTS The proliferative abilities of the three granulosa cells differed, but all had the ability to secrete exosomes. In the exosomes of SVOG, KGN, and PGC cells, 218, 327, and 471 miRNAs were detected, respectively. When compared to the exosomal miRNAs of PGC cells, 111 miRNAs were significantly different in SVOG, and 70 miRNAs were washed two significantly different in KGN cells. These differential miRNA functions were mainly enriched in the cell cycle, cell division/differentiation, multicellular biogenesis, and protein binding. CONCLUSION Human ovarian granulosa cells of different origins are capable of secreting exosomes, but there are still some differences in their exosomes and exosomal miRNAs, and experimental subjects should be selected rationally according to the actual situation.
Collapse
Affiliation(s)
- Xiaorong Li
- Department of the Center for Reproductive Medicine, General Hospital of Ningxia Medical University, No. 1106 of Shengli Road, Xingqin District, Yinchuan, 750004, China.
- Key Laboratory of Fertility Maintenance, Ningxia Medical University, Yinchuan, 750004, China.
| | - Ting Gao
- Key Laboratory of Fertility Maintenance, Ningxia Medical University, Yinchuan, 750004, China
- Key Laboratory of Modernization of Hui Medicine, Ministry of Education, School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiaohong Ma
- Department of the Center for Reproductive Medicine, General Hospital of Ningxia Medical University, No. 1106 of Shengli Road, Xingqin District, Yinchuan, 750004, China
| | - Jiawen Zhong
- Key Laboratory of Fertility Maintenance, Ningxia Medical University, Yinchuan, 750004, China
- Key Laboratory of Modernization of Hui Medicine, Ministry of Education, School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Ling Qin
- Key Laboratory of Modernization of Hui Medicine, Ministry of Education, School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Yan Nian
- Department of the Center for Reproductive Medicine, General Hospital of Ningxia Medical University, No. 1106 of Shengli Road, Xingqin District, Yinchuan, 750004, China
| | - Xueyi Wang
- Key Laboratory of Fertility Maintenance, Ningxia Medical University, Yinchuan, 750004, China
- Key Laboratory of Modernization of Hui Medicine, Ministry of Education, School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Yuxue Luo
- Key Laboratory of Fertility Maintenance, Ningxia Medical University, Yinchuan, 750004, China
- Key Laboratory of Modernization of Hui Medicine, Ministry of Education, School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, 750004, China
| |
Collapse
|
33
|
Jasim SA, Al-Hawary SIS, Kaur I, Ahmad I, Hjazi A, Petkov I, Ali SHJ, Redhee AH, Shuhata Alubiady MH, Al-Ani AM. Critical role of exosome, exosomal non-coding RNAs and non-coding RNAs in head and neck cancer angiogenesis. Pathol Res Pract 2024; 256:155238. [PMID: 38493725 DOI: 10.1016/j.prp.2024.155238] [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: 01/25/2024] [Revised: 02/13/2024] [Accepted: 03/02/2024] [Indexed: 03/19/2024]
Abstract
Head and neck cancer (HNC) refers to the epithelial malignancies of the upper aerodigestive tract. HNCs have a constant yet slow-growing rate with an unsatisfactory overall survival rate globally. The development of new blood vessels from existing blood conduits is regarded as angiogenesis, which is implicated in the growth, progression, and metastasis of cancer. Aberrant angiogenesis is a known contributor to human cancer progression. Representing a promising therapeutic target, the blockade of angiogenesis aids in the reduction of the tumor cells oxygen and nutrient supplies. Despite the promise, the association of existing anti-angiogenic approaches with severe side effects, elevated cancer regrowth rates, and limited survival advantages is incontrovertible. Exosomes appear to have an essential contribution to the support of vascular proliferation, the regulation of tumor growth, tumor invasion, and metastasis, as they are a key mediator of information transfer between cells. In the exocrine region, various types of noncoding RNAs (ncRNAs) identified to be enriched and stable and contribute to the occurrence and progression of cancer. Mounting evidence suggest that exosome-derived ncRNAs are implicated in tumor angiogenesis. In this review, the characteristics of angiogenesis, particularly in HNC, and the impact of ncRNAs on HNC angiogenesis will be outlined. Besides, we aim to provide an insight on the regulatory role of exosomes and exosome-derived ncRNAs in angiogenesis in different types of HNC.
Collapse
Affiliation(s)
| | | | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Iliya Petkov
- Medical University - Sofia, Department of Neurology, Sofia, Bulgaria
| | - Saad Hayif Jasim Ali
- Department of medical laboratory, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq
| | - Ahmed Huseen Redhee
- Medical laboratory technique college, the Islamic University, Najaf, Iraq; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | | | | |
Collapse
|
34
|
Peng YW, Tang R, Xu QY, Mei SY, Zhou Y, Feng JH, Zhang SY, He ZY. Worldwide productivity and research trend of publications concerning extracellular vesicles role in fibrosis: A bibliometric study from 2013 to 2022. Heliyon 2024; 10:e24357. [PMID: 38293443 PMCID: PMC10826165 DOI: 10.1016/j.heliyon.2024.e24357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 12/04/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
Background Fibrosis is a heavy burden on the global healthcare system. Recently, an increasing number of studies have demonstrated that Extracellular vesicles play an important role in intercellular communication under both physiological and pathological conditions. This study aimed to explore the role of extracellular vesicles' in fibrosis using bibliometric methods. Methods Original articles and reviews related to extracellular vesicles and fibrosis were obtained from the Web of Science Core Collection database on November 9, 2022. VOSviewer was used to obtain general information, including co-institution, co-authorship, and co-occurrence visualization maps. The CiteSpace software was used to analyze citation bursts of keywords and references, a timeline view of the top clusters of keywords and cited articles, and the dual map. R package "bibliometrix" was used to analyze annual production, citation per year, collaboration network between countries/regions, thematic evolution map, and historiography network. Results In total, 3376 articles related to extracellular vesicles and fibrosis published from 2013 to 2022 were included in this study, with China and the United States being the top contributors. Shanghai Jiao Tong University has the highest number of publications. The main collaborators were Giovanni Camussi, Stefania Bruno, Marta Tepparo, and Cristina Grange. Journals related to molecular, biology, genetics, health, immunology, and medicine tended to publish literature on extracellular vesicles and fibrosis. "Recovery," "heterogeneity," "degradation," "inflammation," and "mesenchymal stem cells" are the keywords in this research field. Literature on extracellular vesicles and fibrosis associated with several diseases, including "kidney disease," "rheumatoid arthritis," and "skin regeneration" may be the latest hot research field. Conclusions This study provides a comprehensive perspective on extracellular vesicles and fibrosis through a bibliometric analysis of articles published between 2013 and 2022. We identified the most influential countries, institutions, authors, and journals. We provide information on recent research frontiers and trends for scholars interested in the field of extracellular vesicles and fibrosis. Their role in biological processes has great potential to initiate a new upsurge in future research.
Collapse
Affiliation(s)
| | | | - Qiao-Yi Xu
- Department of Critical Care Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shu-Ya Mei
- Department of Critical Care Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yang Zhou
- Department of Critical Care Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jin-Hua Feng
- Department of Critical Care Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shu-Yi Zhang
- Department of Critical Care Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zheng-Yu He
- Department of Critical Care Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| |
Collapse
|
35
|
Li N, Li K, Zhao W, Wang Y, Xu C, Wang Q, Pan L, Li Q, Ji K, He N, Liu Y, Wang J, Zhang M, Yang M, Du L, Liu Q. Small extracellular vesicles from irradiated lung epithelial cells promote the activation of fibroblasts in pulmonary fibrosis. Int J Radiat Biol 2024; 100:268-280. [PMID: 37747344 DOI: 10.1080/09553002.2023.2263550] [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: 11/30/2022] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Alveolar epithelial injury and dysfunction are the risk factors for radiation-induced pulmonary fibrosis (RIPF). However, it is not clear about the relationship between RIPF and the small extracellular vesicles (sEV) secreted by irradiated alveolar epithelial cells. Based on the activation of fibroblasts, this study explored the role of sEV derived from alveolar epithelial cells in RIPF and the potential mechanisms. METHODS Transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and western blotting were used to characterize sEV. Western blotting was used to detect fibrosis-associated proteins. Cell counts and transwell assays were used to evaluate the proliferation and migration ability of fibroblasts. RT-PCR was used to observe the extracellular matrix (ECM) synthesized by fibroblasts, miRNA changes in the sEV were determined by second-generation sequencing. RESULTS TEM, NTA, and western blotting showed the extracellular vesicles with a double-layer membrane structure of approximately 100 nm in diameter. The sEV derived from irradiated A549, HBEC3-KT, and MLE12 cells upregulated FN1 and alpha-SMA proteins expression in fibroblasts and drove the fibroblast to myofibroblast transition, and the sEV from irradiated mouse bronchoalveolar lavage fluid (BALF) affirmed the same results. In addition, the sEV derived from irradiated alveolar epithelial cells significantly increased the migration ability of fibroblasts and the expression of extracellular matrix proteins such as FN1. The results of miRNA sequencing of sEV in BALF of rats with RIPF showed that the metabolic pathway may be important for miRNA to regulate the activation of fibroblasts. CONCLUSION The sEV derived from radiated pulmonary epithelial cells promote the activation, migration and extracellular matrix proteins expression of lung fibroblasts; miRNA in sEV may be an important molecular that affects the activation of lung fibroblasts.
Collapse
Affiliation(s)
- Na Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Kejun Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wenyue Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Chang Xu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Qin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lifeng Pan
- The general surgery department of Chu Hsien-I Memorial Hospital of Tianjin Medical University, Tianjin, China
| | - Qiang Li
- The general surgery department of Chu Hsien-I Memorial Hospital of Tianjin Medical University, Tianjin, China
| | - Kaihua Ji
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ningning He
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jinhan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Manman Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Mengmeng Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Liqing Du
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Qiang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
36
|
Guo C, Cui Y, Jiao M, Yao J, Zhao J, Tian Y, Dong J, Liao L. Crosstalk between proximal tubular epithelial cells and other interstitial cells in tubulointerstitial fibrosis after renal injury. Front Endocrinol (Lausanne) 2024; 14:1256375. [PMID: 38260142 PMCID: PMC10801024 DOI: 10.3389/fendo.2023.1256375] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/22/2023] [Indexed: 01/24/2024] Open
Abstract
The energy needs of tubular epithelial components, especially proximal tubular epithelial cells (PTECs), are high and they heavily depend on aerobic metabolism. As a result, they are particularly vulnerable to various injuries caused by factors such as ischemia, proteinuria, toxins, and elevated glucose levels. Initial metabolic and phenotypic changes in PTECs after injury are likely an attempt at survival and repair. Nevertheless, in cases of recurrent or prolonged injury, PTECs have the potential to undergo a transition to a secretory state, leading to the generation and discharge of diverse bioactive substances, including transforming growth factor-β, Wnt ligands, hepatocyte growth factor, interleukin (IL)-1β, lactic acid, exosomes, and extracellular vesicles. By promoting fibroblast activation, macrophage recruitment, and endothelial cell loss, these bioactive compounds stimulate communication between epithelial cells and other interstitial cells, ultimately worsening renal damage. This review provides a summary of the latest findings on bioactive compounds that facilitate the communication between these cellular categories, ultimately leading to the advancement of tubulointerstitial fibrosis (TIF).
Collapse
Affiliation(s)
- Congcong Guo
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuying Cui
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicin, Jinan, Shandong, China
| | - Mingwen Jiao
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jinming Yao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Junyu Zhao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yutian Tian
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Jianjun Dong
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong Institute of Nephrology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- First Clinical Medical College, Shandong University of Traditional Chinese Medicin, Jinan, Shandong, China
| |
Collapse
|
37
|
Shao Z, Xu J, Xu X, Wang X, Zhou Y, Li Y, Li K. Exosomes Derived from Human Adipose Mesenchymal Stem Cells Inhibits Fibrosis and Treats Oral Submucous Fibrosis via the miR-181a-5p/Smad2 Axis. Tissue Eng Regen Med 2024; 21:123-135. [PMID: 37755664 PMCID: PMC10764695 DOI: 10.1007/s13770-023-00579-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Oral submucous fibrosis (OSF) is a chronic disease with carcinogenic tendency that poses a non-negligible threat to human health. Exosomes derived from human adipose mesenchymal stem cells (ADSC-Exo) reduces visceral and cutaneous fibroses, but their role in OSF has received little attention. The aim of this study was to investigate the effects of ADSC-Exo on OSF and elucidate the mechanism. METHODS In brief, ADSCs were extracted from adipose tissues and subjected to flow cytometry and induction culture. Fibroblasts were isolated from human buccal mucosa and subjected to immunofluorescence. Myofibroblasts were obtained from fibroblasts induced by arecoline and identified. Immunofluorescence assay confirmed that myofibroblasts could take up ADSC-Exo. The effects of ADSC-Exo on the proliferative and migratory capacities of myofibroblasts were examined using the Cell Counting Kit-8 and scratch assay. Real-time quantitative polymerase chain reaction (qPCR) was performed to evaluate mothers against decapentaplegic homolog 2 (Smad2), Smad3, Smad7, collagen type 1 (Col1), Col3, alpha smooth muscle actin (α-SMA), fibronectin, and vimentin. Western blotting was performed to detect phospho (p)-Smad2, Smad2, p-Smad2/3, Smad2/3, Smad7, Col1, Col3, α-SMA, fibronectin, and vimentin. Furthermore, the dual-luciferase reporter assay was performed to prove that miR-181a-5p in ADSC-Exo directly inhibited the expression of Smad2 mRNA to regulate the transforming growth factor beta (TGF-β) pathway. We also performed qPCR and western blotting to verify the results. RESULTS ADSC-Exo could promote the proliferation and migration of myofibroblasts, reduce the expressions of p-smad2, Smad2, p-smad2/3, Smad2/3, Col1, αSMA, fibronectin, and vimentin and elevated the levels of Smad7 and Col3. In addition, miR-181a-5p was highly expressed in ADSC-Exo and bound to the 3'-untranslated region of Smad2. ADSC-Exo enriched with miR-181a-5p reduced collagen production in myofibroblasts and modulated the TGF-β pathway. CONCLUSIONS ADSC-Exo promoted the proliferative and migratory capacities of myofibroblasts and inhibited collagen deposition and trans-differentiation of myofibroblasts in vitro. miR-181a-5p in exosomes targets Smad2 to regulate the TGF-β pathway in myofibroblasts. ADSC-Exo perform antifibrotic actions through the miR-181a-5p/Smad2 axis and may be a promising clinical treatment for OSF.
Collapse
Affiliation(s)
- Zifei Shao
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, 410000, China
| | - Jinhao Xu
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, 410000, China
| | - Xiaoyang Xu
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, 410000, China
| | - Xiang Wang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, 410000, China
| | - Yuxi Zhou
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, 410000, China
| | - Yiyang Li
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, 410000, China
| | - Kun Li
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, 410000, China.
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China.
| |
Collapse
|
38
|
Palma C, Lai A, Scholz‐Romero K, Chittoory H, Van Haeringen B, Carrion F, Handberg A, Lappas M, Lakhani SR, McCart Reed AE, McIntyre HD, Nair S, Salomon C. Differential response of placental cells to high D-glucose and its impact on extracellular vesicle biogenesis and trafficking via small GTPase Ras-related protein RAB-7A. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e135. [PMID: 38938672 PMCID: PMC11080917 DOI: 10.1002/jex2.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 06/29/2024]
Abstract
Placental extracellular vesicles (EVs) can be found in the maternal circulation throughout gestation, and their concentration, content and bioactivity are associated with pregnancy outcomes, including gestational diabetes mellitus (GDM). However, the effect of changes in the maternal microenvironment on the mechanisms associated with the secretion of EVs from placental cells remains to be fully established. Here, we evaluated the effect of high glucose on proteins associated with the trafficking and release of different populations of EVs from placental cells. BeWo and HTR8/SVneo cells were used as placental models and cultured under 5-mM D-glucose (i.e. control) or 25-mM D-glucose (high glucose). Cell-conditioned media (CCM) and cell lysate were collected after 48 h. Different populations of EVs were isolated from CCM by ultracentrifugation (i.e. pellet 2K-g, pellet 10K-g, and pellet 100K-g) and characterised by Nanoparticle Tracking Analysis. Quantitative proteomic analysis (IDA/SWATH) and multiple reaction monitoring protocols at high resolution (MRMHR) were developed to quantify 37 proteins related to biogenesis, trafficking/release and recognition/uptake of EVs. High glucose increased the secretion of total EVs across the pellets from BeWo cells, an effect driven mainly by changes in the small EVs concentration in the CCM. Interestingly, no effect of high glucose on HTR8/SVneo cells EVs secretion was observed. High glucose induces changes in proteins associated with vesicle trafficking in BeWo cells, including Heat Shock Protein Family A (Hsp70) Member 9 (HSPA9) and Member 8 (HSPA8). For HTR8/SVneo, altered proteins including prostaglandin F2α receptor regulatory protein (FPRP), RAB5A, RAB35, RAB5B, and RB11B, STAM1 and TSG101. These proteins are associated with the secretion and trafficking of EVs, which could explain in part, changes in the levels of circulating EVs in diabetic pregnancies. Further, we identified that proteins RAB11B, PDCD6IP, STAM, HSPA9, HSPA8, SDCBP, RAB5B, RAB5A, RAB7A and ERAP1 regulate EV release in response to high and low glucose when overexpressed in cells. Interestingly, immunohistochemistry analysis of RAB7A revealed distinct changes in placental tissues obtained from women with normal glucose tolerance (NGT, n = 6) and those with GDM (n = 6), influenced by diet or insulin treatment. High glucose regulation of proteins involved in intercellular dynamics and the trafficking of multivesicular bodies to the plasma membrane in placental cells is relevant in the context of GDM pregnancies.
Collapse
Affiliation(s)
- Carlos Palma
- Translational Extracellular Vesicles in Obstetrics and Gynae‐Oncology Group, Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's HospitalThe University of QueenslandBrisbaneQueenslandAustralia
| | - Andrew Lai
- Translational Extracellular Vesicles in Obstetrics and Gynae‐Oncology Group, Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's HospitalThe University of QueenslandBrisbaneQueenslandAustralia
| | - Katherin Scholz‐Romero
- Translational Extracellular Vesicles in Obstetrics and Gynae‐Oncology Group, Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's HospitalThe University of QueenslandBrisbaneQueenslandAustralia
| | - Haarika Chittoory
- UQ Centre for Clinical Research, Faculty of MedicineThe University of QueenslandBrisbaneAustralia
| | - Benjamin Van Haeringen
- UQ Centre for Clinical Research, Faculty of MedicineThe University of QueenslandBrisbaneAustralia
- Pathology QueenslandThe Royal Brisbane and Women's HospitalBrisbaneAustralia
| | - Flavio Carrion
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la SaludUniversidad del AlbaSantiagoChile
| | - Aase Handberg
- Department of Clinical BiochemistryAalborg University HospitalAalborgDenmark
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and GynaecologyUniversity of MelbourneVictoriaAustralia
- Mercy Perinatal Research CentreMercy Hospital for WomenVictoriaAustralia
| | - Sunil R Lakhani
- UQ Centre for Clinical Research, Faculty of MedicineThe University of QueenslandBrisbaneAustralia
- Pathology QueenslandThe Royal Brisbane and Women's HospitalBrisbaneAustralia
| | - Amy E McCart Reed
- UQ Centre for Clinical Research, Faculty of MedicineThe University of QueenslandBrisbaneAustralia
| | - H. David McIntyre
- Department of Obstetric Medicine, Mater Health Brisbane, Queensland and Mater ResearchThe University of QueenslandSouth BrisbaneQueenslandAustralia
| | - Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae‐Oncology Group, Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's HospitalThe University of QueenslandBrisbaneQueenslandAustralia
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae‐Oncology Group, Faculty of Medicine, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's HospitalThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
39
|
Grange C, Dalmasso A, Cortez JJ, Spokeviciute B, Bussolati B. Exploring the role of urinary extracellular vesicles in kidney physiology, aging, and disease progression. Am J Physiol Cell Physiol 2023; 325:C1439-C1450. [PMID: 37842748 PMCID: PMC10861146 DOI: 10.1152/ajpcell.00349.2023] [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: 07/27/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023]
Abstract
Extracellular vesicles (EVs), membranous vesicles present in all body fluids, are considered important messengers, carrying their information over long distance and modulating the gene expression profile of recipient cells. EVs collected in urine (uEVs) are mainly originated from the apical part of urogenital tract, following the urine flow. Moreover, bacterial-derived EVs are present within urine and may reflect the composition of microbiota. Consolidated evidence has established the involvement of uEVs in renal physiology, being responsible for glomerular and tubular cross talk and among different tubular segments. uEVs may also be involved in other physiological functions such as modulation of innate immunity, coagulation, or metabolic activities. Furthermore, it has been recently remonstrated that age, sex, endurance excise, and lifestyle may influence uEV composition and release, modifying their cargo. On the other hand, uEVs appear modulators of different urogenital pathological conditions, triggering disease progression. uEVs sustain fibrosis and inflammation processes, both involved in acute and chronic kidney diseases, aging, and stone formation. The molecular signature of uEVs collected from diseased patients can be of interest for understanding kidney physiopathology and for identifying diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Cristina Grange
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alessia Dalmasso
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Judiel John Cortez
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Beatrice Spokeviciute
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
40
|
Ganguin AA, Skorup I, Streb S, Othman A, Luciani P. Formation and Investigation of Cell-Derived Nanovesicles as Potential Therapeutics against Chronic Liver Disease. Adv Healthc Mater 2023; 12:e2300811. [PMID: 37669775 PMCID: PMC11468924 DOI: 10.1002/adhm.202300811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/04/2023] [Indexed: 09/07/2023]
Abstract
A new therapeutic approach using cell-derived nanovesicles (cdNVs) is offered here to overcome the lack of effective treatments for liver fibrosis, a reversible chronic liver disease. To achieve this goal the formation and purification of cdNVs from untreated, quiescent-like, or activated LX-2 cells, an immortalized human hepatic stellate cell (HSC) line with key features of transdifferentiated HSCs are established. Analysis of the genotype and phenotype of naïve and transdifferentiated LX-2 cells activated through transforming growth factor beta 1, following treatment with cdNVs, reveals a concentration-dependent fibrosis regression. The beneficial fibrosis-resolving effects of cdNVs are linked to their biomolecular corona. Liposomes generated using lipids extracted from cdNVs exhibit a reduced antifibrotic response in perpetuated LX-2 cells and show a reduced cellular uptake. However, incubation with soluble factors collected during purification results in a new corona, thereby restoring fibrosis regression activity. Overall, cdNVs display encouraging therapeutic properties, making them a promising candidate for the development of liver fibrosis resolving therapeutics.
Collapse
Affiliation(s)
- Aymar Abel Ganguin
- Department of ChemistryBiochemistry and Pharmaceutical SciencesUniversity of BernBern3012Switzerland
| | - Ivo Skorup
- Department of ChemistryBiochemistry and Pharmaceutical SciencesUniversity of BernBern3012Switzerland
| | - Sebastian Streb
- Functional Genomics Center Zurich (FGCZ)University of Zurich/ETH ZurichZurich8057Switzerland
| | - Alaa Othman
- Functional Genomics Center Zurich (FGCZ)University of Zurich/ETH ZurichZurich8057Switzerland
| | - Paola Luciani
- Department of ChemistryBiochemistry and Pharmaceutical SciencesUniversity of BernBern3012Switzerland
| |
Collapse
|
41
|
Wang Y, Shen X, Song S, Chen Y, Wang Y, Liao J, Chen N, Zeng L. Mesenchymal stem cell-derived exosomes and skin photoaging: From basic research to practical application. PHOTODERMATOLOGY, PHOTOIMMUNOLOGY & PHOTOMEDICINE 2023; 39:556-566. [PMID: 37605539 DOI: 10.1111/phpp.12910] [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: 02/20/2023] [Revised: 06/25/2023] [Accepted: 08/01/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND Skin photoaging is a condition caused by long-term exposure to ultraviolet irradiation, resulting in a variety of changes in the skin, such as capillary dilation, increased or absent pigmentation, dryness, sagging, and wrinkles. Stem cells possess a remarkable antioxidant capacity and the ability to proliferate, differentiate, and migrate, and their main mode of action is through paracrine secretion, with exosomes being the primary form of secretion. Stem cell-derived exosomes contain a variety of growth factors and cytokines and may have great potential to promote skin repair and delay skin ageing. METHODS This review focuses on the mechanisms of UV-induced skin photoaging, the research progress of stem cell exosomes against skin photoaging, emerging application approaches and limitations in the application of exosome therapy. RESULT Exosomes derived from various stem cells have the potential to prevent skin photoaging. CONCLUSION The combination with novel materials may be a key step for their practical application, which could be an important direction for future basic research and practical applications.
Collapse
Affiliation(s)
- Yihao Wang
- Center of Burn & Plastic and Wound Healing Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Xu Shen
- Center of Burn & Plastic and Wound Healing Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Shenghua Song
- Center of Burn & Plastic and Wound Healing Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yan Chen
- Center of Burn & Plastic and Wound Healing Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yiping Wang
- Center of Burn & Plastic and Wound Healing Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Junlin Liao
- Center of Burn & Plastic and Wound Healing Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Nian Chen
- Center of Burn & Plastic and Wound Healing Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Li Zeng
- Center of Burn & Plastic and Wound Healing Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, China
| |
Collapse
|
42
|
Abinti M, Favi E, Alfieri CM, Zanoni F, Armelloni S, Ferraresso M, Cantaluppi V, Castellano G. Update on current and potential application of extracellular vesicles in kidney transplantation. Am J Transplant 2023; 23:1673-1693. [PMID: 37517555 DOI: 10.1016/j.ajt.2023.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/28/2023] [Accepted: 07/17/2023] [Indexed: 08/01/2023]
Abstract
Kidney transplantation (KT) is the best treatment for end-stage kidney disease. However, early diagnosis of graft injury remains challenging, mainly because of the lack of accurate and noninvasive diagnostic techniques. Improving graft outcomes is equally demanding, as is the development of innovative therapies. Many research efforts are focusing on extracellular vesicles, cellular particles free in each body fluid that have shown promising results as precise markers of damage and potential therapeutic targets in many diseases, including the renal field. In fact, through their receptors and cargo, they act in damage response and immune modulation. In transplantation, they may be used to determine organ quality and aging, the presence of delayed graft function, rejection, and many other transplant-related pathologies. Moreover, their low immunogenicity and safe profile make them ideal for drug delivery and the development of therapies to improve KT outcomes. In this review, we summarize current evidence about extracellular vesicles in KT, starting with their characteristics and major laboratory techniques for isolation and characterization. Then, we discuss their use as potential markers of damage and as therapeutic targets, discussing their promising use in clinical practice as a form of liquid biopsy.
Collapse
Affiliation(s)
- Matteo Abinti
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Evaldo Favi
- Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Carlo Maria Alfieri
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Francesca Zanoni
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, New York, USA
| | - Silvia Armelloni
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Mariano Ferraresso
- Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Vincenzo Cantaluppi
- Nephrology and Kidney Transplant Unit, Department of Translational Medicine (DIMET), University of Piemonte Orientale (UPO), "Maggiore della Carita" University Hospital, Novara, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
43
|
Nørgård MØ, Svenningsen P. Acute Kidney Injury by Ischemia/Reperfusion and Extracellular Vesicles. Int J Mol Sci 2023; 24:15312. [PMID: 37894994 PMCID: PMC10607034 DOI: 10.3390/ijms242015312] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Acute kidney injury (AKI) is often caused by ischemia-reperfusion injury (IRI). IRI significantly affects kidney metabolism, which elicits pro-inflammatory responses and kidney injury. The ischemia/reperfusion of the kidney is associated with transient high mitochondrial-derived reactive oxygen species (ROS) production rates. Excessive mitochondrial-derived ROS damages cellular components and, together with other pathogenic mechanisms, elicits a range of acute injury mechanisms that impair kidney function. Mitochondrial-derived ROS production also stimulates epithelial cell secretion of extracellular vesicles (EVs) containing RNAs, lipids, and proteins, suggesting that EVs are involved in AKI pathogenesis. This literature review focuses on how EV secretion is stimulated during ischemia/reperfusion and how cell-specific EVs and their molecular cargo may modify the IRI process. Moreover, critical pitfalls in the analysis of kidney epithelial-derived EVs are described. In particular, we will focus on how the release of kidney epithelial EVs is affected during tissue analyses and how this may confound data on cell-to-cell signaling. By increasing awareness of methodological pitfalls in renal EV research, the risk of false negatives can be mitigated. This will improve future EV data interpretation regarding EVs contribution to AKI pathogenesis and their potential as biomarkers or treatments for AKI.
Collapse
Affiliation(s)
| | - Per Svenningsen
- Department of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark;
| |
Collapse
|
44
|
Liu X, Liu Z, Wang C, Miao J, Zhou S, Ren Q, Jia N, Zhou L, Liu Y. Kidney tubular epithelial cells control interstitial fibroblast fate by releasing TNFAIP8-encapsulated exosomes. Cell Death Dis 2023; 14:672. [PMID: 37828075 PMCID: PMC10570316 DOI: 10.1038/s41419-023-06209-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 09/18/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023]
Abstract
Kidney fibrosis, characterized by the activation and expansion of the matrix-producing fibroblasts, is the common outcome of chronic kidney disease (CKD). While fibroblast proliferation is well studied in CKD, little is known about the regulation and mechanism of fibroblast depletion. Here, we show that exosomes derived from stressed/injured tubules play a pivotal role in dictating fibroblast apoptosis and fate. When human kidney tubular cells (HK-2) were stimulated with TGF-β1, they produced and released increased amounts of exosomes (TGFβ-Exo), which prevented renal interstitial fibroblasts from apoptosis. In vivo, injections of TGFβ-Exo promoted renal fibroblast survival, whereas blockade of exosome secretion accelerated fibroblast apoptosis in obstructive nephropathy. Proteomics profiling identified the tumor necrosis factor-α-induced protein 8 (TNFAIP8) as a key component enriched in TGFβ-Exo. TNFAIP8 was induced in renal tubular epithelium and enriched in the exosomes from fibrotic kidneys. Knockdown of TNFAIP8 in tubular cells abolished the ability of TGFβ-Exo to prevent fibroblast apoptosis. In vivo, gain- or loss- of TNFAIP8 prevented or aggravated renal fibroblast apoptosis after obstructive injury. Mechanistically, exosomal-TNFAIP8 promoted p53 ubiquitination leading to its degradation, thereby inhibiting fibroblasts apoptosis and inducing their proliferation. Collectively, these results indicate that tubule-derived exosomes play a critical role in controlling the size of fibroblast population during renal fibrogenesis through shuttling TNFAIP8 to block p53 signaling. Strategies to target exosomes may be effective strategies for the therapy of fibrotic CKD.
Collapse
Affiliation(s)
- Xi Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Zhao Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cong Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinhua Miao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Ren
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Nan Jia
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Guangdong Provincial Institute of Nephrology, Guangzhou, China.
| |
Collapse
|
45
|
Fu J, Song W, Hao Z, Fan M, Li Y. Research trends and hotspots of exosomes in respiratory diseases. Medicine (Baltimore) 2023; 102:e35381. [PMID: 37773786 PMCID: PMC10545307 DOI: 10.1097/md.0000000000035381] [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: 06/13/2023] [Accepted: 09/04/2023] [Indexed: 10/01/2023] Open
Abstract
Currently, theoretical studies on exosomes in respiratory diseases have received much attention from many scholars and have made remarkable progress, which has inestimable value and potential in future clinical and scientific research. Unfortunately, no scholar has yet addressed this field's bibliometric analysis and summary. We aim to comprehensively and profoundly study and explore the present situation and highlights of exosome research at the stage of respiratory diseases and to provide meaningful insights for the future development of this field. The WOSCC literature was gathered for the study using bibliometrics, and the data were collected and analyzed using CiteSpace, VOSviewer, Microsoft Excel, and Endnote software. The publication language is "English," and the search strategy is TS = (exosome OR exosomes OR exosomal) AND TS = (respiratory OR lung). The search time is from the beginning of the WOS construction, and the deadline is July 11, 2022, at 22:00 hours. The literature types selected were dissertation, review paper, and online published paper. The analysis includes 2456 publications in 738 journals from 76 countries, 2716 institutions, and 14,568 authors. The field's annual publications have been rising, especially in recent years. China and the US lead research, and prominent universities, including Harvard Medical School, Shanghai Jiao Tong University, and Fudan University, are essential research institutes. Takahiro Ochiya, whose research focuses on exosomes and lung cancer, and Clotilde Théry, a pioneering exosome researcher, are the most cited authors in this field. The key terms include lung cancer, non-small cell lung cancer, mesenchymal stem cells, intercellular communication, exosomal miRNAs, and oncology. Cell biology, biochemistry & biotechnology, and oncology are related fields. The final summary of research hotspots is exosomes and lung cancer, mesenchymal stem cell-derived exosomes and lung inflammation, and miRNAs in exosomes as biomarkers for respiratory illnesses. The present research situation and relevant hotspots of the area were analyzed through bibliometric studies on exosomes in respiratory diseases. The research development in this field has a considerable upside, and the exosome's function in diagnosing, treating, monitoring, and prognosis of respiratory illnesses cannot be taken lightly. Moreover, we believe the research results will bring the gospel to many patients with clinical respiratory diseases shortly.
Collapse
Affiliation(s)
- Jinjie Fu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenjie Song
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Medical History and Literature Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory Innovation and Transformation, Tianjin, China
| | - Zheng Hao
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Medical History and Literature Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory Innovation and Transformation, Tianjin, China
| | - Mengzhen Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
46
|
Wu M, Jin Q, Xu X, Fan J, Chen W, Miao M, Gu R, Zhang S, Guo Y, Huang S, Zhang Y, Zhang A, Jia Z. TP53RK Drives the Progression of Chronic Kidney Disease by Phosphorylating Birc5. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301753. [PMID: 37382161 PMCID: PMC10477881 DOI: 10.1002/advs.202301753] [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: 03/17/2023] [Indexed: 06/30/2023]
Abstract
Renal fibrosis is a common characteristic of various chronic kidney diseases (CKDs) driving the loss of renal function. During this pathological process, persistent injury to renal tubular epithelial cells and activation of fibroblasts chiefly determine the extent of renal fibrosis. In this study, the role of tumor protein 53 regulating kinase (TP53RK) in the pathogenesis of renal fibrosis and its underlying mechanisms is investigated. TP53RK is upregulated in fibrotic human and animal kidneys with a positive correlation to kidney dysfunction and fibrotic markers. Interestingly, specific deletion of TP53RK either in renal tubule or in fibroblasts in mice can mitigate renal fibrosis in CKD models. Mechanistic investigations reveal that TP53RK phosphorylates baculoviral IAP repeat containing 5 (Birc5) and facilitates its nuclear translocation; enhanced Birc5 displays a profibrotic effect possibly via activating PI3K/Akt and MAPK pathways. Moreover, pharmacologically inhibiting TP53RK and Birc5 using fusidic acid (an FDA-approved antibiotic) and YM-155(currently in clinical phase 2 trials) respectively both ameliorate kidney fibrosis. These findings demonstrate that activated TP53RK/Birc5 signaling in renal tubular cells and fibroblasts alters cellular phenotypes and drives CKD progression. A genetic or pharmacological blockade of this axis serves as a potential strategy for treating CKDs.
Collapse
Affiliation(s)
- Mengqiu Wu
- Department of NephrologyNanjing Key Laboratory of PediatricsJiangsu Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210008P. R. China
| | - Qianqian Jin
- Department of NephrologyNanjing Key Laboratory of PediatricsJiangsu Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210008P. R. China
| | - Xinyue Xu
- School of MedicineSoutheast UniversityNanjing210009P. R. China
| | - Jiaojiao Fan
- School of MedicineSoutheast UniversityNanjing210009P. R. China
| | - Weiyi Chen
- Department of Emergency MedicineChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
| | - Mengqiu Miao
- Department of NephrologyNanjing Key Laboratory of PediatricsJiangsu Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210008P. R. China
| | - Ran Gu
- Department of NephrologyNanjing Key Laboratory of PediatricsJiangsu Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210008P. R. China
| | - Shengnan Zhang
- Department of NephrologyNanjing Key Laboratory of PediatricsJiangsu Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210008P. R. China
| | - Yan Guo
- Department of NephrologyNanjing Key Laboratory of PediatricsJiangsu Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210008P. R. China
| | - Songming Huang
- Department of NephrologyNanjing Key Laboratory of PediatricsJiangsu Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210008P. R. China
| | - Yue Zhang
- Department of NephrologyNanjing Key Laboratory of PediatricsJiangsu Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210008P. R. China
| | - Aihua Zhang
- Department of NephrologyNanjing Key Laboratory of PediatricsJiangsu Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210008P. R. China
| | - Zhanjun Jia
- Department of NephrologyNanjing Key Laboratory of PediatricsJiangsu Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210008P. R. China
| |
Collapse
|
47
|
van Zonneveld AJ, Zhao Q, Rotmans JI, Bijkerk R. Circulating non-coding RNAs in chronic kidney disease and its complications. Nat Rev Nephrol 2023; 19:573-586. [PMID: 37286733 DOI: 10.1038/s41581-023-00725-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/09/2023]
Abstract
Post-transcriptional regulation by non-coding RNAs (ncRNAs) can modulate the expression of genes involved in kidney physiology and disease. A large variety of ncRNA species exist, including microRNAs, long non-coding RNAs, piwi-interacting RNAs, small nucleolar RNAs, circular RNAs and yRNAs. Despite early assumptions that some of these species may exist as by-products of cell or tissue injury, a growing body of literature suggests that these ncRNAs are functional and participate in a variety of processes. Although they function intracellularly, ncRNAs are also present in the circulation, where they are carried by extracellular vesicles, ribonucleoprotein complexes or lipoprotein complexes such as HDL. These systemic, circulating ncRNAs are derived from specific cell types and can be directly transferred to a variety of cells, including endothelial cells of the vasculature and virtually any cell type in the kidney, thereby affecting the function of the host cell and/or its response to injury. Moreover, chronic kidney disease itself, as well as injury states associated with transplantation and allograft dysfunction, is associated with a shift in the distribution of circulating ncRNAs. These findings may provide opportunities for the identification of biomarkers with which to monitor disease progression and/or the development of therapeutic interventions.
Collapse
Affiliation(s)
- Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Qiao Zhao
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Joris I Rotmans
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Roel Bijkerk
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands.
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
48
|
Alli AA. Extracellular Vesicles: Investigating the Pathophysiology of Diabetes-Associated Hypertension and Diabetic Nephropathy. BIOLOGY 2023; 12:1138. [PMID: 37627022 PMCID: PMC10452642 DOI: 10.3390/biology12081138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023]
Abstract
Extracellular vesicles (EVs) include exosomes, microvesicles, and apoptotic bodies. EVs are released by all cell types and are found in biological fluids including plasma and urine. Urinary extracellular vesicles (uEVs) are a mixed population of EVs that comprise small EVs that are filtered and excreted, EVs secreted by tubular epithelial cells, and EVs released from the bladder, urethra, and prostate. The packaged cargo within uEVs includes bioactive molecules such as metabolites, lipids, proteins, mRNAs, and miRNAs. These molecules are involved in intercellular communication, elicit changes in intracellular signaling pathways, and play a role in the pathogenesis of various diseases including diabetes-associated hypertension and diabetic nephropathy. uEVs represent a rich source of biomarkers, prognosis markers, and can be loaded with small-molecule drugs as a vehicle for delivery.
Collapse
Affiliation(s)
- Abdel A. Alli
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA; ; Tel.: +1-352-273-7877
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
49
|
Jin Y, Li S, Yu Q, Chen T, Liu D. Application of stem cells in regeneration medicine. MedComm (Beijing) 2023; 4:e291. [PMID: 37337579 PMCID: PMC10276889 DOI: 10.1002/mco2.291] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 06/21/2023] Open
Abstract
Regeneration is a complex process affected by many elements independent or combined, including inflammation, proliferation, and tissue remodeling. Stem cells is a class of primitive cells with the potentiality of differentiation, regenerate with self-replication, multidirectional differentiation, and immunomodulatory functions. Stem cells and their cytokines not only inextricably linked to the regeneration of ectodermal and skin tissues, but also can be used for the treatment of a variety of chronic wounds. Stem cells can produce exosomes in a paracrine manner. Stem cell exosomes play an important role in tissue regeneration, repair, and accelerated wound healing, the biological properties of which are similar with stem cells, while stem cell exosomes are safer and more effective. Skin and bone tissues are critical organs in the body, which are essential for sustaining life activities. The weak repairing ability leads a pronounced impact on the quality of life of patients, which could be alleviated by stem cell exosomes treatment. However, there are obstacles that stem cells and stem cells exosomes trough skin for improved bioavailability. This paper summarizes the applications and mechanisms of stem cells and stem cells exosomes for skin and bone healing. We also propose new ways of utilizing stem cells and their exosomes through different nanoformulations, liposomes and nanoliposomes, polymer micelles, microspheres, hydrogels, and scaffold microneedles, to improve their use in tissue healing and regeneration.
Collapse
Affiliation(s)
- Ye Jin
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Shuangyang Li
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Qixuan Yu
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Tianli Chen
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| | - Da Liu
- School of PharmacyChangchun University of Chinese MedicineChangchunJilinChina
| |
Collapse
|
50
|
Yang S, Sun Y, Long M, Zhou X, Yuan M, Yang L, Luo W, Cheng Y, Zhang X, Jiang W, Chao J. Single-cell transcriptome sequencing-based analysis: probing the mechanisms of glycoprotein NMB regulation of epithelial cells involved in silicosis. Part Fibre Toxicol 2023; 20:29. [PMID: 37468937 DOI: 10.1186/s12989-023-00543-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/13/2023] [Indexed: 07/21/2023] Open
Abstract
Chronic exposure to silica can lead to silicosis, one of the most serious occupational lung diseases worldwide, for which there is a lack of effective therapeutic drugs and tools. Epithelial mesenchymal transition plays an important role in several diseases; however, data on the specific mechanisms in silicosis models are scarce. We elucidated the pathogenesis of pulmonary fibrosis via single-cell transcriptome sequencing and constructed an experimental silicosis mouse model to explore the specific molecular mechanisms affecting epithelial mesenchymal transition at the single-cell level. Notably, as silicosis progressed, glycoprotein non-metastatic melanoma protein B (GPNMB) exerted a sustained amplification effect on alveolar type II epithelial cells, inducing epithelial-to-mesenchymal transition by accelerating cell proliferation and migration and increasing mesenchymal markers, ultimately leading to persistent pulmonary pathological changes. GPNMB participates in the epithelial-mesenchymal transition in distant lung epithelial cells by releasing extracellular vesicles to accelerate silicosis. These vesicles are involved in abnormal changes in the composition of the extracellular matrix and collagen structure. Our results suggest that GPNMB is a potential target for fibrosis prevention.
Collapse
Affiliation(s)
- Shaoqi Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Rd, Nanjing, Jiangsu, 210009, China
- Key Laboratory of Environmental Medicine Engineering, School of Public Health, Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yuheng Sun
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Rd, Nanjing, Jiangsu, 210009, China
- Key Laboratory of Environmental Medicine Engineering, School of Public Health, Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Min Long
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, 29 Jiangjun Avenue, Nanjing, Jiangsu, 211106, China
| | - Xinbei Zhou
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Rd, Nanjing, Jiangsu, 210009, China
- Key Laboratory of Environmental Medicine Engineering, School of Public Health, Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Mengqin Yuan
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, 29 Jiangjun Avenue, Nanjing, Jiangsu, 211106, China
| | - Liliang Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Rd, Nanjing, Jiangsu, 210009, China
| | - Wei Luo
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Rd, Nanjing, Jiangsu, 210009, China
| | - Yusi Cheng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Rd, Nanjing, Jiangsu, 210009, China
| | - Xinxin Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Rd, Nanjing, Jiangsu, 210009, China
| | - Wei Jiang
- Department of Biomedical Engineering, College of Automation Engineering, Nanjing University of Aeronautics and Astronautics, 29 Jiangjun Avenue, Nanjing, Jiangsu, 211106, China.
| | - Jie Chao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Zhongda Hospital, Southeast University, 87 Dingjiaqiao Rd, Nanjing, Jiangsu, 210009, China.
- Key Laboratory of Environmental Medicine Engineering, School of Public Health, Ministry of Education, Southeast University, Nanjing, Jiangsu, 210009, China.
- School of Medicine, Xizang Minzu University, Xianyang, Shanxi, 712082, China.
| |
Collapse
|