1
|
Wang L, Yu M, Yang Y, Lv Y, Xie H, Chen J, Peng X, Peng Z, Zhou L, Wang Y, Huang Y, Chen F. Porous Photocrosslinkable Hydrogel Functionalized with USC Derived Small Extracellular Vesicles for Corpus Spongiosum Repair. Adv Healthc Mater 2024; 13:e2304387. [PMID: 39036844 DOI: 10.1002/adhm.202304387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/21/2024] [Indexed: 07/23/2024]
Abstract
Reconstruction of a full-thickness spongy urethra is difficult because a corpus spongiosum (CS) defect cannot be repaired using self-healing or substitution urethroplasty. Small extracellular vesicles (sEVs) secreted by urine-derived stem cells (USC-sEVs) strongly promote vascular regeneration. In this study, it is aimed to explore whether USC-sEVs promote the repair of CS defects. To prolong the in vivo effects of USC-sEVs, a void-forming photoinduced imine crosslinking hydrogel (vHG) is prepared and mixed with the USC-sEV suspension. vHG encapsulated with USC-sEVs (vHG-sEVs) is used to repair a CS defect with length of 1.5 cm and width of 0.8 cm. The results show that vHG-sEVs promote the regeneration and repair of CS defects. Histological analysis reveals abundant sinusoid-like vascular structures in the vHG-sEV group. Photoacoustic microscopy indicates that blood flow and microvascular structure of the defect area in the vHG-sEV group are similar to those in the normal CS group. This study confirms that the in situ-formed vHG-sEV patch appears to be a valid and promising strategy for repairing CS defects.
Collapse
Affiliation(s)
- Lin Wang
- Department of Urology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, 200233, China
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Mingming Yu
- Department of Urology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
- Department of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yunlong Yang
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yiqing Lv
- Department of Urology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hua Xie
- Department of Urology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiasheng Chen
- Department of Urology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xufeng Peng
- Department of Urology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhiwei Peng
- Department of Urology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lijun Zhou
- Department of Urology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Wang
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yichen Huang
- Department of Urology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Fang Chen
- Department of Urology, Shanghai Children's Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, 200233, China
| |
Collapse
|
2
|
Andersson KE, Williams K. Cellular regenerative therapy in stress urinary incontinence: new frontiers?-a narrative review. Transl Androl Urol 2024; 13:1709-1716. [PMID: 39280677 PMCID: PMC11399031 DOI: 10.21037/tau-22-682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 07/10/2023] [Indexed: 09/18/2024] Open
Abstract
Background and Objective Even if treatment with stem cells has been shown to be safe and effective in many patients with stress urinary incontinence (SUI), there is still room for improvement using other regenerative medicine alternatives. Since the beneficial effects of stem cells are probably mediated by secretion of factors rather than by the cells themselves there is a good rationale for further exploring the therapeutic effects of the secretome and/or its components. However, homing factors such as stromal derived growth factor 1 (SDF-1; CXCL12), stimulation of stem cell growth and stem cell mobilization in vivo using low intensity shock wave therapy (Li-ESWT) or regenerative electrical stimulation (RES), are also promising approaches. Methods A literature search was performed based on PubMed, Scopus and Google Scholar. The search criteria included original basic science articles, systematic reviews and randomized control trials. All studies were published between 2000 and 2023. Selected, peer-reviewed studies were further analyzed to identify those of relevance. Keywords searched included: "female stress incontinence", "homing factors", "CXCL12", "secretome", "low intensity shockwave therapy" and "regenerative electrical stimulation". The peer-reviewed publications on the key word subjects that contained a novel addition to the existing body of literature were included. Key Content and Findings There is evidence from studies on non-human primates (NHPs) with experimental urinary sphincter injury that CXCL12 can restore sphincter structure and function. Studies with homing factors in human patients with SUI are still to be performed. A large number of clinical studies on the use of secretome or secretome products from mesenchymal stem cells (MSCs) on indications other than human SUI are already available. However, controlled clinical trials on patients with SUI, have to the best of our knowledge, not yet been performed. Also, RES has not been studied in patients with SUI. In contrast, there is clinical evidence that Li-ESWT may improve female SUI. Conclusions Treatment with homing factors, MSC secretome/secretome components, Li-ESWT and RES are promising frontiers in the treatment of human SUI caused by sphincter damage.
Collapse
Affiliation(s)
- Karl-Erik Andersson
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
- Institute for Laboratory Medicine, Lund University, Lund, Sweden
| | - Koudy Williams
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| |
Collapse
|
3
|
Tan X, Li G, Li C, Kong C, Li H, Wu S. Animal models, treatment options, and biomaterials for female stress urinary incontinence. Front Bioeng Biotechnol 2024; 12:1414323. [PMID: 39267906 PMCID: PMC11390547 DOI: 10.3389/fbioe.2024.1414323] [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: 04/08/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
In the quest to tackle stress urinary incontinence (SUI), the synthesis of cutting-edge biomaterials and regenerative materials has emerged as a promising frontier. Briefly, animal models like vaginal distension and bilateral ovariectomy serve as crucial platforms for unraveling the intricacies of SUI, facilitating the evaluation of innovative treatments. The spotlight, however, shines on the development and application of novel biomaterials-ranging from urethral bulking agents to nano-gel composites-which aim to bolster urethral support and foster tissue regeneration. Furthermore, the exploration of stem cell therapies, particularly those derived from adipose tissues and urine, heralds a new era of regenerative medicine, offering potential for significant improvements in urinary function. This review encapsulates the progress in biomaterials and regenerative strategies, highlighting their pivotal role in advancing the treatment of SUI, thereby opening new avenues for effective and minimally invasive solutions.
Collapse
Affiliation(s)
- Xiyang Tan
- Shenzhen Hospital, Shanghai University of Traditional Chinese Medicine, Shenzhen, China
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Department of Urology, The Affiliated South China Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Guangzhi Li
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Chenchen Li
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Chenfan Kong
- Shenzhen Hospital, Shanghai University of Traditional Chinese Medicine, Shenzhen, China
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Department of Urology, The Affiliated South China Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Huizhen Li
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Song Wu
- Shenzhen Hospital, Shanghai University of Traditional Chinese Medicine, Shenzhen, China
- Department of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
- Department of Urology, The Affiliated South China Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| |
Collapse
|
4
|
Alwan A, Khalil F, Bowlby J, Peko G, Estrada EV, Singh S, Deep G, Zhang Y, Farney AC, Opara EC. Effect of controlled release of HGF on extracellular vesicle secretion by urine-derived stem cells. Front Bioeng Biotechnol 2024; 12:1436296. [PMID: 39234273 PMCID: PMC11371732 DOI: 10.3389/fbioe.2024.1436296] [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: 05/21/2024] [Accepted: 08/01/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction The hepatic growth factor (HGF) stimulates DNA synthesis and cell proliferation and plays a role in tissue protection and regeneration. In this study, we have examined the effect of incubation of HGF with urine-derived stem cells (USCs) on the secretion of small extracellular vesicles (sEV) by the cells. Materials and Methods HGF in the incubation medium was either a bolus administration or a controlled release of an equivalent amount from microbeads within the size range of 50-200 µm made with ultrapurified low-viscosity high-guluronic acid (UP-LVG) alginate. USCs were incubated with or without HGF for 3 days or 7 days before removal of the incubation media, followed by harvesting sEV by the precipitation method. The protein content of isolated sEV was measured by bicinchoninic acid assay (BCA) for these three groups: control (no HGF beads), bolus HGF, and HGF beads. We also performed nanoparticle tracking analysis (NTA), Western blot assay, and ELISA for the HGF content of samples. Results We found a significantly higher concentration of proteins in the HGF microbead group (control release group) compared to the bolus group and the control group after 7 days (p < 0.0017). The NTA data aligned with the BCA; they showed a significantly higher concentration of particles within the size range of sEV (<200 nm) in the group treated with HGF beads compared to the two other groups on day 7 (p < 0.0001). Conclusion We found that administration of HGF to USCs by controlled release of the growth factor significantly enhances the levels of sEV secretion during 7 days of incubation.
Collapse
Affiliation(s)
- Abdelrahman Alwan
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Fatma Khalil
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
- Department of Histology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Joshua Bowlby
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Gabrielle Peko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Exel Valle Estrada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Sangeeta Singh
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Gagan Deep
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Alan C Farney
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Emmanuel C Opara
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
5
|
Yu P, Bosholm CC, Zhu H, Duan Z, Atala A, Zhang Y. Beyond waste: understanding urine's potential in precision medicine. Trends Biotechnol 2024; 42:953-969. [PMID: 38369434 PMCID: PMC11741143 DOI: 10.1016/j.tibtech.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/20/2024]
Abstract
Urine-derived stem cells (USCs) are a promising source of stem cells for cell therapy, renal toxicity drug testing, and renal disease biomarker discovery. Patients' own USCs can be used for precision medicine. In this review we first describe the isolation and characterization of USCs. We then discuss preclinical studies investigating the use of USCs in cell therapy, exploring the utility of USCs and USC-derived induced pluripotent stem cells (u-iPSCs) in drug toxicity testing, and investigating the use of USCs as biomarkers for renal disease diagnosis. Finally, we discuss the challenges of using USCs in these applications and provide insights into future research directions. USCs are a promising tool for advancing renal therapy, drug testing, and biomarker discovery. Further research is needed to explore their potential.
Collapse
Affiliation(s)
- Pengfei Yu
- The Fourth Department of Liver Disease, Beijing YouAn Hospital, Capital Medical University, Beijing, China; Wake Forest Institute for Regeneration Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Carol Christine Bosholm
- Wake Forest Institute for Regeneration Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Hainan Zhu
- Wake Forest Institute for Regeneration Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Zhongping Duan
- The Fourth Department of Liver Disease, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Anthony Atala
- Wake Forest Institute for Regeneration Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regeneration Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
6
|
Khanabdali R, Mandrekar M, Grygiel R, Vo PA, Palma C, Nikseresht S, Barton S, Shojaee M, Bhuiyan S, Asari K, Belzer S, Ansari K, Coward JI, Perrin L, Hooper J, Guanzon D, Lai A, Salomon C, Kershner K, Newton C, Horejsh D, Rice G. High-throughput surface epitope immunoaffinity isolation of extracellular vesicles and downstream analysis. Biol Methods Protoc 2024; 9:bpae032. [PMID: 39070184 PMCID: PMC11272960 DOI: 10.1093/biomethods/bpae032] [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: 04/03/2024] [Revised: 04/09/2024] [Accepted: 05/16/2024] [Indexed: 07/30/2024] Open
Abstract
Extracellular vesicles (EVs), including exosomes, have significant potential for diagnostic and therapeutic applications. The lack of standardized methods for efficient and high-throughput isolation and analysis of EVs, however, has limited their widespread use in clinical practice. Surface epitope immunoaffinity (SEI) isolation utilizes affinity ligands, including antibodies, aptamers, or lectins, that target specific surface proteins present on EVs. Paramagnetic bead-SEI isolation represents a fit-for-purpose solution for the reproducible, high-throughput isolation of EVs from biofluids and downstream analysis of RNA, protein, and lipid biomarkers that is compatible with clinical laboratory workflows. This study evaluates a new SEI isolation method for enriching subpopulations of EVs. EVs were isolated from human plasma using a bead-based SEI method designed for on-bead and downstream analysis of EV-associated RNA and protein biomarkers. Western blot analysis confirmed the presence of EV markers in the captured nanoparticles. Mass spectrometry analysis of the SEI lysate identified over 1500 proteins, with the top 100 including known EV-associated proteins. microRNA (miRNA) sequencing followed by RT-qPCR analysis identified EV-associated miRNA transcripts. Using SEI, EVs were isolated using automated high-throughput particle moving instruments, demonstrating equal or higher protein and miRNA yield and recovery compared to manual processing. SEI is a rapid, efficient, and high-throughput method for isolating enriched populations of EVs; effectively reducing contamination and enabling the isolation of a specific subpopulation of EVs. In this study, high-throughput EV isolation and RNA extraction have been successfully implemented. This technology holds great promise for advancing the field of EV research and facilitating their application for biomarker discovery and clinical research.
Collapse
Affiliation(s)
| | | | - Rick Grygiel
- Promega Corporation, Madison, WI 53711, United States
| | - Phuoc-An Vo
- Promega Corporation, Madison, WI 53711, United States
| | | | | | | | | | | | | | | | | | - Jermaine I Coward
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
- ICON Cancer Care, South Brisbane, QLD 4101, Australia
| | - Lewis Perrin
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - John Hooper
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Dominic Guanzon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Andrew Lai
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | | | | | | | - Gregory Rice
- INOVIQ Ltd., Notting Hill, VIC 3168, Australia
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women’s Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
7
|
Zou Y, Li S, Chen W, Xu J. Urine-derived stem cell therapy for diabetes mellitus and its complications: progress and challenges. Endocrine 2024; 83:270-284. [PMID: 37801228 DOI: 10.1007/s12020-023-03552-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/24/2023] [Indexed: 10/07/2023]
Abstract
Diabetes mellitus (DM) is a chronic and relentlessly progressive metabolic disease characterized by a relative or absolute deficiency of insulin in the body, leading to increased production of advanced glycosylation end products that further enhance oxidative and nitrosative stresses, often leading to multiple macrovascular (cardiovascular disease) and microvascular (e.g., diabetic nephropathy, diabetic retinopathy, and neuropathy) complications, representing the ninth leading cause of death worldwide. Existing medical treatments do not provide a complete cure for DM; thus, stem cell transplantation therapy has become the focus of research on DM and its complications. Urine-derived stem cells (USCs), which are isolated from fresh urine and have biological properties similar to those of mesenchymal stem cells (MSCs), were demonstrated to exert antiapoptotic, antifibrotic, anti-inflammatory, and proangiogenic effects through direct differentiation or paracrine mechanisms and potentially treat patients with DM. USCs also have the advantages of simple noninvasive sample collection procedures, minimal ethical issues, low cost, and easy cell isolation methods and thus have received more attention in regenerative therapies in recent years. This review outlines the biological properties of USCs and the research progress and current limitations of their role in DM and related complications. In summary, USCs have shown good versatility in treating hyperglycemia-impaired target organs in preclinical models, and many challenges remain in translating USC therapies to the clinic.
Collapse
Affiliation(s)
- Yun Zou
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shanshan Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wen Chen
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jixiong Xu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
8
|
Knoll J, Amend B, Harland N, Isser S, Bézière N, Kraushaar U, Stenzl A, Aicher WK. Cell Therapy by Mesenchymal Stromal Cells Versus Myoblasts in a Pig Model of Urinary Incontinence. Tissue Eng Part A 2024; 30:14-30. [PMID: 37933911 DOI: 10.1089/ten.tea.2023.0103] [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: 11/08/2023] Open
Abstract
The leading cause of stress urinary incontinence (SUI) in women is the urethral sphincter muscle deficiency caused by mechanical stress during pregnancy and vaginal delivery. In men, prostate cancer surgery and injury of local nerves and muscles are associated with incontinence. Current treatment often fails to satisfy the patient's needs. Cell therapy may improve the situation. We therefore investigated the regeneration potential of cells in ameliorating sphincter muscle deficiency and UI in a large animal model. Urethral sphincter deficiency was induced surgically in gilts by electrocautery and balloon dilatation. Adipose tissue-derived stromal cells (ADSCs) and myoblasts from Musculus semitendinosus were isolated from male littermates, expanded, characterized in depth for expression of marker genes and in vitro differentiation, and labeled. The cells were injected into the deficient sphincter complex of the incontinent female littermates. Incontinent gilts receiving no cell therapy served as controls. Sphincter deficiency and functional regeneration were recorded by monitoring the urethral wall pressure during follow-up by two independent methods. Cells injected were detected in vivo during follow-up by transurethral fluorimetry, ex vivo by fluorescence imaging, and in cryosections of tissues targeted by immunofluorescence and by polymerase chain reaction of the sex-determining region Y (SRY) gene. Partial spontaneous regeneration of sphincter muscle function was recorded in control gilts, but the sphincter function remained significantly below levels measured before induction of incontinence (67.03% ± 14.00%, n = 6, p < 0.05). Injection of myoblasts yielded an improved sphincter regeneration within 5 weeks of follow-up but did not reach significance compared to control gilts (81.54% ± 25.40%, n = 5). A significant and full recovery of the urethral sphincter function was observed upon injection of ADSCs within 5 weeks of follow-up (100.4% ± 23.13%, n = 6, p < 0.05). Injection of stromal cells provoked slightly stronger infiltration of CD45pos leukocytes compared to myoblasts injections and controls. The data of this exploratory study indicate that ADSCs inherit a significant potential to regenerate the function of the urethral sphincter muscle.
Collapse
Affiliation(s)
- Jasmin Knoll
- Department of Urology at UKT, Center for Medical Research, Eberhard-Karls-University, Tuebingen, Germany
| | - Bastian Amend
- Department of Urology, University of Tuebingen Hospital, Tuebingen, Germany
| | - Niklas Harland
- Department of Urology, University of Tuebingen Hospital, Tuebingen, Germany
| | - Simon Isser
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University of Tübingen, Germany
| | - Nicolas Bézière
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University of Tübingen, Germany
- Cluster of Excellence CMFI (EXC 2124) "Controlling Microbes to Fight Infections," Eberhard Karls University Tübingen, Germany
| | - Udo Kraushaar
- Naturwissenschaftlich-Medizinisches Institut, Reutlingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tuebingen Hospital, Tuebingen, Germany
| | - Wilhelm K Aicher
- Department of Urology at UKT, Center for Medical Research, Eberhard-Karls-University, Tuebingen, Germany
| |
Collapse
|
9
|
Huang RL, Li Q, Ma JX, Atala A, Zhang Y. Body fluid-derived stem cells - an untapped stem cell source in genitourinary regeneration. Nat Rev Urol 2023; 20:739-761. [PMID: 37414959 PMCID: PMC11639537 DOI: 10.1038/s41585-023-00787-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 07/08/2023]
Abstract
Somatic stem cells have been obtained from solid organs and tissues, including the bone marrow, placenta, corneal stroma, periosteum, adipose tissue, dental pulp and skeletal muscle. These solid tissue-derived stem cells are often used for tissue repair, disease modelling and new drug development. In the past two decades, stem cells have also been identified in various body fluids, including urine, peripheral blood, umbilical cord blood, amniotic fluid, synovial fluid, breastmilk and menstrual blood. These body fluid-derived stem cells (BFSCs) have stemness properties comparable to those of other adult stem cells and, similarly to tissue-derived stem cells, show cell surface markers, multi-differentiation potential and immunomodulatory effects. However, BFSCs are more easily accessible through non-invasive or minimally invasive approaches than solid tissue-derived stem cells and can be isolated without enzymatic tissue digestion. Additionally, BFSCs have shown good versatility in repairing genitourinary abnormalities in preclinical models through direct differentiation or paracrine mechanisms such as pro-angiogenic, anti-apoptotic, antifibrotic, anti-oxidant and anti-inflammatory effects. However, optimization of protocols is needed to improve the efficacy and safety of BFSC therapy before therapeutic translation.
Collapse
Affiliation(s)
- Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Anthony Atala
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
10
|
Xu L, Sima Y, Xiao C, Chen Y. Exosomes derived from mesenchymal stromal cells: a promising treatment for pelvic floor dysfunction. Hum Cell 2023; 36:937-949. [PMID: 36940057 DOI: 10.1007/s13577-023-00887-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/25/2023] [Indexed: 03/21/2023]
Abstract
Pelvic floor dysfunction (PFDs), which include pelvic organ prolapse (POP), stress urinary incontinence (SUI) and anal incontinence (AI), are common degenerative diseases in women that have dramatic effects on quality of life. The pathology of PFDs is based on impaired pelvic connective tissue supportive strength due to an imbalance in extracellular matrix (ECM) metabolism, the loss of a variety of cell types, such as fibroblasts, muscle cells, peripheral nerve cells, and oxidative stress and inflammation in the pelvic environment. Fortunately, exosomes, which are one of the major secretions of mesenchymal stromal cells (MSCs), are involved in intercellular communication and the modulation of molecular activities in recipient cells via their contents, which are bioactive proteins and genetic factors such as mRNAs and miRNAs. These components modify fibroblast activation and secretion, facilitate ECM modelling, and promote cell proliferation to enhance pelvic tissue regeneration. In this review, we focus on the molecular mechanisms and future directions of exosomes derived from MSCs that are of great value in the treatment of PFD.
Collapse
Affiliation(s)
- Leimei Xu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 128 ShenYang Road, Shanghai, 200011, People's Republic of China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Yizhen Sima
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 128 ShenYang Road, Shanghai, 200011, People's Republic of China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Chengzhen Xiao
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 128 ShenYang Road, Shanghai, 200011, People's Republic of China
| | - Yisong Chen
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, 128 ShenYang Road, Shanghai, 200011, People's Republic of China. .,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China.
| |
Collapse
|
11
|
Shan S, Li Q, Criswell T, Atala A, Zhang Y. Stem cell therapy combined with controlled release of growth factors for the treatment of sphincter dysfunction. Cell Biosci 2023; 13:56. [PMID: 36927578 PMCID: PMC10018873 DOI: 10.1186/s13578-023-01009-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Sphincter dysfunction often occurs at the end of tubule organs such as the urethra, anus, or gastroesophageal sphincters. It is the primary consequence of neuromuscular impairment caused by trauma, inflammation, and aging. Despite intensive efforts to recover sphincter function, pharmacological treatments have not achieved significant improvement. Cell- or growth factor-based therapy is a promising approach for neuromuscular regeneration and the recovery of sphincter function. However, a decrease in cell retention and viability, or the short half-life and rapid degradation of growth factors after implantation, remain obstacles to the translation of these therapies to the clinic. Natural biomaterials provide unique tools for controlled growth factor delivery, which leads to better outcomes for sphincter function recovery in vivo when stem cells and growth factors are co-administrated, in comparison to the delivery of single therapies. In this review, we discuss the role of stem cells combined with the controlled release of growth factors, the methods used for delivery, their potential therapeutic role in neuromuscular repair, and the outcomes of preclinical studies using combination therapy, with the hope of providing new therapeutic strategies to treat incontinence or sphincter dysfunction of the urethra, anus, or gastroesophageal tissues, respectively.
Collapse
Affiliation(s)
- Shengzhou Shan
- 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.
| | - Tracy Criswell
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
12
|
Wang XX, Zhang L, Lu Y. Advances in the molecular pathogenesis and cell therapy of stress urinary incontinence. Front Cell Dev Biol 2023; 11:1090386. [PMID: 36846586 PMCID: PMC9944745 DOI: 10.3389/fcell.2023.1090386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Stress urinary incontinence (SUI) is very common in women. It affects patients' mental and physical health, and imposed huge socioeconomic pressure. The therapeutic effect of conservative treatment is limited, and depends heavily on patient persistence and compliance. Surgical treatment often brings procedure-related adverse complications and higher costs for patients. Therefore, it is necessary to better understand the potential molecular mechanisms underlying stress urinary incontinence and develop new treatment methods. Although some progress has been made in the basic research in recent years, the specific molecular pathogenic mechanisms of SUI are still unclear. Here, we reviewed the published studies on the molecular mechanisms associated with nerves, urethral muscles, periurethral connective tissue and hormones in the pathogenesis of SUI. In addition, we provide an update on the recent progresses in research on the use of cell therapy for treating SUI, including research on stem cells therapy, exosome differentiation and gene regulation.
Collapse
Affiliation(s)
- Xiao-xiao Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Lei Zhang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | | |
Collapse
|
13
|
Abstract
Skeletal muscle plays a paramount role in physical activity, metabolism, and energy balance, while its homeostasis is being challenged by multiple unfavorable factors such as injury, aging, or obesity. Exosomes, a subset of extracellular vesicles, are now recognized as essential mediators of intercellular communication, holding great clinical potential in the treatment of skeletal muscle diseases. Herein, we outline the recent research progress in exosomal isolation, characterization, and mechanism of action, and emphatically discuss current advances in exosomes derived from multiple organs and tissues, and engineered exosomes regarding the regulation of physiological and pathological development of skeletal muscle. These remarkable advances expand our understanding of myogenesis and muscle diseases. Meanwhile, the engineered exosome, as an endogenous nanocarrier combined with advanced design methodologies of biomolecules, will help to open up innovative therapeutic perspectives for the treatment of muscle diseases.
Collapse
|
14
|
Liu M, Wang Y, Gao G, Zhao WX, Fu Q. Stem Cell Application for Stress Urinary Incontinence: From Bench to Bedside. Curr Stem Cell Res Ther 2023; 18:17-26. [PMID: 35249506 DOI: 10.2174/1574888x17666220304213057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/29/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022]
Abstract
Stress urinary incontinence (SUI) is a common urinary system disease worldwide. Nowadays, medical therapy and surgery can control the symptoms and improve the life quality of patients. However, they might also bring about complications as the standard therapy fails to address the underlying problem of urethral sphincter dysfunction. Recent advances in cell technology have aroused interest in the use of autologous stem cell therapy to restore the ability of urinary control. The present study reviewed several types of stem cells for the treatment of SUI in the experimental and clinical stages.
Collapse
Affiliation(s)
- Meng Liu
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Ying Wang
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Guo Gao
- Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei-Xin Zhao
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Qiang Fu
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai 200233, China
| |
Collapse
|
15
|
Najafi-Ghalehlou N, Feizkhah A, Mobayen M, Pourmohammadi-Bejarpasi Z, Shekarchi S, Roushandeh AM, Roudkenar MH. Plumping up a Cushion of Human Biowaste in Regenerative Medicine: Novel Insights into a State-of-the-Art Reserve Arsenal. Stem Cell Rev Rep 2022; 18:2709-2739. [PMID: 35505177 PMCID: PMC9064122 DOI: 10.1007/s12015-022-10383-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 12/03/2022]
Abstract
Major breakthroughs and disruptive methods in disease treatment today owe their thanks to our inch by inch developing conception of the infinitive aspects of medicine since the very beginning, among which, the role of the regenerative medicine can on no account be denied, a branch of medicine dedicated to either repairing or replacing the injured or diseased cells, organs, and tissues. A novel means to accomplish such a quest is what is being called "medical biowaste", a large assortment of biological samples produced during a surgery session or as a result of physiological conditions and biological activities. The current paper accentuating several of a number of promising sources of biowaste together with their plausible applications in routine clinical practices and the confronting challenges aims at inspiring research on the existing gap between clinical and basic science to further extend our knowledge and understanding concerning the potential applications of medical biowaste.
Collapse
Affiliation(s)
- Nima Najafi-Ghalehlou
- Department of Medical Laboratory Sciences, Faculty of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Feizkhah
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammadreza Mobayen
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Pourmohammadi-Bejarpasi
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Shima Shekarchi
- Anatomical Sciences Department, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amaneh Mohammadi Roushandeh
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| | - Mehryar Habibi Roudkenar
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
16
|
Zhang X, Chen JL, Xing F, Duan X. Three-dimensional printed polylactic acid and hydroxyapatite composite scaffold with urine-derived stem cells as a treatment for bone defects. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 33:71. [PMID: 36190568 PMCID: PMC9529701 DOI: 10.1007/s10856-022-06686-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 08/22/2022] [Indexed: 06/16/2023]
Abstract
Bone defects still pose various challenges in osteology. As one treatment method for bone defects, tissue engineering requires biomaterials with good biocompatibility and stem cells with good differentiation. This study aimed to fabricate a 3D-printed polylactic acid and hydroxyapatite (PLA/HA) composite scaffold with urine-derived stem cells (USCs) to study its therapeutic effect in a rat model of skull defects. USCs were isolated and extracted from the urine of healthy adult males and inoculated onto PLA/HA and PLA scaffolds fabricated by 3D printing technology. A total of 36 skull defect models in eighteen Sprague-Dawley rats were randomly divided into a control group (no treatment of the defects), PLA group (treated with PLA scaffolds with USCs), and PLA/HA group (treated with PLA/HA scaffolds with USCs). The therapeutic efficacy was evaluated by real-time PCR, microcomputed tomography (micro-CT), and immunohistochemistry at 4, 8, and 12 weeks. We found that the PLA/HA scaffold loaded with USCs effectively promoted new bone regeneration in the defect area. CT images showed that in the PLA/HA group, the defect area was almost entirely covered by newly formed bone (coverage of 96.7 ± 1.6%), and the coverage was greater than that in the PLA group (coverage of 74.6 ± 1.9%) at 12 weeks. Histology and immunohistochemical staining showed the highest new bone formation on the PLA/HA scaffolds containing USCs in the defect site at 12 weeks. These findings demonstrate the broad application prospects of PLA/HA scaffolds with USCs in bone tissue engineering. Graphical abstract.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Jia-Lei Chen
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Fei Xing
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China
| | - Xin Duan
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041, Sichuan, China.
- Department of Orthopedics, Ganzi Tibetan Autonomous Prefecture People's Hospital, Ganzi Prefecture, 626700, Sichuan, China.
| |
Collapse
|
17
|
Urine-Derived Stem Cells for Epithelial Tissues Reconstruction and Wound Healing. Pharmaceutics 2022; 14:pharmaceutics14081669. [PMID: 36015295 PMCID: PMC9415563 DOI: 10.3390/pharmaceutics14081669] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/09/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Epithelial tissue injury can occur on any surface site of the body, particularly in the skin or urethral mucosa tissue, due to trauma, infection, inflammation, and toxic compounds. Both internal and external body epithelial tissue injuries can significantly affect patients’ quality of life, increase healthcare spending, and increase the global economic burden. Transplantation of epithelial tissue grafts is an effective treatment strategy in clinical settings. Autologous bio-engineered epithelia are common clinical skin substitutes that have the specific advantages of avoiding tissue rejection, obviating ethical concerns, reducing the risk of infection, and decreasing scarring compared to donor grafts. However, epithelial cells are often obtained from the individual’s skin and mucosa through invasive methods, which cause further injury or damage. Urine-derived stem cells (USC) of kidney origin, obtained via non-invasive acquisition, possess high stemness properties, self-renewal ability, trophic effects, multipotent differentiation potential, and immunomodulatory ability. These cells show versatile potential for tissue regeneration, with extensive evidence supporting their use in the repair of epidermal and urothelial injuries. We discuss the collection, isolation, culture, characterization, and differentiation of USC. We also discuss the use of USC for cellular therapies as well as the administration of USC-derived paracrine factors for epidermal and urothelial tissue repair. Specifically, we will discuss 3D constructions involving multiple types of USC-loaded hydrogels and USC-seeded scaffolds for use in cosmetic production testing, drug development, and disease modeling. In conclusion, urine-derived stem cells are a readily accessible autologous stem cell source well-suited for developing personalized medical treatments in epithelial tissue regeneration and drug testing.
Collapse
|
18
|
Fang J, Peng T, Liu J, Liu H, Liu T, Zhang Z, Zhao C, Li Y, Wang Q, Chen H, Li T, Huang S, Pu X. Muscle-derived Stem Cells Combined With Nerve Growth Factor Transplantation in the Treatment of Stress Urinary Incontinence. Urology 2022; 166:126-132. [PMID: 35490902 DOI: 10.1016/j.urology.2022.04.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To investigate the efficacy of muscle-derived stem cells (MDSCs) combined with nerve growth factor (NGF) in the treatment of stress urinary incontinence (SUI) METHODS: MDSCs were isolated and extracted from 90 SD rats, and the stem cell characteristics of the cells were identified using flow cytometry. NGF overexpression (oe-NGF) plasmid was coated with adenovirus and qRT-PCR was applied to verify adenovirus transfection efficiency. The rat models of SUI were constructed and randomly divided into 5 groups: control group, phosphate buffer (PBS) group, MDSCs + oe-NGF group, MDSCs + vector group, and MDSCs group. After 8 weeks of feeding, the leakage point pressure (LPP) rats, and Masson staining of rat urethral sections were detected. The expression of NGF and vascular endothelial growth factor (VEGF) was detected by western blot and IHC staining. RESULTS Compared with the control group, the LPP and the ratio of muscle fibers/collagen fibers were significantly increased in the MDSCs treated groups, with the highest increase in the MDSCs + oe-NGF group. Western blot and IHC results showed that the expression of NGF and VEGF in the urethral tissues in the MDSCs treated groups were significantly up-regulated comparing with the control group, with the highest increase in the MDSCs + oe-NGF group. CONCLUSION MDSCs alone can relieve SUI, while MDSCs combined with NGF is more effective, which may be related to the up-regulating of VEGF.
Collapse
Affiliation(s)
- Jianxiong Fang
- Department of Urology, Jiangmen Central Hospital, Jiangmen, PR China; Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Tianming Peng
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Jiumin Liu
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Haosheng Liu
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Tianqi Liu
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China; Shantou University Medical College, Shantou, PR China
| | - Zhenhui Zhang
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Chao Zhao
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Yong Li
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Qianqian Wang
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Hanzhong Chen
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Teng Li
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Shang Huang
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Xiaoyong Pu
- Department of Urology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
19
|
Ye YC, Chang ZH, Wang P, Wang YW, Liang J, Chen C, Wang JJ, Sun HT, Wang Y, Li XH. Infarct-preconditioning exosomes of umbilical cord mesenchymal stem cells promoted vascular remodeling and neurological recovery after stroke in rats. Stem Cell Res Ther 2022; 13:378. [PMID: 35902882 PMCID: PMC9330700 DOI: 10.1186/s13287-022-03083-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/21/2022] [Indexed: 12/30/2022] Open
Abstract
Background Stroke is the leading cause of disability worldwide, resulting in severe damage to the central nervous system and disrupting neurological functions. There is no effective therapy for promoting neurological recovery. Growing evidence suggests that the composition of exosomes from different microenvironments may benefit stroke. Therefore, it is reasonable to assume that exosomes secreted in response to infarction microenvironment could have further therapeutic effects. Methods In our study, cerebral infarct tissue extracts were used to pretreat umbilical cord mesenchymal stem cells (UCMSC). Infarct-preconditioned exosomes were injected into rats via tail vein after middle cerebral artery occlusion (MCAO). The effect of infarct-preconditioned exosomes on the neurological recovery of rats was examined using Tunel assay, 2,3,5-triphenyltetrazolium chloride (TTC) assay, magnetic resonance imaging (MRI) analyses, modified Neurological Severity Score (mNSS), Morris water maze (MWM), and vascular remodeling analysis. Mi-RNA sequencing and functional enrichment analysis were used to validate the signal pathway involved in the effect of infarct-preconditioned exosomes. Human umbilical vein endothelial cells (HUVECs) were co-cultured with the isolated exosomes. Cell Counting Kit-8 (CCK-8) assay, scratch healing, and Western blot analysis were used to detect the biological behavior of HUVECs. Results The results showed that compared with normal exosomes, infarct-preconditioned exosomes further promoted vascular remodeling and recovery of neurological function after stroke. The function of upregulated miRNAs and their target genes which is beneficial to vascular smooth muscle cells verified the importance of vascular remodeling in improving stroke. Better resistance to oxygen–glucose deprivation/reoxygenation (OGD/R), reduced apoptosis, and enhanced migration were observed in infarct-preconditioned exosomes-treated umbilical vein endothelial cells. Conclusions Our results demonstrated that infarct-preconditioned exosomes promoted neurological recovery after stroke by enhancing vascular endothelial remodeling, suggested that infarct-preconditioned exosomes could be a novel way to alleviate brain damage following a stroke. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03083-9.
Collapse
Affiliation(s)
- Yi-Chao Ye
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Zhe-Han Chang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Peng Wang
- Health Management Department, Tianjin Hospital, Tianjin University, Tianjin, 300299, China
| | - You-Wei Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jun Liang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China.,Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Jing-Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Hong-Tao Sun
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Yi Wang
- Neurology Department, Tianjin Hospital, Tianjin University, Tianjin, 300299, China.
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
20
|
Sadeghmousavi S, Soltani Khaboushan A, Jafarnezhad-Ansariha F, Nejad-Gashti R, Farsi M, Esmaeil-Pour R, Alijani M, Majidi Zolbin M, Niknejad H, Kajbafzadeh AM. The role of spinal cord tractography in detecting lesions following selective bladder afferent and efferent fibers: A novel method for induction of neurogenic lower urinary tract dysfunction in rabbit. Neurourol Urodyn 2022; 41:1539-1552. [PMID: 35842827 DOI: 10.1002/nau.25009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/07/2022] [Accepted: 06/19/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Neurogenic lower urinary tract dysfunction (NLUTD), a challenging disorder, is defined by lack of bladder control due to the abnormalities in neural pathways and can be classified based on the location of lesions within the nervous system, thus investigating the neural pathways can help us to know the site of the lesion and specify the class of the NLUTD. Diffusion Tensor Imaging (DTI) tractography, a noninvasive advanced imaging method, is capable of detecting central nervous system pathologies, even if routine magnetic resonance imaging shows no abnormality. Accordingly, tractography is an ideal technique to evaluate patients with NLUTD and visualize the pathology site within the spine. This study aimed to introduce a novel method of spinal cord injury (SCI) to establish NLUTD in the rabbit and to investigate the potential of tractography in tracing neural tracts of the spinal cord in an induced NLUTD animal model. MATERIALS AND METHODS An animal model of NLUTD was induced through cauterization of the spinal cord at the level T12-L1 in 12 rabbits. Then rabbits were assessed via DTI, urodynamic studies (UDS), voiding cystourethrogram (VCUG), and pathology assessments using antineurofilament 200 (NF200) antibody, anti-S100, anti-Smooth Muscle Actin, anti-Myogenin, and anti-MyoD1. RESULTS The tractography visualized lesions within spinal cord fibers. DTI parameters including fractional anisotropy (FA) value and tract density were significantly decreased (FA: p-value = 0.01, Tract density: p-value = 0.05) after injury. The mean diffusivity (MD) was insignificantly increased compared to before the injury. Also, the results of UDS and pathology assessments corroborated that applying SCI and the establishment of the NLUTD model was completely successful. CONCLUSION In the present study, we investigated the auxiliary role of tractography in detecting the spinal cord lesions in the novel established rabbit model of NLUTD. The introduced method of NLUTD induction was without the leg's neurological deficit, easily applicable, low-cost, and was accompanied by minimal surgical preparation and a satisfactory survival rate in comparison with other SCI animal models.
Collapse
Affiliation(s)
- Shaghayegh Sadeghmousavi
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatrics' Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran.,School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Soltani Khaboushan
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatrics' Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Fahimeh Jafarnezhad-Ansariha
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatrics' Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Nejad-Gashti
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatrics' Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Farsi
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatrics' Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Esmaeil-Pour
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatrics' Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Alijani
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatrics' Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatrics' Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Pediatrics' Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran.,Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Childern's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Zhang S, Li J, Li C, He J, Ling F, Liu G. Isolation and identification of a mesenchymal stem/stromal cell-like population from pediatric urethral tissue. In Vitro Cell Dev Biol Anim 2022; 58:503-511. [PMID: 35817989 DOI: 10.1007/s11626-022-00697-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/23/2022] [Indexed: 11/25/2022]
Abstract
Mesenchymal stem cells (MSCs) are important seed cells for cell therapy and tissue engineering because of their multidirectional differentiation potential, high proliferative capacity, low immunogenicity, and immunomodulatory ability. In this study, we successfully isolated and cultured a population of mesenchymal stem-like cells from pediatric urethra (PU-MSLCs). The cells had a spindle-shaped fibroblast-like morphology, similar to MSCs derived from other tissues. The PU-MSLCs highly expressed MSC surface markers CD29, CD73, CD90, and CD105 but were negative for leukocyte common antigen CD45, and MHC class II-encoded molecule HLA-DR. After in vitro induction, the PU-MSLCs had the potential to differentiate into adipocytes, osteocytes, and chondrocytes. The PU-MSLCs maintained a normal karyotype and showed no tumorigenicity during long-term cultivation. We thus demonstrated that the mesenchymal stem/stromal cell-like population obtained from pediatric urethra tissue is capable of self-renewal and multidirectional differentiation, has promising application prospects for cell therapy and tissue engineering, and is expected to contribute to urethral tissue reconstruction.
Collapse
Affiliation(s)
- Shilin Zhang
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China.
| | - Jierong Li
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China
| | - Chunjing Li
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China
| | - Jun He
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China
| | - Fengsheng Ling
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China
| | - Guoqing Liu
- Department of Urology, Child Healthcare Hospital, Southern Medical University, Chancheng District, Affiliated Foshan Maternity &No.11, Renmin West Road, 528000, Foshan, Guangdong, China
| |
Collapse
|
22
|
Cell Technologies in the Stress Urinary Incontinence Correction. Biomedicines 2022; 10:biomedicines10020309. [PMID: 35203518 PMCID: PMC8869383 DOI: 10.3390/biomedicines10020309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
The scientific literature of recent years contains a lot of data about using multipotent stromal cells (MSCs) for urinary incontinence correction. Despite this, the ideal treatment method for urinary incontinence has not yet been created. The cell therapy results in patients and experimental animals with incontinence have shown promising results, but the procedures require further optimization, and more research is needed to focus on the clinical phase. The MSC use appears to be a feasible, safe, and effective method of treatment for patients with urinary incontinence. However, the best mode for application of cell technology is still under study. Most clinical investigations have been performed on only a few patients and during rather short follow-up periods, which, together with an incomplete knowledge of the mechanisms of MSC action, does not make it possible for their widespread implementation. The technical details regarding the MSC application remain to be identified in more rigorous preclinical and clinical trials.
Collapse
|
23
|
Zhou Q, Cheng Y, Sun F, Shen J, Nasser MI, Zhu P, Zhang X, Li Y, Yin G, Wang Y, Wu X, Zhao M. A Comprehensive Review of the Therapeutic Value of Urine-Derived Stem Cells. Front Genet 2022; 12:781597. [PMID: 35047009 PMCID: PMC8762167 DOI: 10.3389/fgene.2021.781597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022] Open
Abstract
Stem cells possess regenerative powers and multidirectional differentiation potential and play an important role in disease treatment and basic medical research. Urine-derived stem cells (USCs) represent a newly discovered type of stem cell with biological characteristics similar to those of mesenchymal stromal cells (MSCs), including their doubling time and immunophenotype. USCs are noninvasive and can be readily obtained from voided urine and steadily cultured. Based on advances in this field, USCs and their secretions have increasingly emerged as ideal sources. USCs may play regulatory roles in the cellular immune system, oxidative stress, revascularization, apoptosis and autophagy. This review summarizes the applications of USCs in tissue regeneration and various disease treatments. Furthermore, by analysing their limitations, we anticipate the development of more feasible therapeutic strategies to promote USC-based individualized treatment.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yiyu Cheng
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Fang Sun
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jie Shen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xueyan Zhang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuxiang Li
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Guangming Yin
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuequn Wang
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiushan Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
24
|
Huang YZ, He T, Cui J, Jiang YL, Zeng JF, Zhang WQ, Xie HQ. Urine-Derived Stem Cells for Regenerative Medicine: Basic Biology, Applications, and Challenges. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:978-994. [PMID: 35049395 DOI: 10.1089/ten.teb.2021.0142] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Regenerative medicine based on stem cell research has the potential to provide advanced health care for human beings. Recent studies demonstrate that stem cells in human urine can serve as an excellent source of graft cells for regenerative therapy, mainly due to simple, low-cost, and noninvasive cell isolation. These cells, termed human urine-derived stem cells (USCs), are highly expandable and can differentiate into various cell lineages. They share many biological properties with mesenchymal stem cells, such as potent paracrine effects and immunomodulation ability. The advantage of USCs has motivated researchers to explore their applications in regenerative medicine, including genitourinary regeneration, musculoskeletal repair, skin wound healing, and disease treatment. Although USCs have showed many positive outcomes in preclinical studies, and although the possible applications of USCs for animal therapy have been reported, many issues need to be addressed before clinical translation. This article provides a comprehensive review of USC biology and recent advances in their application for tissue regeneration. Challenges in the clinical translation of USC-based therapy are also discussed. Impact statement Recently, stem cells isolated from urine, referred to as urine-derived stem cells (USCs), have gained much interest in the field of regenerative medicine. Many advantages of human USCs have been found for cell-based therapy: (i) the cell isolation procedure is simple and low cost; (ii) they have remarkable proliferation ability, multidifferentiation potential, and paracrine effects; and (iii) they facilitate tissue regeneration in many animal models. With the hope to facilitate the development of USC-based therapy, we describe the current understanding of USC biology, summarize recent advances in their applications, and discuss future challenges in clinical translation.
Collapse
Affiliation(s)
- Yi-Zhou Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Tao He
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China.,Department of Breast Surgery, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, China
| | - Jing Cui
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yan-Lin Jiang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Jun-Feng Zeng
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Wen-Qian Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| |
Collapse
|
25
|
Fakhredini F, Mansouri E, Mard SA, Valizadeh Gorji A, Rashno M, Orazizadeh M. Effects of Exosomes Derived from Kidney Tubular Cells on Diabetic Nephropathy in Rats. CELL JOURNAL 2022; 24:28-35. [PMID: 35182062 PMCID: PMC8876258 DOI: 10.22074/cellj.2022.7591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 08/16/2020] [Indexed: 11/21/2022]
Abstract
OBJECTIVE One of the severe complications and well-known sources of end stage renal disease (ESRD) from diabetes mellitus is diabetic nephropathy (DN). Exosomes secreted from diverse cells are one of the novel encouraging therapies for chronic renal injuries. In this study, we assess whether extracted exosomes from kidney tubular cells (KTCs) could prevent early stage DN in vivo. MATERIALS AND METHODS In this experimental, exosomes from conditioned medium of rabbit KTCs (RK13) were purified by ultracentrifuge procedures. The exosomes were assessed in terms of morphology and size, and particular biomarkers were evaluated by transmission electron microscopy (TEM), scanning electron microscopy (SEM), Western blot, atomic force microscopy (AFM) and Zetasizer Nano analysis. The rats were divided into four groups: DN, control, DN treated with exosomes and sham. First, diabetes was induced in the rats by intraperitoneial (i.p.) administration of streptozotocin (STZ, 50 mg/kg body weight). Then, the exosomes were injected each week into their tail vein for six weeks. We measured 24-hour urine protein, blood urea nitrogen (BUN), and serum creatinine (Scr) levels with detection kits. The histopathological effects of the exosomes on kidneys were evaluated by periodic acid-Schiff (PAS) staining and expressions of miRNA-29a and miRNA-377 by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS The KTC-Exos were approximately 50-150 nm and had a spherical morphology. They expressed the CD9 and CD63 specific markers. Intravenous injections of KTC-Exos potentially reduced urine volume (P<0.0001), and 24- hour protein (P<0.01), BUN (P<0.001) and Scr (P<0.0001) levels. There was a decrease in miRNA-377 (P<0.01) and increase in miRNA-29a (P<0.001) in the diabetic rats. KTC-Exos ameliorated the renal histopathology with regulatory changes in microRNAs (miRNA) expressions. CONCLUSION KTC-Exos plays a role in attenuation of kidney injury from diabetes by regulating the miRNAs associated with DN.
Collapse
Affiliation(s)
- Fereshtesadat Fakhredini
- Cell and Molecular Research Centre, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Esrafil Mansouri
- Cell and Molecular Research Centre, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Ali Mard
- Department of Physiology, Physiology Research Centre, Research Institute for Infectious Diseases of the Digestive System, School of
Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Armita Valizadeh Gorji
- Department of Bone Marrow Transplantation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Rashno
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Orazizadeh
- Cell and Molecular Research Centre, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran,P.O.Box: 61335Department of Anatomical SciencesFaculty of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| |
Collapse
|
26
|
De La Torre P, Pérez-Lorenzo MJ, Alcázar-Garrido Á, Collado J, Martínez-López M, Forcén L, Masero-Casasola AR, García A, Gutiérrez-Vélez MC, Medina-Polo J, Muñoz E, Flores AI. Perinatal mesenchymal stromal cells of the human decidua restore continence in rats with stress urinary incontinence induced by simulated birth trauma and regulate senescence of fibroblasts from women with stress urinary incontinence. Front Cell Dev Biol 2022; 10:1033080. [PMID: 36742196 PMCID: PMC9893794 DOI: 10.3389/fcell.2022.1033080] [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: 08/31/2022] [Accepted: 12/29/2022] [Indexed: 01/19/2023] Open
Abstract
Stress urinary incontinence (SUI) is a condition that causes the involuntary loss of urine when making small efforts, which seriously affects daily life of people who suffer from it. Women are more affected by this form of incontinence than men, since parity is the main risk factor. Weakening of the pelvic floor tissues is the cause of SUI, although a complete understanding of the cellular and molecular mechanisms of the pathology is still lacking. Reconstructive surgery to strengthen tissue in SUI patients is often associated with complications and/or is ineffective. Mesenchymal stromal cells from the maternal side of the placenta, i.e. the decidua, are proposed here as a therapeutic alternative based on the regenerative potential of mesenchymal cells. The animal model of SUI due to vaginal distention simulating labor has been used, and decidual mesenchymal stromal cell (DMSC) transplantation was effective in preventing a drop in pressure at the leak point in treated animals. Histological analysis of the urethras from DMSC-treated animals after VD showed recovery of the muscle fiber integrity, low or no extracellular matrix (ECM) infiltration and larger elastic fibers near the external urethral sphincter, compared to control animals. Cells isolated from the suburethral connective tissue of SUI patients were characterized as myofibroblasts, based on the expression of several specific genes and proteins, and were shown to achieve premature replicative senescence. Co-culture of SUI myofibroblasts with DMSC via transwell revealed a paracrine interaction between the cells through signals that mediated DMSC migration, SUI myofibroblast proliferation, and modulation of the proinflammatory and ECM-degrading milieu that is characteristic of senescence. In conclusion, DMSC could be an alternative therapeutic option for SUI by counteracting the effects of senescence in damaged pelvic tissue.
Collapse
Affiliation(s)
- Paz De La Torre
- Regenerative Medicine Group, Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Álvaro Alcázar-Garrido
- Regenerative Medicine Group, Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Jennifer Collado
- Regenerative Medicine Group, Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Laura Forcén
- Regenerative Medicine Group, Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
- Obstetrics and Gynecology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ana R. Masero-Casasola
- Regenerative Medicine Group, Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
- Obstetrics and Gynecology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alicia García
- Regenerative Medicine Group, Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
- Obstetrics and Gynecology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Mª Carmen Gutiérrez-Vélez
- Regenerative Medicine Group, Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
- Obstetrics and Gynecology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - José Medina-Polo
- Male’s Integral Health Group, Urology Department, Research Institute Hospital 12 de Octubre (imas12), Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Eloy Muñoz
- Regenerative Medicine Group, Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
- Obstetrics and Gynecology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ana I. Flores
- Regenerative Medicine Group, Research Institute Hospital 12 de Octubre (imas12), Madrid, Spain
- *Correspondence: Ana I. Flores,
| |
Collapse
|
27
|
Zhang H, Wang L, Xiang Y, Wang Y, Li H. Nampt promotes fibroblast extracellular matrix degradation in stress urinary incontinence by inhibiting autophagy. Bioengineered 2021; 13:481-495. [PMID: 34967693 PMCID: PMC8805819 DOI: 10.1080/21655979.2021.2009417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stress urinary incontinence (SUI) is defined as involuntary urinary leakage happening in exertion. Nicotinamide phosphoribosyltransferase (Nampt) is seldom researched in the pathogenesis of SUI. Accordingly, the current study set out to elucidate the role of Nampt in SUI progression. Firstly, we determined Nampt expression patterns in SUI patients and rat models. In addition, fibroblasts were obtained from the anterior vaginal wall tissues of non-SUI patients and subjected to treatment with different concentrations of interleukin-1β (IL-1β), followed by quantification of Nampt expressions in fibroblasts. Subsequently, an appropriate concentration of IL-1β was selected to treat anterior vaginal wall fibroblasts. Nampt was further silenced in IL-1β-treated fibroblasts to assess the role of Nampt in autophagy and extracellular matrix (ECM) degradation. Lastly, functional rescue assays were carried out to inhibit autophagy and evaluate the role of autophagy in the mechanism of Nampt modulating IL-1β-treated fibroblast ECM degradation. It was found that Nampt was highly-expressed in SUI patients and rat models and IL-1β-treated fibroblasts. On the other hand, Nampt silencing was found to suppress ECM degradation and promote SUI fibroblast autophagy. Additionally, inhibition of autophagy attenuated the inhibitory effects of Nampt silencing on SUI fibroblast ECM degradation. Collectively, our findings revealed that Nampt was over-expressed in SUI, whereas Nampt silencing enhanced SUI fibroblast autophagy, and thereby inhibited ECM degradation.
Collapse
Affiliation(s)
- Hui Zhang
- Gynecology II Ward, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province, China
| | - Lu Wang
- Gynecology II Ward, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yuancui Xiang
- Gynecology II Ward, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yali Wang
- Gynecology II Ward, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hongjuan Li
- Gynecology II Ward, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
28
|
Zhang Y, Chen J, Fu H, Kuang S, He F, Zhang M, Shen Z, Qin W, Lin Z, Huang S. Exosomes derived from 3D-cultured MSCs improve therapeutic effects in periodontitis and experimental colitis and restore the Th17 cell/Treg balance in inflamed periodontium. Int J Oral Sci 2021; 13:43. [PMID: 34907166 PMCID: PMC8671433 DOI: 10.1038/s41368-021-00150-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 01/07/2023] Open
Abstract
Although mesenchymal stem cell-derived exosomes (MSC-exos) have been shown to have therapeutic effects in experimental periodontitis, their drawbacks, such as low yield and limited efficacy, have hampered their clinical application. These drawbacks can be largely reduced by replacing the traditional 2D culture system with a 3D system. However, the potential function of MSC-exos produced by 3D culture (3D-exos) in periodontitis remains elusive. This study showed that compared with MSC-exos generated via 2D culture (2D-exos), 3D-exos showed enhanced anti-inflammatory effects in a ligature-induced model of periodontitis by restoring the reactive T helper 17 (Th17) cell/Treg balance in inflamed periodontal tissues. Mechanistically, 3D-exos exhibited greater enrichment of miR-1246, which can suppress the expression of Nfat5, a key factor that mediates Th17 cell polarization in a sequence-dependent manner. Furthermore, we found that recovery of the Th17 cell/Treg balance in the inflamed periodontium by the local injection of 3D-exos attenuated experimental colitis. Our study not only showed that by restoring the Th17 cell/Treg balance through the miR-1246/Nfat5 axis, the 3D culture system improved the function of MSC-exos in the treatment of periodontitis, but also it provided a basis for treating inflammatory bowel disease (IBD) by restoring immune responses in the inflamed periodontium.
Collapse
Affiliation(s)
- Yong Zhang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiayao Chen
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Haijun Fu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuhong Kuang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Feng He
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Min Zhang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zongshan Shen
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wei Qin
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhengmei Lin
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Shuheng Huang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
29
|
Zhang W, Hu J, Huang Y, Wu C, Xie H. Urine-derived stem cells: applications in skin, bone and articular cartilage repair. BURNS & TRAUMA 2021; 9:tkab039. [PMID: 34859109 PMCID: PMC8633594 DOI: 10.1093/burnst/tkab039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/18/2021] [Indexed: 02/05/2023]
Abstract
As an emerging type of adult stem cell featuring non-invasive acquisition, urine-derived stem cells (USCs) have shown great potential for applications in tissue engineering and regenerative medicine. With a growing amount of research on the topic, the effectiveness of USCs in various disease models has been shown and the underlying mechanisms have also been explored, though many aspects still remain unclear. In this review, we aim to provide an up-to-date overview of the biological characteristics of USCs and their applications in skin, bone and articular cartilage repair. In addition to the identification procedure of USCs, we also summarize current knowledge of the underlying repair mechanisms and application modes of USCs. Potential concerns and perspectives have also been summarized.
Collapse
Affiliation(s)
- Wenqian Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jungen Hu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yizhou Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chenyu Wu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huiqi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
30
|
Singh A, Raghav A, Shiekh PA, Kumar A. Transplantation of engineered exosomes derived from bone marrow mesenchymal stromal cells ameliorate diabetic peripheral neuropathy under electrical stimulation. Bioact Mater 2021; 6:2231-2249. [PMID: 33553812 PMCID: PMC7829156 DOI: 10.1016/j.bioactmat.2021.01.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/23/2020] [Accepted: 01/08/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a long-term complication associated with nerve dysfunction and uncontrolled hyperglycemia. In spite of new drug discoveries, development of effective therapy is much needed to cure DPN. Here, we have developed a combinatorial approach to provide biochemical and electrical cues, considered to be important for nerve regeneration. Exosomes derived from bone marrow mesenchymal stromal cells (BMSCs) were fused with polypyrrole nanoparticles (PpyNps) containing liposomes to deliver both the cues in a single delivery vehicle. We developed DPN rat model and injected intramuscularly the fused exosomal system to understand its long-term therapeutic effect. We found that the fused system along with electrical stimulation normalized the nerve conduction velocity (57.60 ± 0.45 m/s) and compound muscle action potential (16.96 ± 0.73 mV) similar to healthy control (58.53 ± 1.10 m/s; 18.19 ± 1.45 mV). Gastrocnemius muscle morphology, muscle mass, and integrity were recovered after treatment. Interestingly, we also observed paracrine effect of delivered exosomes in controlling hyperglycemia and loss in body weight and also showed attenuation of damage to the tissues such as the pancreas, kidney, and liver. This work provides a promising effective treatment and also contribute cutting edge therapeutic approach for the treatment of DPN.
Collapse
Affiliation(s)
- Anamika Singh
- Biomaterial and Tissue Engineering Group, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Alok Raghav
- Biomaterial and Tissue Engineering Group, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Parvaiz Ahmad Shiekh
- Biomaterial and Tissue Engineering Group, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| | - Ashok Kumar
- Biomaterial and Tissue Engineering Group, Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
- Centre for Nanosciences, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
- Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, UP, India
| |
Collapse
|
31
|
Integrin-Linked Kinase (ILK) Regulates Urinary Stem Cells Differentiation into Smooth Muscle via NF- κB Signal Pathway. Stem Cells Int 2021; 2021:6633111. [PMID: 33854551 PMCID: PMC8019365 DOI: 10.1155/2021/6633111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/28/2021] [Accepted: 03/09/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives Urinary stem cells (USCs) have the capacity for unlimited growth and are promising tools for the investigations of cell differentiation and urinary regeneration. However, the limited life span significantly restricts their usefulness. This study is aimed at exploring the effect of integrin-linked kinase (ILK) on the smooth muscle cells (SMCs) differentiation of the dog USCs and investigating its molecular mechanism. Methods An immortalized USCs cell line with the molecular markers and biological functions was prepared. After successfully inducing the differentiation of USCs into SMCs, the expression level of the unique key factor and its mechanisms in this process was determined through real-time polymerase chain reaction, Western blot, or Immunofluorescence staining. Results We found that high cell density promoted USCs differentiation SMCs, and ILK was necessary for USCs differentiation into SMCs. Knocking down ILK decreased the expression of SMCs specific-marker, while using a selective ILK agonist increased the expression of SMCs specific-marker. Furthermore, ILK regulated SMCs differentiation in part through the activation of NF-κB pathway in USCs. A NF-κB activity assay showed overexpression of ILK could significantly upregulate NF-κB p50 expression, and NF-κB p50 acts as downstream signal molecular of ILK. Conclusion High cell density induces the differentiation of USCs into SMCs, and ILK is a key regulator of myogenesis. Furthermore, NF-κB signaling pathway might play a crucial role in this process.
Collapse
|
32
|
Fuloria S, Subramaniyan V, Dahiya R, Dahiya S, Sudhakar K, Kumari U, Sathasivam K, Meenakshi DU, Wu YS, Sekar M, Malviya R, Singh A, Fuloria NK. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Regenerative Potential and Challenges. BIOLOGY 2021; 10:172. [PMID: 33668707 PMCID: PMC7996168 DOI: 10.3390/biology10030172] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Evidence suggests that stem cells exert regenerative potential via the release of extracellular vesicles. Mesenchymal stem cell extracellular vesicles (MSCEVs) offer therapeutic benefits for various pathophysiological ailments by restoring tissues. Facts suggest that MSCEV action can be potentiated by modifying the mesenchymal stem cells culturing methodology and bioengineering EVs. Limited clinical trials of MSCEVs have questioned their superiority, culturing quality, production scale-up and isolation, and administration format. Translation of preclinically successful MSCEVs into a clinical platform requires paying attention to several critical matters, such as the production technique, quantification/characterization, pharmacokinetics/targeting/transfer to the target site, and the safety profile. Keeping these issues as a priority, the present review was designed to highlight the challenges in translating preclinical MSCEV research into clinical platforms and provide evidence for the regenerative potential of MSCEVs in various conditions of the liver, kidney, heart, nervous system, bone, muscle, cartilage, and other organs/tissues.
Collapse
Affiliation(s)
| | - Vetriselvan Subramaniyan
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur 42610, Malaysia; (V.S.); (Y.S.W.)
| | - Rajiv Dahiya
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago;
| | - Sunita Dahiya
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA;
| | - Kalvatala Sudhakar
- School of Pharmaceutical Sciences (LIT-Pharmacy), Lovely Professional University, Jalandhar 144411, India;
| | - Usha Kumari
- Faculty of Medicine, AIMST University, Kedah 08100, Malaysia;
| | | | | | - Yuan Seng Wu
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur 42610, Malaysia; (V.S.); (Y.S.W.)
| | - Mahendran Sekar
- Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh 30450, Malaysia;
| | - Rishabha Malviya
- Department of Pharmacy, SMAS, Galgotias University, Greater Noida 203201, India; (R.M.); (A.S.)
| | - Amit Singh
- Department of Pharmacy, SMAS, Galgotias University, Greater Noida 203201, India; (R.M.); (A.S.)
| | | |
Collapse
|
33
|
Characterization of stem cells from human ovarian follicular fluid; a potential source of autologous stem cell for cell-based therapy. Hum Cell 2021; 34:300-309. [PMID: 33543452 DOI: 10.1007/s13577-020-00439-2] [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: 05/24/2020] [Accepted: 09/19/2020] [Indexed: 10/22/2022]
Abstract
Human ovarian follicular fluid (HOFF) contains proteins, extracellular matrixes necessary for growth and maturation of oocytes as well as granulosa cells. Epithelial cells and stem cells can be isolated from HOFF. However, information regarding stem cells derived from HOFF is still lacking. The objectives of the present study were to isolate, characterize, and differentiate cells derived from HOFF. HOFF was collected during the routine aspiration of oocytes in an assisted fertilization program and subjected to cell isolation, characterization, and in vitro culture. After 24 h of culture, different cell morphologies including epithelial-like-, neural-like- and fibroblast-like cells were observed. Immunocytochemistry reveals the expression of pluripotent stem cell markers (OCT4, NANOG, SSEA4), epithelial marker (CK18), FSH- and LH-receptor. For in vitro culture, the isolated cells were continuously cultured in a growth medium; alpha MEM containing 10% FBS and epidermal growth factor (EGF). After 2 weeks of in vitro culture, cells with fibroblast-like morphology dominantly grow in the culture vessels and resemble mesenchymal stem cells (MSCs). HOFF-derived cells exhibited MSC expression of CD44, CD73, CD90, CD105, CD146, and STRO-1, and were capable of differentiation into osteoblasts, chondrocytes, and adipocytes. After induction of neural differentiation, HOFF-derived cells formed spheroidal structures and expressed neural stem cell markers including Nestin, β-tubulin III, and O4. Besides, the oocyte-like structure was observed after prolonged culture of HOFF. In conclusion, cells derived from follicular fluid exhibited stem cell characteristics, which could be useful for regenerative medicine applications and cell-based therapies.
Collapse
|
34
|
Guan F, Wu J, Li J, Liu S, Weng Y, Chen T, Yang T, Fang K. RETRACTED ARTICLE: Inducible nitric oxide synthase promotes differentiation of satellite cells and prevents stress urinary incontinence via HGF-mediated p38/MAPK signaling. World J Urol 2021; 39:633. [PMID: 32556674 DOI: 10.1007/s00345-020-03289-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 05/28/2020] [Indexed: 01/06/2023] Open
Affiliation(s)
- Fei Guan
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China
- The 4th Hospital of Changde, Changde, 415000, People's Republic of China
| | - Jing Wu
- Department of Biochemistry and Molecular Biology, The Primary Medicine School of Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Jiang Li
- Department of Urology, The 1st Hospital of Qujing, Qujing, 650500, People's Republic of China
| | - Shang Liu
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China
| | - Yuting Weng
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China
| | - Tao Chen
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China
| | - Tongxin Yang
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China
| | - Kewei Fang
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China.
| |
Collapse
|
35
|
Zidan AA, Al-Hawwas M, Perkins GB, Mourad GM, Stapledon CJM, Bobrovskaya L, Zhou XF, Hurtado PR. Characterization of Urine Stem Cell-Derived Extracellular Vesicles Reveals B Cell Stimulating Cargo. Int J Mol Sci 2021; 22:E459. [PMID: 33466423 PMCID: PMC7796485 DOI: 10.3390/ijms22010459] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 12/31/2020] [Accepted: 01/01/2021] [Indexed: 12/22/2022] Open
Abstract
Elucidation of the biological functions of extracellular vesicles (EVs) and their potential roles in physiological and pathological processes is an expanding field of research. In this study, we characterized USC-derived EVs and studied their capacity to modulate the human immune response in vitro. We found that the USC-derived EVs are a heterogeneous population, ranging in size from that of micro-vesicles (150 nm-1 μm) down to that of exosomes (60-150 nm). Regarding their immunomodulatory functions, we found that upon isolation, the EVs (60-150 nm) induced B cell proliferation and IgM antibody secretion. Analysis of the EV contents unexpectedly revealed the presence of BAFF, APRIL, IL-6, and CD40L, all known to play a central role in B cell stimulation, differentiation, and humoral immunity. In regard to their effect on T cell functions, they resembled the function of mesenchymal stem cell (MSC)-derived EVs previously described, suppressing T cell response to activation. The finding that USC-derived EVs transport a potent bioactive cargo opens the door to a novel therapeutic avenue for boosting B cell responses in immunodeficiency or cancer.
Collapse
Affiliation(s)
- Asmaa A. Zidan
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.A.Z.); (M.A.-H.); (L.B.)
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria 21568, Egypt;
- Centre of Excellence for Research in Regenerative Medicine Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria 21568, Egypt
| | - Mohammed Al-Hawwas
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.A.Z.); (M.A.-H.); (L.B.)
| | - Griffith B. Perkins
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA 5000, Australia;
| | - Ghada M. Mourad
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria 21568, Egypt;
- Centre of Excellence for Research in Regenerative Medicine Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria 21568, Egypt
| | | | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.A.Z.); (M.A.-H.); (L.B.)
| | - Xin-Fu Zhou
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (A.A.Z.); (M.A.-H.); (L.B.)
| | - Plinio R. Hurtado
- Department of Renal Medicine, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- School of Medicine, University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
36
|
Wang L, Wang Y, Xiang Y, Ma J, Zhang H, Dai J, Hou Y, Yang Y, Ma J, Li H. An In Vitro Study on Extracellular Vesicles From Adipose-Derived Mesenchymal Stem Cells in Protecting Stress Urinary Incontinence Through MicroRNA-93/F3 Axis. Front Endocrinol (Lausanne) 2021; 12:693977. [PMID: 34484115 PMCID: PMC8415505 DOI: 10.3389/fendo.2021.693977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/12/2021] [Indexed: 11/18/2022] Open
Abstract
Since the potential roles of extracellular vesicles secreted by adipose-derived mesenchymal stem cells (ADSCs) are not well understood in collagen metabolism, the purpose of this research was to evaluate the effects of ADSCs-extracellular vesicles in stress urinary incontinence and the regulatory mechanism of delivered microRNA-93 (miR-93). ADSCs were isolated and cultured, and ADSCs-extracellular vesicles were extracted and identified. Stress urinary incontinence primary fibroblasts or satellite cells were treated with ADSCs-extracellular vesicles to detect the expression of Elastin, Collagen I, and Collagen III in fibroblasts and Pax7 and MyoD in satellite cells. After transfecting ADSCs with miR-93 mimics or inhibitors, extracellular vesicles were isolated and treated with stress urinary incontinence primary fibroblasts or satellite cells to observe cell function changes. The online prediction and luciferase activity assay confirmed the targeting relationship between miR-93 and coagulation factor III (F3). The rescue experiment verified the role of ADSCs-extracellular vesicles carrying miR-93 in stress urinary incontinence primary fibroblasts and satellite cells by targeting F3. ADSCs-extracellular vesicles treatment upregulated expression of Elastin, Collagen I, and Collagen III in stress urinary incontinence primary fibroblasts and expression of Pax7 and MyoD in stress urinary incontinence primary satellite cells. miR-93 expression was increased in stress urinary incontinence primary fibroblasts or satellite cells treated with ADSCs-extracellular vesicles. Extracellular vesicles secreted by ADSCs could deliver miR-93 to fibroblasts and then negatively regulate F3 expression; ADSCs-extracellular vesicles could reverse the effect of F3 on extracellular matrix remodeling in stress urinary incontinence fibroblasts. miR-93 expression was also increased in stress urinary incontinence primary satellite cells treated by ADSCs-extracellular vesicles. Extracellular vesicles secreted by ADSCs were delivered to satellite cells through miR-93, which directly targets F3 expression and upregulates Pax7 and MyoD expression in satellite cells. Our study indicates that miR-93 delivered by ADSCs-extracellular vesicles could regulate extracellular matrix remodeling of stress urinary incontinence fibroblasts and promote activation of stress urinary incontinence satellite cells through targeting F3.
Collapse
|
37
|
Zhang Y, Wang J, Yang B, Qiao R, Li A, Guo H, Ding J, Li H, Ye H, Wu D, Cui L, Yang S. Transfer of MicroRNA-216a-5p From Exosomes Secreted by Human Urine-Derived Stem Cells Reduces Renal Ischemia/Reperfusion Injury. Front Cell Dev Biol 2020; 8:610587. [PMID: 33415108 PMCID: PMC7783217 DOI: 10.3389/fcell.2020.610587] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/23/2020] [Indexed: 12/19/2022] Open
Abstract
Human urine-derived stem cells (USCs) protect rats against kidney ischemia/reperfusion (I/R) injury. Here we investigated the role of USCs exosomes (USCs-Exos) in protecting tubular endothelial cells and miRNA transfer in the kidney. Human USCs and USCs-Exos were isolated and verified by morphology and specific biomarkers. USC-Exos played a protective role in human proximal tubular epithelial cells (HK-2) exposed to hypoxia/reoxygenation (H/R). USCs-Exos were rich in miR-216a-5p, which targeted phosphatase and tensin homolog (PTEN) and regulated cell apoptosis through the Akt pathway. In HK-2 cells exposed to H/R, incubation with USC-Exos increased miR-216-5p, decreased PTEN levels, and stimulated Akt phosphorylation. Exposure of hypoxic HK-2 cells to USCs-Exos pretreated with anti-miR-216a-5p can prevent the increase of miR-216-5p and Akt phosphorylation levels, restore PTEN expression, and promote apoptosis. The dual-luciferase reported gene assay in HK-2 cells confirmed that miR-216a-5p targeted PTEN. In rats with I/R injury, intravenous infusion of USCs-Exos can effectively induce apoptosis suppression and functional protection, which is associated with decreased PTEN. Infusion of exosomes from anti-miR-216a-5p-transfected USCs weakened the protective effect in the I/R model. Therefore, USCs-Exos can reduce renal I/R injury by transferring miR-216a-5p targeting PTEN. Potentially, USCs-Exos rich in miR-216a-5p can serve as a promising therapeutic option for AKI.
Collapse
Affiliation(s)
- Yinmei Zhang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Junxiong Wang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Rui Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Aiwei Li
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Han Guo
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Jie Ding
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Hui Li
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Hong Ye
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Di Wu
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
38
|
Abstract
Mesenchymal stem cells (MSCs), also referred to as multipotent stromal cells or mesenchymal stromal cells, are present in multiple tissues and capable of differentiating into diverse cell lineages, holding a great promise in developing cell-based therapy for a wide range of conditions. Pelvic floor disorders (PFDs) is a common degenerative disease in women and may diminish a woman's quality of life at any age. Since the treatments for this disease are limited by the high rates of recurrence and surgical complications, seeking an ideal therapy in the restoration of pelvic floor function is an urgent issue at present. Herein, we summarize the cell sources of MSCs used for PFDs and discuss the potential mechanisms of MSCs in treating PFDs. Specifically, we also provide a comprehensive review of current preclinical and clinical trials dedicated to investigating MSC-based therapy for PFDs. The novel therapy has presented promising therapeutic effects which include relieving the symptoms of urinary or fecal incontinence, improving the biological properties of implanted meshes and promoting the injured tissue repair. Nevertheless, MSC-based therapies for PFDs are still experimental and the unstated issues on their safety and efficacy should be carefully addressed before their clinical applications.
Collapse
|
39
|
Garcia-Arranz M, Alonso-Gregorio S, Fontana-Portella P, Bravo E, Diez Sebastian J, Fernandez-Santos ME, Garcia-Olmo D. Two phase I/II clinical trials for the treatment of urinary incontinence with autologous mesenchymal stem cells. Stem Cells Transl Med 2020; 9:1500-1508. [PMID: 32864818 PMCID: PMC7695632 DOI: 10.1002/sctm.19-0431] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 06/06/2020] [Accepted: 07/19/2020] [Indexed: 12/21/2022] Open
Abstract
We evaluated the safety and feasibility of adipose‐derived mesenchymal stem cells to treat endoscopically urinary incontinence after radical prostatectomy in men or female stress urinary. We designed two prospective, nonrandomized phase I‐IIa clinical trials of urinary incontinence involving 9 men (8 treated) and 10 women to test the feasibility and safety of autologous mesenchymal stem cells for this use. Cells were obtained from liposuction containing 150 to 200 g of fat performed on every patient. After 4 to 6 weeks and under sedation, endoscopic intraurethral injection of the cells was performed. On each visit (baseline, 1, 3, 6, and 12 months), clinical parameters were measured, and blood samples, urine culture, and uroflowmetry were performed. Every patient underwent an urethrocystoscopy and urodynamic studies on the first and last visit. Data from pad test, quality‐of‐life and incontinence questionnaires, and pads used per day were collected at every visit. Statistical analysis was done by Wilcoxon signed‐rank test. No adverse effects were observed. Three men (37.5%) and five women (50%) showed an objective improvement of >50% (P < .05) and a subjective improvement of 70% to 80% from baseline. In conclusion, intraurethral application of stem cells derived from adipose tissue is a safe and feasible procedure to treat urinary incontinence after radical prostatectomy or in female stress urinary incontinence. A statistically significant difference was obtained for pad‐test improvement in 3/8 men and 5/10 women. Our results encourage studies to confirm safety and to analyze efficacy.
Collapse
Affiliation(s)
- Mariano Garcia-Arranz
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Madrid, Spain.,Surgery Department, Autonoma University of Madrid, Madrid, Spain
| | | | | | - Elena Bravo
- Department of Plastic Surgery, La Paz University Hospital, Madrid, Spain
| | | | | | - Damian Garcia-Olmo
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Madrid, Spain.,Surgery Department, Autonoma University of Madrid, Madrid, Spain
| |
Collapse
|
40
|
Magnetic targeting enhances the cutaneous wound healing effects of human mesenchymal stem cell-derived iron oxide exosomes. J Nanobiotechnology 2020; 18:113. [PMID: 32799868 PMCID: PMC7429707 DOI: 10.1186/s12951-020-00670-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Human mesenchymal stem cell (MSC)-derived exosomes (Exos) are a promising therapeutic agent for cell-free regenerative medicine. However, their poor organ-targeting ability and therapeutic efficacy have been found to critically limit their clinical applications. In the present study, we fabricated iron oxide nanoparticle (NP)-labeled exosomes (Exo + NPs) from NP-treated MSCs and evaluated their therapeutic efficacy in a clinically relevant model of skin injury. We found that the Exos could be readily internalized by human umbilical vein endothelial cells (HUVECs), and could significantly promote their proliferation, migration, and angiogenesis both in vitro and in vivo. Moreover, the protein expression of proliferative markers (Cyclin D1 and Cyclin A2), growth factors (VEGFA), and migration-related chemokines (CXCL12) was significantly upregulated after Exo treatment. Unlike the Exos prepared from untreated MSCs, the Exo + NPs contained NPs that acted as a magnet-guided navigation tool. The in vivo systemic injection of Exo + NPs with magnetic guidance significantly increased the number of Exo + NPs that accumulated at the injury site. Furthermore, these accumulated Exo + NPs significantly enhanced endothelial cell proliferation, migration, and angiogenic tubule formation in vivo; moreover, they reduced scar formation and increased CK19, PCNA, and collagen expression in vivo. Collectively, these findings confirm the development of therapeutically efficacious extracellular nanovesicles and demonstrate their feasibility in cutaneous wound repair.
Collapse
|
41
|
Follicle-stimulating hormone promotes the proliferation of epithelial ovarian cancer cells by activating sphingosine kinase. Sci Rep 2020; 10:13834. [PMID: 32796926 PMCID: PMC7428003 DOI: 10.1038/s41598-020-70896-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 08/05/2020] [Indexed: 01/01/2023] Open
Abstract
Follicle-stimulating hormone (FSH) is closely related to the pathogenesis and progression of epithelial ovarian cancer (EOC). However, until now, knowledge relating to FSH-driven signalling pathways that lead to the growth of EOC remained incomplete. We sought to explore whether sphingosine kinase (SphK) could mediate FSH-induced ovarian cancer cell proliferation and which pathway might be involved in this process. The expression of phospho-SphK1 and phospho-SphK2 was detected in sections of EOC tissues by Immunohistochemical staining, and clinical significances were analyzed by statistical analysis. EOC cells were treated with FSH or/and SKI-II. CCK8 assays and colony formation assays were used to investigate cell proliferation. Western blot was carried out to detect protein expression in EOC cell line after treated with FSH. Here, for the first time, we provide evidence that high expression levels of phospho-SphK1 and phospho-SphK2 were both prognostic indicators of overall survival (OS) in EOC. Additionally, the expression levels of both phospho-SphK1 and phospho-SphK2 were closely correlated with the expression level of follicle-stimulating hormone receptor (FSHR) in ovarian cancer tissues. FSH stimulated the phosphorylation of both SphK1 and SphK2 and was able to regulate the survival and growth of ovarian cancer cells by activating SphK1 and SphK2 through ERK1/2. Both isoenzymes of SphK were equally responsible for FSH-induced cell proliferation of EOC. Both Erk1/2 and Akt activation play important roles in mediating FSH-induced cell proliferation after phosphorylation of SphK. Moreover, our data demonstrated that S1P receptor 1 (S1PR1) and S1PR3, key components of the SphK signalling system, were involved in FSH-mediated proliferation of EOC. Taken together, the results of the current study revealed that SphK is an essential mediator in FSH-induced proliferation of ovarian cancer cells in EOC, which indicates a new signalling pathway that controls FSH-mediated growth in EOC and suggests a new strategy that pharmaceutically targets both isoenzymes of SphK for the management of ovarian cancer.
Collapse
|
42
|
Wang Y, Zhang Y, Cai G, Li Q. Exosomes as Actively Targeted Nanocarriers for Cancer Therapy. Int J Nanomedicine 2020; 15:4257-4273. [PMID: 32606676 PMCID: PMC7306454 DOI: 10.2147/ijn.s239548] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
In recent years, it has been found that exosomes can be used as nanocarriers, which can be used in the treatment of tumors by carrying contents. The exosomes are derived from the secretion of the organism's own cells and are characterized by a phospholipid bilayer structure and a small particle size. These characteristics guarantee that the exosomes can carry a wide range of tumor drugs, deliver the drug to the cancer, and reduce or eliminate the tumor drug band. The toxic side effects were significantly eliminated; meanwhile, the therapeutic effects of the drug on the tumor were remarkably improved. This paper reviewed the strategies and drugs presented by different scholars for the treatment of tumors based on the drugs carried by exosomes.
Collapse
Affiliation(s)
- Yan Wang
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai201203, People’s Republic of China
| | - Yingru Zhang
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai201203, People’s Republic of China
| | - Gang Cai
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai201203, People’s Republic of China
| | - Qi Li
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai201203, People’s Republic of China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai201203, People’s Republic of China
| |
Collapse
|
43
|
Urinary Extracellular Vesicles as Biomarkers of Kidney Disease: From Diagnostics to Therapeutics. Diagnostics (Basel) 2020; 10:diagnostics10050311. [PMID: 32429335 PMCID: PMC7277956 DOI: 10.3390/diagnostics10050311] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
Cell-derived extracellular vesicles (EVs) can be isolated from various body fluids, including urine. Urinary EVs have gained important recognition as potential diagnostic biomarkers in renal disease since their cargo includes nucleic acids, proteins, and other cellular components, which likely mirror the physiological and possibly pathophysiological state of cells along the nephron. Accumulating evidence highlights the feasibility of using EVs as biomarkers for diagnostic, prognostic, and therapeutic purposes in several forms of renal disease, such as acute kidney injury, glomerulonephritis, and renal transplantation. Additionally, exogenous delivery of EVs released in vitro by cells in culture may have salutary benefits for renal diseases. In this review, we introduce recent studies that attempt to identify urinary EVs as candidate biomarkers for human kidney diseases and consider their potential implication as a therapeutic option in key kidney diseases.
Collapse
|
44
|
Tsiapalis D, O’Driscoll L. Mesenchymal Stem Cell Derived Extracellular Vesicles for Tissue Engineering and Regenerative Medicine Applications. Cells 2020; 9:E991. [PMID: 32316248 PMCID: PMC7226943 DOI: 10.3390/cells9040991] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are being extensively investigated for their potential in tissue engineering and regenerative medicine. However, recent evidence suggests that the beneficial effects of MSCs may be manifest by their released extracellular vesicles (EVs); typically not requiring the administration of MSCs. This evidence, predominantly from pre-clinical in vitro and in vivo studies, suggests that MSC-EVs may exhibit substantial therapeutic properties in many pathophysiological conditions, potentially restoring an extensive range of damaged or diseased tissues and organs. These benefits of MSC EVs are apparently found, regardless of the anatomical or body fluid origin of the MSCs (and include e.g., bone marrow, adipose tissue, umbilical cord, urine, etc). Furthermore, early indications suggest that the favourable effects of MSC-EVs could be further enhanced by modifying the way in which the donor MSCs are cultured (for example, in hypoxic compared to normoxic conditions, in 3D compared to 2D culture formats) and/or if the EVs are subsequently bio-engineered (for example, loaded with specific cargo). So far, few human clinical trials of MSC-EVs have been conducted and questions remain unanswered on whether the heterogeneous population of EVs is beneficial or some specific sub-populations, how best we can culture and scale-up MSC-EV production and isolation for clinical utility, and in what format they should be administered. However, as reviewed here, there is now substantial evidence supporting the use of MSC-EVs in tissue engineering and regenerative medicine and further research to establish how best to exploit this approach for societal and economic benefit is warranted.
Collapse
Affiliation(s)
| | - Lorraine O’Driscoll
- School of Pharmacy and Pharmaceutical Sciences & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland;
| |
Collapse
|
45
|
Floriano JF, Willis G, Catapano F, de Lima PR, Reis FVDS, Barbosa AMP, Rudge MVC, Emanueli C. Exosomes Could Offer New Options to Combat the Long-Term Complications Inflicted by Gestational Diabetes Mellitus. Cells 2020; 9:E675. [PMID: 32164322 PMCID: PMC7140615 DOI: 10.3390/cells9030675] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/20/2020] [Accepted: 02/29/2020] [Indexed: 02/08/2023] Open
Abstract
Gestational diabetes Mellitus (GDM) is a complex clinical condition that promotes pelvic floor myopathy, thus predisposing sufferers to urinary incontinence (UI). GDM usually regresses after birth. Nonetheless, a GDM history is associated with higher risk of subsequently developing type 2 diabetes, cardiovascular diseases (CVD) and UI. Some aspects of the pathophysiology of GDM remain unclear and the associated pathologies (outcomes) are poorly addressed, simultaneously raising public health costs and diminishing women's quality of life. Exosomes are small extracellular vesicles produced and actively secreted by cells as part of their intercellular communication system. Exosomes are heterogenous in their cargo and depending on the cell sources and environment, they can mediate both pathogenetic and therapeutic functions. With the advancement in knowledge of exosomes, new perspectives have emerged to support the mechanistic understanding, prediction/diagnosis and ultimately, treatment of the post-GMD outcomes. Here, we will review recent advances in knowledge of the role of exosomes in GDM and related areas and discuss the possibilities for translating exosomes as therapeutic agents in the GDM clinical setting.
Collapse
Affiliation(s)
- Juliana Ferreira Floriano
- Botucatu Medical School, Sao Paulo State University, 18618687 Botucatu, Brazil; (J.F.F.); (P.R.d.L.); (F.V.D.S.R.); (A.M.P.B.)
| | - Gareth Willis
- Division of Newborn Medicine/Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Francesco Catapano
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| | - Patrícia Rodrigues de Lima
- Botucatu Medical School, Sao Paulo State University, 18618687 Botucatu, Brazil; (J.F.F.); (P.R.d.L.); (F.V.D.S.R.); (A.M.P.B.)
| | | | - Angélica Mercia Pascon Barbosa
- Botucatu Medical School, Sao Paulo State University, 18618687 Botucatu, Brazil; (J.F.F.); (P.R.d.L.); (F.V.D.S.R.); (A.M.P.B.)
| | - Marilza Vieira Cunha Rudge
- Botucatu Medical School, Sao Paulo State University, 18618687 Botucatu, Brazil; (J.F.F.); (P.R.d.L.); (F.V.D.S.R.); (A.M.P.B.)
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK;
| |
Collapse
|
46
|
Hu Y, Zhang Y, Ni CY, Chen CY, Rao SS, Yin H, Huang J, Tan YJ, Wang ZX, Cao J, Liu ZZ, Xie PL, Wu B, Luo J, Xie H. Human umbilical cord mesenchymal stromal cells-derived extracellular vesicles exert potent bone protective effects by CLEC11A-mediated regulation of bone metabolism. Am J Cancer Res 2020; 10:2293-2308. [PMID: 32089743 PMCID: PMC7019162 DOI: 10.7150/thno.39238] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/30/2019] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis and osteoporotic fractures severely compromise quality of life in elderly people and lead to early death. Human umbilical cord mesenchymal stromal cell (MSC)-derived extracellular vesicles (hucMSC-EVs) possess considerable therapeutic effects in tissue repair and regeneration. Thus, in the present study, we investigated the effects of hucMSC-EVs on primary and secondary osteoporosis and explored the underlying mechanisms. Methods: hucMSCs were isolated and cultured. EVs were obtained from the conditioned medium of hucMSCs and determined by using transmission electron microscopy, dynamic light scattering and Western Blot analyses. The effects of hucMSC-EVs on ovariectomy-induced postmenopausal osteoporosis and tail suspension-induced hindlimb disuse osteoporosis in mouse models were assessed by using microcomputed tomography, biomechanical, histochemical and immunohistochemical, as well as histomorphometric analyses. Proteomic analysis was applied between hucMSC-EVs and hucMSCs to screen the candidate proteins that mediate hucMSC-EVs function. The effects of hucMSC-EVs on osteogenic and adipogenic differentiation of bone marrow mesenchymal stromal cells (BMSCs), and osteoclastogenesis of the macrophage cell line RAW264.7 in vitro were determined by using cytochemical staining and quantitative real-time PCR analysis. Subsequently, the roles of the key protein in hucMSC-EVs-induced regulation on BMSCs and RAW264.7 cells were evaluated. Results: hucMSCs were able to differentiate into osteoblasts, adipocytes or chondrocytes and positively expressed CD29, CD44, CD73 and CD90, but negatively expressed CD34 and CD45. The morphological assessment revealed the typical cup- or sphere-shaped morphology of hucMSC-EVs with diameters predominantly ranging from 60 nm to 150 nm and expressed CD9, CD63, CD81 and TSG101. The systemic administration of hucMSC-EVs prevented bone loss and maintained bone strength in osteoporotic mice by enhancing bone formation, reducing marrow fat accumulation and decreasing bone resorption. Proteomic analysis showed that the potently pro-osteogenic protein, CLEC11A (C-type lectin domain family 11, member A) was very highly enriched in hucMSC-EVs. In addition, hucMSC-EVs enhanced the shift from adipogenic to osteogenic differentiation of BMSCs via delivering CLEC11A in vitro. Moreover, CLEC11A was required for the inhibitory effects of hucMSC-EVs on osteoclast formation. Conclusion: Our results suggest that hucMSC-EVs serve as a critical regulator of bone metabolism by transferring CLEC11A and may represent a potential agent for prevention and treatment of osteoporosis.
Collapse
|
47
|
Therapeutic Effects of Human Urine-Derived Stem Cells in a Rat Model of Cisplatin-Induced Acute Kidney Injury In Vivo and In Vitro. Stem Cells Int 2019; 2019:8035076. [PMID: 31885626 PMCID: PMC6893246 DOI: 10.1155/2019/8035076] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/18/2019] [Accepted: 08/30/2019] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) is an extremely dangerous clinical syndrome with high morbidity and mortality. Stem cell-based therapies have shown great promise for AKI treatment. Urine-derived stem cells (USCs) are a novel cell source in tissue engineering and cell therapy which provide advantages of simple, noninvasive, and low-cost harvest methods, efficient proliferation, and multi-differentiation potential. Here, we described the therapeutic effects of USCs in a rat model of cisplatin-induced AKI as a novel therapy. In vivo, the intravenous administration of USCs alleviated the renal functional damage in AKI rats, for the levels of blood urea nitrogen (BUN) and serum creatinine (SCr) were significantly decreased. The USCs-treated group also exhibited improved histological and ultrastructural changes, promoted proliferation, and inhibited apoptosis in renal tissues. After the USC therapy, the expression levels of proinflammatory cytokines (TNF-α and IL-6) and apoptosis-related proteins (BAX and cleaved caspase-3) were downregulated. In addition, the presence of a few GFP-labeled USCs was confirmed in rat renal tissues. In vitro, rat tubular epithelial (NRK-52E) cells were incubated with cisplatin to induce cell damage and then cocultured with USCs. After coculture with USCs, the cisplatin-induced NRK-52E cells showed higher cell viability and a lower apoptosis ratio than those of the control group, and cell cycle arrest was improved. In conclusion, our results demonstrated that USC therapy significantly improved the renal function and histological damage, inhibited the inflammation and apoptosis processes in the kidney, and promoted tubular epithelial proliferation. Our study exhibited the potential of USCs in the treatment of AKI, representing a new clinical therapeutic strategy.
Collapse
|