1
|
Zununi Vahed S, Hejazian SM, Bakari WN, Landon R, Gueguen V, Meddahi-Pellé A, Anagnostou F, Barzegari A, Pavon-Djavid G. Milking mesenchymal stem cells: Updated protocols for cell lysate, secretome, and exosome extraction, and comparative analysis of their therapeutic potential. Methods 2025; 238:40-60. [PMID: 40058715 DOI: 10.1016/j.ymeth.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/21/2025] Open
Abstract
The potential of the cell lysate, secretome, and extracellular vesicles (EVs) of mesenchymal stem cells (MSCs) to modulate the immune response and promote tissue regeneration has positioned them as a promising option for cell-free therapy. Currently, many clinical trials in stem cells-derived EVs and secretome are in progress various diseases and sometimes the results are failing. The major challenge on this roadmap is the lack of a standard extraction method for exosome, secretome, and lysate. The most optimal method for obtaining the secretome of MSCs for clinical utilization involves a comprehensive approach that includes non-destructive collection methods, time optimization, multiple collection rounds, optimization of culture conditions, and quality control measures. Further research and clinical studies are warranted to validate and refine these methods for safe and effective utilization of the MSC exosome, secretome, and lysate in various clinical applications. To address these challenges, it is imperative to establish a standardized and unified methodology to ensure reliable evaluation of these extractions in clinical trials. This review seeks to outline the pros and cons of methods for the preparation of MSCs-derived exosome, and secretome/lysate, and comparative analysis of their therapeutic potential.
Collapse
Affiliation(s)
| | | | - William Ndjidda Bakari
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France; Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Rebecca Landon
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Virginie Gueguen
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France
| | - Anne Meddahi-Pellé
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France
| | - Fani Anagnostou
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France.
| |
Collapse
|
2
|
Zhang L, Yuan X, Song R, Yuan Z, Zhao Y, Zhang Y. Engineered 3D mesenchymal stem cell aggregates with multifunctional prowess for bone regeneration: Current status and future prospects. J Adv Res 2025:S2090-1232(25)00227-9. [PMID: 40220897 DOI: 10.1016/j.jare.2025.04.008] [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: 11/17/2024] [Revised: 03/29/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Impaired efficacy of in vitro expanded mesenchymal stem cells (MSCs) is a universal and thorny situation, which cast a shadow on further clinical translation of exogenous MSCs. Moreover, the relatively lengthy healing process, host metabolic heterogeneity and the sophisticated cell recognition and crosstalk pose rigorous challenges towards MSC-based bone regeneration strategies. Three-dimensional (3D) cell aggregates facilitate more robust intercellular communications and cell-extracellular matrix (ECM) interactions, providing a better mimicry of microarchitectures and biochemical milieus in vivo, which is conducive for stemness maintenance and downstream bone formation. AIM OF REVIEW This review enunciates the phenotypic features of MSCs in aggregates, which deepens the knowledge of the MSC fate determination in 3D microenvironment. By summarizing current empowerment methods and biomaterial-combined techniques for establishing functionalized MSC aggregates, this review aims to spark innovative and promising solutions for exalting the translational value of MSCs and improve their therapeutic applications in bone tissue repair. KEY SCIENTIFIC CONCEPTS OF REVIEW 3D aggregates optimize regenerative behaviors of in vitro cultured MSCs including cell adhesion, viability, proliferation, pluripotency and immunoregulation capacity, etc. Biomaterials hybridization endows MSC aggregates with tailored mechanical and biological properties, which offers more possibilities in adapting various clinical scenarios.
Collapse
Affiliation(s)
- Linxue Zhang
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China
| | - Xiaojing Yuan
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China
| | - Rui Song
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China
| | - Zuoying Yuan
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, PR China; Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, PR China.
| | - Yuming Zhao
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing 100081, PR China.
| | - Yunfan Zhang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, 22 Zhongguancun South Avenue, Haidian District, Beijing, PR China.
| |
Collapse
|
3
|
Sakhare S, Sanap A, Bhonde R, Behera S, Potdar P, Kheur S, Kharat A. Dialysis and lyophilization of the mesenchymal stromal cell secretome for wound healing. Cytotherapy 2025; 27:544-551. [PMID: 39818643 DOI: 10.1016/j.jcyt.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/26/2024] [Accepted: 12/26/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND AIMS The clinical translation of mesenchymal stromal cell secretome (MSC-S) has been challenging owing to a lack of appropriate methods in downstream processing. Dialysis is an age-old method of protein purification by the exchange of small molecules through a semi-permeable membrane. In this study, we investigated the potential of three forms of umbilical cord-derived MSC secretome (UC-MSC-S)-native (S), dialyzed (DS), and lyophilized (LDS)-for wound healing applications. METHODS AND RESULTS We dialyzed the UC-MSC-S using Slide-A-Lyzer G3 Dialysis Cassettes (20K MWCO) and then lyophilized it to obtain secretome powder. The DS fraction exhibited an 86.01-fold decrease compared with S, whereas LDS showed a 613.71-fold increase in the total protein concentration. Growth factor analysis revealed a significant decrease in the levels of interleukin-6 (IL-6; 54.44-fold), angiopoietin-1 (79.56-fold), angiopoietin-2 (51.76-fold), IL-8 (54.4-fold), platelet endothelial cell adhesion molecule-1 (PECAM-1; 63.25-fold), phosphatidylinositol glycan anchor biosynthesis class F (PIGF; 40.42-fold), vascular endothelial growth factor (VEGF; 39.64-fold), and tumor necrosis factor alpha (TNF-α; 24.62-fold) after dialysis as analyzed by the LEGEND plex multi-analyte flow assay kit on a FACS analyzer. Post-lyophilization, the levels of IL-6 (392.21-fold), angiopoietin-1 (823.04-fold), angiopoietin-2 (397.69-fold), IL-8 (584.83-fold), PECAM-1 (341.28-fold), PIGF (342.85-fold), VEGF (2209.42-fold), and TNF-α (194.4-fold) were enriched in LDS. The highest wound closure (64.07%) and a significant increase in angiogenesis were seen in DLS at the concentration of 1 µg/µL of protein by wound scratch and in ovo yolk sac membrane assay, respectively. CONCLUSIONS Dialysis followed by lyophilization is a simple and cost-effective method to fractionate and enrich the bioactive components of MSC-S without compromising the bioactivity for tailor-made applications.
Collapse
Affiliation(s)
- Swapnali Sakhare
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India; Cellom Biologicals Pvt Ltd, Lab Bay 5, 100, NCL Innovation Park, Dr. Homi Bhaba Road, Pune 411008, India.
| | - Ramesh Bhonde
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India; Cellom Biologicals Pvt Ltd, Lab Bay 5, 100, NCL Innovation Park, Dr. Homi Bhaba Road, Pune 411008, India
| | - Shubhanath Behera
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India
| | - Pranjali Potdar
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India
| | - Supriya Kheur
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India
| | - Avinash Kharat
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India
| |
Collapse
|
4
|
He Y, Lu Y, Li R, Tang Y, Du W, Zhang L, Wu J, Li K, Zhuang W, Lv S, Han Y, Tao B, Deng F, Zhao W, Yu D. CircAars-Engineered ADSCs Facilitate Maxillofacial Bone Defects Repair Via Synergistic Capability of Osteogenic Differentiation, Macrophage Polarization and Angiogenesis. Adv Healthc Mater 2025; 14:e2404501. [PMID: 40035523 PMCID: PMC12004435 DOI: 10.1002/adhm.202404501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/12/2025] [Indexed: 03/05/2025]
Abstract
Adipose-derived stem cells (ADSCs) hold significant promise in bone tissue engineering due to their self-renewal capacity and easy accessibility. However, their limited osteogenic potential remains a critical challenge for clinical application in bone repair. Emerging evidence suggests that circular RNAs (circRNAs) play a key role in regulating stem cell fate and osteogenesis. Despite this, the specific mechanisms by which circRNAs influence ADSCs in the context of bone tissue engineering are largely unexplored. This study introduces a novel strategy utilizing circAars, a specific circRNA, to modify ADSCs, which are then incorporated into gelatin methacryloyl (GelMA) hydrogels for the repair of critical-sized maxillofacial bone defects. The findings reveal that circAars predominantly localizes in the cytoplasm of ADSCs, where it acts as a competitive sponge for miR-128-3p, enhancing the osteogenic differentiation and migration capabilities of ADSCs. Furthermore, circAars-engineered ADSCs facilitate macrophage polarization from the M1 to M2 phenotype and enhance endothelial cell (EC) angiogenic potential through a paracrine mechanism. Additionally, GelMA scaffolds loaded with circAars-engineered ADSCs accelerate the repair of critical-sized maxillofacial bone defects by synergistically promoting osteogenesis, macrophage M2 polarization, and angiogenesis. This approach offers a promising therapeutic strategy for the treatment of critical-sized maxillofacial defects.
Collapse
Affiliation(s)
- Yi He
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Yunyang Lu
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Runze Li
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Yuquan Tang
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
- Guangzhou Liwan District Stomatological HospitalGuangzhou510080P. R. China
| | - Weidong Du
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Lejia Zhang
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Jie Wu
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Kechen Li
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Weijie Zhuang
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Shiyu Lv
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Yaoling Han
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Bailong Tao
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
- Laboratory Research CenterThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016P. R. China
| | - Feilong Deng
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Wei Zhao
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| | - Dongsheng Yu
- Hospital of StomatologyGuanghua School of StomatologyInstitute of Stomatological ResearchSun Yat‐sen UniversityGuangZhou510080P. R. China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510055P. R. China
| |
Collapse
|
5
|
Trigo CM, Rodrigues JS, Camões SP, Solá S, Miranda JP. Mesenchymal stem cell secretome for regenerative medicine: Where do we stand? J Adv Res 2025; 70:103-124. [PMID: 38729561 PMCID: PMC11976416 DOI: 10.1016/j.jare.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-based therapies have yielded beneficial effects in a broad range of preclinical models and clinical trials for human diseases. In the context of MSC transplantation, it is widely recognized that the main mechanism for the regenerative potential of MSCs is not their differentiation, with in vivo data revealing transient and low engraftment rates. Instead, MSCs therapeutic effects are mainly attributed to its secretome, i.e., paracrine factors secreted by these cells, further offering a more attractive and innovative approach due to the effectiveness and safety of a cell-free product. AIM OF REVIEW In this review, we will discuss the potential benefits of MSC-derived secretome in regenerative medicine with particular focus on respiratory, hepatic, and neurological diseases. Both free and vesicular factors of MSC secretome will be detailed. We will also address novel potential strategies capable of improving their healing potential, namely by delivering important regenerative molecules according to specific diseases and tissue needs, as well as non-clinical and clinical studies that allow us to dissect their mechanisms of action. KEY SCIENTIFIC CONCEPTS OF REVIEW MSC-derived secretome includes both soluble and non-soluble factors, organized in extracellular vesicles (EVs). Importantly, besides depending on the cell origin, the characteristics and therapeutic potential of MSC secretome is deeply influenced by external stimuli, highlighting the possibility of optimizing their characteristics through preconditioning approaches. Nevertheless, the clarity around their mechanisms of action remains ambiguous, whereas the need for standardized procedures for the successful translation of those products to the clinics urges.
Collapse
Affiliation(s)
- Catarina M Trigo
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sérgio P Camões
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P Miranda
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
6
|
Giannasi C, Cadelano F, Della Morte E, Baserga C, Mazzucato C, Niada S, Baj A. Unlocking the Therapeutic Potential of Adipose-Derived Stem Cell Secretome in Oral and Maxillofacial Medicine: A Composition-Based Perspective. BIOLOGY 2024; 13:1016. [PMID: 39765683 PMCID: PMC11673083 DOI: 10.3390/biology13121016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 01/03/2025]
Abstract
The adipose-derived stem cell (ADSC) secretome is widely studied for its immunomodulatory and regenerative properties, yet its potential in maxillofacial medicine remains largely underexplored. This review takes a composition-driven approach, beginning with a list of chemokines, cytokines, receptors, and inflammatory and growth factors quantified in the ADSC secretome to infer its potential applications in this medical field. First, a review of the literature confirmed the presence of 107 bioactive factors in the secretome of ADSCs or other types of mesenchymal stem cells. This list was then analyzed using the Search Tool for Retrieval of Interacting Genes/Proteins (STRING) software, revealing 844 enriched biological processes. From these, key processes were categorized into three major clinical application areas: immunoregulation (73 factors), bone regeneration (13 factors), and wound healing and soft tissue regeneration (27 factors), with several factors relevant to more than one area. The most relevant molecules were discussed in the context of existing literature to explore their therapeutic potential based on available evidence. Among these, TGFB1, IL10, and CSF2 have been shown to modulate immune and inflammatory responses, while OPG, IL6, HGF, and TIMP1 contribute to bone regeneration and tissue repair. Although the ADSC secretome holds great promise in oral and maxillofacial medicine, further research is needed to optimize its application and validate its clinical efficacy.
Collapse
Affiliation(s)
- Chiara Giannasi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy; (F.C.); (A.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Francesca Cadelano
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy; (F.C.); (A.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Elena Della Morte
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Camilla Baserga
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Camilla Mazzucato
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Stefania Niada
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| | - Alessandro Baj
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20100 Milan, Italy; (F.C.); (A.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy; (E.D.M.); (C.B.); (C.M.)
| |
Collapse
|
7
|
Ding Y, Song M, Huang R, Chen W. Adipose-mesenchymal stem cell-derived extracellular vesicles enhance angiogenesis and skin wound healing via bFGF-mediated VEGF expression. Cell Tissue Bank 2024; 26:2. [PMID: 39625539 DOI: 10.1007/s10561-024-10150-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/04/2024] [Indexed: 02/22/2025]
Abstract
This study aimed to investigate whether extracellular vesicles (EVs) derived from adipose-derived mesenchymal stem cells (ASCs) promote skin wound healing by delivering basic fibroblast growth factor (bFGF) to enhance vascular endothelial growth factor (VEGF) expression. ASCs were isolated and transfected with either a bFGF knockdown lentivirus (Lv-sh-bFGF) or a control lentivirus (Lv-sh-NC). EVs were extracted from ASCs cultures and characterized by transmission electron microscopy, nanoparticle tracking analysis, and Western blotting for surface markers. EVs were extracted from the conditioned mediums of ASCs and subjected to different treatments. These EVs or control treatments were injected at the wound edges. Wound healing was assessed using histological techniques, including H&E and Masson's trichrome staining to evaluate tissue regeneration, collagen organization, and immunohistochemistry for CD31 to quantify microvessel density. Protein expression of bFGF and VEGF was measured by Western blotting. ASC-derived EVs significantly promoted angiogenesis and improved skin wound healing. EVs encapsulating bFGF enhanced VEGF expression in the wound tissue, while knockdown of bFGF reduced both bFGF and VEGF expression, leading to delayed wound healing. Further knockdown of VEGF partially reversed the pro-angiogenic and wound-healing effects of bFGF-encapsulated EVs. This study demonstrates that ASC-derived EVs promoted skin wound repair by enhancing angiogenesis and accelerating tissue regeneration through the bFGF/VEGF axis. These findings highlight the therapeutic potential of ASCs-derived EVs in improving skin wound healing.
Collapse
Affiliation(s)
- Yonghu Ding
- Department of Orthopedics, The Third People's Hospital Health Care Group of Cixi, 51-139 Zhouxi Road, Zhouxiang Town, Cixi City, Ningbo, 315000, China
| | - Mengsheng Song
- Department of Orthopedics, The Third People's Hospital Health Care Group of Cixi, 51-139 Zhouxi Road, Zhouxiang Town, Cixi City, Ningbo, 315000, China
| | - Rong Huang
- Department of Orthopedics, The Third People's Hospital Health Care Group of Cixi, 51-139 Zhouxi Road, Zhouxiang Town, Cixi City, Ningbo, 315000, China
| | - Weiting Chen
- Department of Orthopedics, The Third People's Hospital Health Care Group of Cixi, 51-139 Zhouxi Road, Zhouxiang Town, Cixi City, Ningbo, 315000, China.
| |
Collapse
|
8
|
Lorentz KL, Marini AX, Bruk LA, Gupta P, Mandal BB, DiLeo MV, Weinbaum JS, Little SR, Vorp DA. Mesenchymal Stem Cell-Conditioned Media-Loaded Microparticles Enhance Acute Patency in Silk-Based Vascular Grafts. Bioengineering (Basel) 2024; 11:947. [PMID: 39329689 PMCID: PMC11428691 DOI: 10.3390/bioengineering11090947] [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: 06/28/2024] [Revised: 08/10/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Coronary artery disease leads to over 360,000 deaths annually in the United States, and off-the-shelf bypass graft options are currently limited and/or have high failure rates. Tissue-engineered vascular grafts (TEVGs) present an attractive option, though the promising mesenchymal stem cell (MSC)-based implants face uncertain regulatory pathways. In this study, "artificial MSCs" (ArtMSCs) were fabricated by encapsulating MSC-conditioned media (CM) in poly(lactic-co-glycolic acid) microparticles. ArtMSCs and control microparticles (Blank-MPs) were incubated over 7 days to assess the release of total protein and the vascular endothelial growth factor (VEGF-A); releasates were also assessed for cytotoxicity and promotion of smooth muscle cell (SMC) proliferation. Each MP type was loaded in previously published "lyogel" silk scaffolds and implanted as interposition grafts in Lewis rats for 1 or 8 weeks. Explanted grafts were assessed for patency and cell content. ArtMSCs had a burst release of protein and VEGF-A. CM increased proliferation in SMCs, but not after encapsulation. TEVG explants after 1 week had significantly higher patency rates with ArtMSCs compared to Blank-MPs, but similar to unseeded lyogel grafts. ArtMSC explants had lower numbers of infiltrating macrophages compared to Blank-MP explants, suggesting a modulation of inflammatory response by the ArtMSCs. TEVG explants after 8 weeks showed no significant difference in patency among the three groups. The ArtMSC explants showed higher numbers of SMCs and endothelial cells within the neotissue layer of the graft compared to Blank-MP explants. In sum, while the ArtMSCs had positive effects acutely, efficacy was lost in the longer term; therefore, further optimization is needed.
Collapse
Affiliation(s)
- Katherine L Lorentz
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ande X Marini
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Liza A Bruk
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Prerak Gupta
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Morgan V DiLeo
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA 15261, USA
- Clinical & Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Justin S Weinbaum
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Steven R Little
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA 15261, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David A Vorp
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA 15261, USA
- Clinical & Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, PA 15261, USA
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Magee Women's Research Institute, Pittsburgh, PA 15213, USA
| |
Collapse
|
9
|
Alquraisy A, Wilar G, Mohammed AFA, El-Rayyes A, Suhandi C, Wathoni N. A Comprehensive Review of Stem Cell Conditioned Media Role for Anti-Aging on Skin. Stem Cells Cloning 2024; 17:5-19. [PMID: 39310304 PMCID: PMC11416772 DOI: 10.2147/sccaa.s480437] [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: 05/29/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
Various studies have been widely conducted on conditioned medium for the development of anti-aging preparations, including the utilization of stem cells, which present a promising alternative solution. This narrative review aims to understand the latest developments in various conditioned medium stem cell applications for anti-aging on the skin. A search of the Scopus database yielded publications of interest. The research focused on articles published without restrictions on the year. After finding 68 articles in the search results, they moved on to the checking phase. Upon comprehensive literature review, 23 articles met the inclusion criteria, while 45 articles were deemed ineligible for participation in this research. The results of the review indicate that conditioned medium from various stem cells has demonstrated success in reducing risk factors for skin aging, as proven in various tests. The successful reduction of the risk of skin aging has been established in vitro, in vivo, and in clinical trials. Given the numerous studies on the progress of exploring and utilizing conditioned medium, it is expected to provide a solution to the problem of skin aging.
Collapse
Affiliation(s)
- Ayatulloh Alquraisy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Padjadjaran, Sumedang, 45363, Indonesia
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, University of Padjadjaran, Sumedang, 45363, Indonesia
| | | | - Ali El-Rayyes
- Department of Chemistry, College of Science, Northern Border University, Arar, Saudi Arabia
| | - Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Padjadjaran, Sumedang, 45363, Indonesia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Padjadjaran, Sumedang, 45363, Indonesia
| |
Collapse
|
10
|
Meshko B, Volatier TLA, Mann J, Kluth MA, Ganss C, Frank MH, Frank NY, Ksander BR, Cursiefen C, Notara M. Anti-Inflammatory and Anti-(Lymph)angiogenic Properties of an ABCB5+ Limbal Mesenchymal Stem Cell Population. Int J Mol Sci 2024; 25:9702. [PMID: 39273646 PMCID: PMC11395824 DOI: 10.3390/ijms25179702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Corneal transparency and avascularity are essential for vision. The avascular cornea transitions into the vascularized conjunctiva at the limbus. Here, we explore a limbal stromal cell sub-population that expresses ABCB5 and has mesenchymal stem cell characteristics. Human primary corneal stromal cells were enriched for ABCB5 by using FACS sorting. ABCB5+ cells expressed the MSC markers CD90, CD73, and CD105. ABCB5+ but not ABCB5- cells from the same donor displayed evidence of pluripotency with a significantly higher colony-forming efficiency and the ability of trilineage differentiation (osteogenic, adipogenic, and chondrogenic). The ABCB5+ cell secretome demonstrated lower levels of the pro-inflammatory protein MIF (macrophage migration inhibitory factor) as well as of the pro-(lymph)angiogenic growth factors VEGFA and VEGFC, which correlated with reduced proliferation of Jurkat cells co-cultured with ABCB5+ cells and decreased proliferation of blood and lymphatic endothelial cells cultured in ABCB5+ cell-conditioned media. These data support the hypothesis that ABCB5+ limbal stromal cells are a putative MSC population with potential anti-inflammatory and anti-(lymph)angiogenic effects. The therapeutic modulation of ABCB5+ limbal stromal cells may prevent cornea neovascularization and inflammation and, if transplanted to other sites in the body, provide similar protective properties to other tissues.
Collapse
Affiliation(s)
- Berbang Meshko
- Department of Ophthalmology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany (T.L.A.V.); (J.M.); (C.C.)
| | - Thomas L. A. Volatier
- Department of Ophthalmology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany (T.L.A.V.); (J.M.); (C.C.)
| | - Johanna Mann
- Department of Ophthalmology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany (T.L.A.V.); (J.M.); (C.C.)
| | - Mark A. Kluth
- RHEACELL GmbH & Co. KG, Im Neuenheimer Feld 517, 69120 Heidelberg, Germany; (M.A.K.); (C.G.)
| | - Christoph Ganss
- RHEACELL GmbH & Co. KG, Im Neuenheimer Feld 517, 69120 Heidelberg, Germany; (M.A.K.); (C.G.)
| | - Markus H. Frank
- Transplant Research Program, Boston Children’s Hospital, Boston, MA 02115, USA;
- Harvard Skin Disease Research Center, Department of Dermatology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA;
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia
| | - Natasha Y. Frank
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA;
- Department of Medicine, VA Boston Healthcare System, Boston, MA 02132, USA
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Bruce R. Ksander
- Massachusetts Eye & Ear Infirmary, Schepens Eye Research Institute, Boston, MA 02114, USA;
| | - Claus Cursiefen
- Department of Ophthalmology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany (T.L.A.V.); (J.M.); (C.C.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50937 Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD) Research Center, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Maria Notara
- Department of Ophthalmology, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50937 Cologne, Germany (T.L.A.V.); (J.M.); (C.C.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50937 Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD) Research Center, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| |
Collapse
|
11
|
Wan X, Ni X, Xie Y, Chen L, Cai B, Lin Q, Ke R, Huang T, Shan X, Wang B. Research progress and application prospect of adipose-derived stem cell secretome in diabetes foot ulcers healing. Stem Cell Res Ther 2024; 15:279. [PMID: 39227906 PMCID: PMC11373215 DOI: 10.1186/s13287-024-03912-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024] Open
Abstract
Diabetic foot ulcers (DFUs) are chronic wounds and one of the most common complications of diabetes, imposing significant physical and mental burdens on patients due to their poor prognosis and treatment efficacy. Adipose-derived stem cells (ADSCs) have been proven to promote wound healing, with studies increasingly attributing these beneficial effects to their paracrine actions. Consequently, research on ADSC secretome as a novel and promising alternative for DFU treatment has been extensively conducted. This article provides a comprehensive review of the mechanisms underlying refractory DFU wounds, the secretome of ADSCs, and its role in promoting wound healing in diabetes foot ulcers. And the review aims to provide reliable evidence for the clinical application of ADSC secretome in the treatment of refractory DFU wounds.
Collapse
Affiliation(s)
- Xiaofen Wan
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xuejun Ni
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yunjia Xie
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Lu Chen
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Beichen Cai
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Qian Lin
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Ruonan Ke
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Tao Huang
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Xiuying Shan
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| | - Biao Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
12
|
Cui Y, He J, Yu Z, Zhou S, Cao D, Jiang T, Fang B, Li G. Adipose-derived stem cells transplantation improves survival and alleviates contraction of skin grafts via promoting macrophages M2 polarization. Skin Res Technol 2024; 30:e13918. [PMID: 39171846 PMCID: PMC11339854 DOI: 10.1111/srt.13918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Full-thickness skin grafts are widely used in plastic and reconstructive surgery. The main limitation of skin grafting is the poor textural durability and associated contracture, which often needs further corrective surgery. Excessive inflammation is the main reason for skin graft contractions, which involve overactivation of myofibroblasts. These problems have prompted the development of new therapeutic approaches, including macrophage polarization modulation and stem cell-based therapies. Currently, adipose-derived stem cells (ASCs) have shown promise in promoting skin grafts survival and regulating macrophage phenotypes. However, the roles of ASCs on macrophages in decreasing skin grafts contraction remain unknown. MATERIALS AND METHODS Rat adipose-derived stem cells (rASCs) were isolated from rat inguinal adipose tissues. Full-thickness skin graft model was constructed on male rats divided into control group and rASCs treatment group. Skin graft was assessed for concentration, elasticity modulus and stiffness. Rat bone marrow-derived macrophages (rBMDMs) were isolated from rat femurs, and subsequent RT-qPCR and coculture assays were carried out to explore the cellular mechanisms. Immunohistochemical and immunofluorescence staining were used to verify mechanisms in vivo. RESULTS In vivo results showed that after injection of ASCs, improved texture, increased survival and inhibited contraction of skin grafts were seen. Vascularization was also improved as illustrated by laser perfusion image and vascular endothelial growth factor (VEGF) concentration. Histological analysis revealed that ASCs injection significantly reduced expression of pro-inflammatory cytokines (TNF-a, IL-1β) and increased expression of anti-inflammatory (IL-10) and pro-healing cytokines (IGF-1). At cellular level, after co-culturing with rASCs, rat bone marrow derived macrophages (rBMDMs) favored M2 polarization even under inflammatory stimulus. CONCLUSION ASCs treatment enhanced vascularization via angiogenic cytokines secretion and alleviated inflammatory environment in skin grafts by driving M2 macrophages polarization, which improved survival and decreased skin grafts contraction. Our work showed that ASCs transplantation can be harnessed to enhance therapeutic efficacy of skin grafting in cutaneous defects treatment.
Collapse
Affiliation(s)
- Yuying Cui
- Department of Plastic and Reconstructive SurgeryThe First Affiliated Hospital of Zhengzhou UniversityHenanChina
| | - Jiahao He
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zheyuan Yu
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Sizheng Zhou
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Dejun Cao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Taoran Jiang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bin Fang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Guangshuai Li
- Department of Plastic and Reconstructive SurgeryThe First Affiliated Hospital of Zhengzhou UniversityHenanChina
| |
Collapse
|
13
|
Vitale F, Cacciottola L, Camboni A, Houeis L, Donnez J, Dolmans MM. Assessing the effect of adipose-tissue-derived stem cell conditioned medium on follicles and stromal cells in bovine ovarian tissue culture. Reprod Biomed Online 2024; 49:103938. [PMID: 38759499 DOI: 10.1016/j.rbmo.2024.103938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/31/2024] [Accepted: 03/05/2024] [Indexed: 05/19/2024]
Abstract
RESEARCH QUESTION Does adipose-tissue-derived stem cell conditioned medium (ASC-CM) supplementation enhance follicle and stromal cell outcomes in vitro? DESIGN Bovine ovaries (n = 8) were sectioned and cultured in vitro for 8 days in two different groups: (i) standard culture (OT Ctrl D8); and (ii) culture with ASC-CM supplementation (OT + CM D8). Half of the culture medium was replaced every other day, and stored to measure the production of oestradiol. Follicle classification was established using haematoxylin and eosin staining. Follicle and stromal cell DNA fragmentation was assessed by TUNEL assays, while growth differentiation factor-9 (GDF-9) staining served as a marker of follicle quality. Additionally, three factors, namely vascular endothelial growth factor (VEGF), interleukin 6 (IL-6) and transforming growth factor beta 1 (TGF-β1), were evaluated in ASC-CM in order to appraise the potential underlying mechanisms of action of ASC. RESULTS The OT + CM D8 group showed a significantly higher proportion of secondary follicles (P = 0.02) compared with the OT Ctrl D8 group. The OT + CM D8 group also demonstrated significantly lower percentages of TUNEL-positive follicles (P = 0.014) and stromal cells (P = 0.001) compared with the OT Ctrl D8 group. Furthermore, follicles in the OT + CM D8 group exhibited a significant increase (P = 0.002) in expression of GDF-9 compared with those in the OT Ctrl D8 group, and oestradiol production was significantly higher (P = 0.04) in the OT + CM D8 group. All studied factors were found to be present in ASC-CM. VEGF and IL-6 were the most widely expressed factors, while TGF-β1 showed the lowest expression. CONCLUSIONS Addition of ASC-CM to culture medium enhances follicle survival, development and oestradiol production, and promotes the viability of stromal cells. VEGF, IL-6 and TGF-β1 could be paracrine mediators underlying the beneficial effects.
Collapse
Affiliation(s)
- Francisco Vitale
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Luciana Cacciottola
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Alessandra Camboni
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Pathology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Lara Houeis
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jacques Donnez
- Société de Recherche pour l'Infertilité, Brussels, Belgium; Professor Em, Université Catholique de Louvain, Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Gynaecology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
14
|
He Y, Tang Y, Zeng B, Chen X, Yuan L, Lu Y, Du W, Li R, Han Y, Deng F, Yu D, Zhao W. Black phosphorus quantum dot-modified ADSCs as a novel therapeutic for periodontitis bone loss coupling of osteogenesis and osteoimmunomodulation. Mater Today Bio 2024; 27:101122. [PMID: 38975241 PMCID: PMC11225909 DOI: 10.1016/j.mtbio.2024.101122] [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: 03/26/2024] [Revised: 05/24/2024] [Accepted: 06/09/2024] [Indexed: 07/09/2024] Open
Abstract
Alveolar bone defect repair remains a persistent clinical challenge for periodontitis treatment. The use of peripheral functional seed cells is a hot topic in periodontitis. Herein, we explored the cellular behaviors and osteogenic ability of adipose-derived mesenchymal stem cells (ADSCs) treated with black phosphorus quantum dots (BPQDs). Additionally, macrophage polarization, osteogenic effects and angiogenesis were investigated through the paracrine pathway regulated by BPQD-modified ADSCs. Our results demonstrated that BPQDs showed good biocompatibility with ADSCs and BPQD-modified ADSCs could improve the bone repair in vivo inflammatory microenvironment by regulating osteogenesis and osteoimmunomodulation. The BPQDs increased the osteogenic differentiation of ADSCs via the Wnt/β-catenin and BMP2/SMAD5/Runx2 signaling pathway. In addition, BPQD-modified ADSCs promoted the osteogenic effect of BMSCs and facilitated the polarization of macrophages from M1 towards M2 phenotype transformation through the paracrine pathway in the periodontitis microenvironment. This strategy provides a novel idea for treatment of alveolar bone defects for periodontitis in the foreseeable future.
Collapse
Affiliation(s)
- Yi He
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuquan Tang
- Zhujiang Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Binghui Zeng
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Xun Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Linyu Yuan
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yunyang Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Weidong Du
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Runze Li
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yaolin Han
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Feilong Deng
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Dongsheng Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Wei Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
15
|
Li Y, Zhao L, Li S, Ruan D, Xiong L, Tang J, Hu M, Wang Y, Huang W, Li L, Zhao Z. Skin-derived precursor conditioned medium alleviated photoaging via early activation of TGF-β/Smad signaling pathway by thrombospondin1: In vitro and in vivo studies. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 253:112873. [PMID: 38412778 DOI: 10.1016/j.jphotobiol.2024.112873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 02/29/2024]
Abstract
Photoaging is one major exogenous factor of skin aging. Efficacy and safety of current anti-photoaging therapies remained to be improved. Our previous studies indicated that skin-derived precursors (SKPs) alleviated photodamage by early activation of TGF-β/Smad signaling pathway via thrombospondin1 (TSP1). However, the research concerning SKP conditioned medium (SKP-CM) has never been reported. In the current study, we aimed to explore the anti-photoaging effects of SKP-CM both in vitro and in vivo, and to elucidate the possible mechanisms. Mouse SKP-CM (mSKP-CM) collection was optimized by a comparative method. The concentration of protein and growth factors in mSKP-CM was detected using BCA protein assay kit and growth factor protein chip. The anti-photoaging effects of mSKP-CM and its regulation of key factors in the TGF-β/Smad signaling pathway were explored using UVA + UVB photoaged mouse fibroblasts (mFBs) and nude mice dorsal skin. The research revealed that mSKP-CM contained significantly higher-concentration of protein and growth factors than mouse mesenchymal stem cell conditioned medium (mDMSC-CM). mSKP-CM alleviated mFBs photoaging by restoring cell viability and relieving senescence and death. ELISA, qRT-PCR, and western blot results implied the potential mechanisms were associated with the early activation of TGF-β/Smad signaling pathway by TSP1. In vivo experiments demonstrated that compared with the topical intradermal mDMSC-CM injection and retinoic acid cream application, the photodamaged mice dorsal skin intradermally injected with mSKP-CM showed significantly better improvement. Consistent with the in vitro results, both western blot and immunohistochemistry results confirmed that protein expression of TSP1, smad2/3, p-smad2/3, TGF-β1, and collagen I increased, and matrix metalloproteinases decreased. In summary, both in vitro and in vivo experiments demonstrated that mSKP-CM alleviated photoaging through an early activation of TGF-β/Smad signaling pathway via TSP1. SKP-CM may serve as a novel and promising cell-free therapeutical approach for anti-photoaging treatment and regenerative medicine.
Collapse
Affiliation(s)
- Yiming Li
- Department of Dermatology, Sichuan Second Hospital of TCM, Chengdu, Sichuan 610041, China; Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Lingyun Zhao
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shiyi Li
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Danhua Ruan
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lidan Xiong
- Center of Cosmetics Evaluation, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jie Tang
- Center of Cosmetics Evaluation, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Meng Hu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yixin Wang
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wen Huang
- Laboratory of Ethnopharmacology, Tissue-orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li Li
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Center of Cosmetics Evaluation, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhiwei Zhao
- Department of Anatomy, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
16
|
Pourhashemi E, Amini A, Ahmadi H, Ahrabi B, Mostafavinia A, Omidi H, Asadi R, Hajihosseintehrani M, Rahmannia M, Fridoni M, Chien S, Bayat M. Photobiomodulation and conditioned medium of adipose-derived stem cells for enhancing wound healing in rats with diabetes: an investigation on the proliferation phase. Lasers Med Sci 2024; 39:46. [PMID: 38270723 DOI: 10.1007/s10103-024-03974-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 01/02/2024] [Indexed: 01/26/2024]
Abstract
This investigation tried to evaluate the combined and solo effects of photobiomodulation (PBM) and conditioned medium derived from human adipose tissue-derived stem cells (h-ASC-CM) on the inflammatory and proliferative phases of an ischemic infected delayed healing wound model (IIDHWM) in rats with type I diabetes mellitus (TIDM). The present investigation consisted of four groups: group 1 served as the control, group 2 treated with h-ASC-CM, group 3 underwent PBM treatment, and group 4 received a combination of h-ASC-CM and PBM. Clinical and laboratory assessments were conducted on days 4 and 8. All treatment groups exhibited significantly higher wound strength than the group 1 (p = 0.000). Groups 4 and 3 demonstrated significantly greater wound strength than group 2 (p = 0.000). Additionally, all therapeutic groups showed reduced methicillin -resistant Staphylococcus aureus (MRSA) in comparison with group 1 (p = 0.000). While inflammatory reactions, including neutrophil and macrophage counts, were significantly lower in all therapeutic groups rather than group 1 on days 4 and 8 (p < 0.01), groups 4 and 3 exhibited superior results compared to group 2 (p < 0.01). Furthermore, proliferative activities, including fibroblast and new vessel counts, as well as the measurement of new epidermal and dermal layers, were significantly increased in all treatment groups on 4 and 8 days after the surgery (p < 0.001). At the same times, groups 4 and 3 displayed significantly higher proliferative activities compared to group 2 (p < 0.001). The treatment groups exhibited significantly higher mast cell counts and degranulation phenotypes in comparison with the group 1 on day 4 (p < 0.05). The treatment groups showed significantly lower mast cell counts and degranulation phenotypes than group 1 on day 8 (p < 0.05).The combined and individual application of h-ASC-CM and PBM remarkably could accelerate the proliferation phase of wound healing in the IIDHWM for TIDM in rats, as indicated by improved MRSA control, wound strength, and stereological evaluation. Furthermore, the combination of h-ASC-CM and PBM demonstrated better outcomes compared to the individual application of either h-ASC-CM or PBM alone.
Collapse
Affiliation(s)
- Erfan Pourhashemi
- School of Medicine, Shahroud University of Medical Sciences, Shahrud, Iran
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Houssein Ahmadi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnaz Ahrabi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atarodalsadat Mostafavinia
- Department of Anatomical Sciences & Cognitive Neuroscience, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Omidi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Robabeh Asadi
- Department of Paramedicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Hajihosseintehrani
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Rahmannia
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadjavad Fridoni
- Department of Biology and Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC of Louisville, Louisville, USA.
| | - Mohammad Bayat
- Department of Biology and Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC of Louisville, Louisville, USA.
| |
Collapse
|
17
|
Xiao W, Shi J. Application of adipose-derived stem cells in ischemic heart disease: theory, potency, and advantage. Front Cardiovasc Med 2024; 11:1324447. [PMID: 38312236 PMCID: PMC10834651 DOI: 10.3389/fcvm.2024.1324447] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) represent an innovative candidate to treat ischemic heart disease (IHD) due to their abundance, renewable sources, minor invasiveness to obtain, and no ethical limitations. Compared with other mesenchymal stem cells, ASCs have demonstrated great advantages, especially in the commercialization of stem cell-based therapy. Mechanistically, ASCs exert a cardioprotective effect not only through differentiation into functional cells but also via robust paracrine of various bioactive factors that promote angiogenesis and immunomodulation. Exosomes from ASCs also play an indispensable role in this process. However, due to the distinct biological functions of ASCs from different origins or donors with varing health statuses (such as aging, diabetes, or atherosclerosis), the heterogeneity of ASCs deserves more attention. This prompts scientists to select optimal donors for clinical applications. In addition, to overcome the primary obstacle of poor retention and low survival after transplantation, a variety of studies have been dedicated to the engineering of ASCs with biomaterials. Besides, clinical trials have confirmed the safety and efficacy of ASCs therapy in the context of heart failure or myocardial infarction. This article reviews the theory, efficacy, and advantages of ASCs-based therapy, the factors affecting ASCs function, heterogeneity, engineering strategies and clinical application of ASCs.
Collapse
Affiliation(s)
| | - Jiahai Shi
- Department of Cardiothoracic Surgery, Affiliated Hospital and Medical School of Nantong University, Nantong, China
| |
Collapse
|
18
|
Widowati W, Faried A, Adam A, Rahmat D, Kusuma HSW, Dewi NSM, Gondokesumo ME, Rizal R, Nainggolan IM, Vosough M. Potential antiaging activity of secretome gel of human Wharton's jelly mesenchymal stem cells (hWJ-MSCs) in UV-induced mice models. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:868-878. [PMID: 38800010 PMCID: PMC11127088 DOI: 10.22038/ijbms.2024.70825.15385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/06/2024] [Indexed: 05/29/2024]
Abstract
Objectives Skin aging is a degenerative process that can be induced by UV irradiation. UV radiation can produce reactive oxidate stress which causes premature aging. This study aims to examine the antiaging potential of secretome gel (SC) from human Wharton Jelly Mesenchymal Stem Cells (hWJ-MSCs) in a UVB-induced mice model. Materials and Methods The secretome was obtained from hWJ-MSCs and made in gel form. Male mice were radiated by UVB for 15 min twice daily for 14 days. The gel was topically applied to the mice's dorsal skin. Two treatments of secretome gel: secretome 1 is applied once and secretome 2 is applied twice daily after UVB radiation. TGF-β1, IL-10, and IL-18 gene expression was determined using RT-PCR. Hematoxylin Eosin staining was used to observe the inflammation and collagen density of skin tissue. An immunohistochemistry assay was used to analyze the protein expression of P53, COL4A1, MMP-2, and MMP-13. The data were statistically analyzed using the ANOVA test followed by the Tukey post hoc test (P<0.05). Results UVB induction caused loss of collagen, increasing inflammation and high expression of aging mediators. SC increased the gene expression of TGF-β1 and IL-10 and decreased IL-18 gene expression. Histopathological tests showed that SG increased collagen density, lowered inflammation, and repaired cell damage in skin tissue. Immunohistochemistry test showed that SC decreased MMP-2, MMP-13, and P53 expression, in contrast, increased COL4A1. Conclusion The secretome gel of hWJ-MSCs showed antiaging activities with potential for preventing and curing skin aging.
Collapse
Affiliation(s)
- Wahyu Widowati
- Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Ahmad Faried
- Department of Neurosurgery, Faculty of Medicine, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, West Java, Indonesia
- Oncology and Stem Cell Working Group, Faculty of Medicine, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, West Java, Indonesia
| | - Achmad Adam
- Department of Neurosurgery, Faculty of Medicine, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, West Java, Indonesia
- Oncology and Stem Cell Working Group, Faculty of Medicine, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, West Java, Indonesia
| | - Deni Rahmat
- Faculty of Pharmacy, Pancasila University, South Jakarta, Indonesia
| | - Hanna Sari Widya Kusuma
- Biomolecular and Biomedical Research Center, Aretha Medika Utama, Bandung, West Java, Indonesia
| | | | | | - Rizal Rizal
- Biomolecular and Biomedical Research Center, Aretha Medika Utama, Bandung, West Java, Indonesia
- Biomedical Engineering Department of Electrical Engineering, Faculty of Engineering University of Indonesia, Jakarta, Indonesia
| | - Ita Margaretha Nainggolan
- Clinical Pathology Department, School of Medicine and Health Sciences, Atma Jaya Catholic University, Jakarta, Indonesia
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Bogor, West Java, Indonesia
| | - Massoud Vosough
- Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska, Institute, Stockholm, Sweden
| |
Collapse
|
19
|
Asadi R, Mostafavinia A, Amini A, Ahmadi H, Ahrabi B, Omidi H, Pourhashemi E, Hajihosseintehrani M, Rezaei F, Mohsenifar Z, Chien S, Bayat M. Acceleration of a delayed healing wound repair model in diabetic rats by additive impacts of photobiomodulation plus conditioned medium of adipose-derived stem cells. J Diabetes Metab Disord 2023; 22:1551-1560. [PMID: 37975122 PMCID: PMC10638220 DOI: 10.1007/s40200-023-01285-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/17/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE This study aimed to investigate the effects of photobiomodulation (PBM) and conditioned medium (CM) derived from human adipose-derived stem cells (h-ASCs), both individually and in combination, on the maturation stage of an ischemic infected delayed healing wound model (IIDHWM) in type I diabetic (TIDM) rats. METHODS The study involved the extraction of h-ASCs from donated fat, assessment of their immunophenotypic markers, cell culture, and extraction and concentration of CM from cultured 1 × 10^6 h-ASCs. TIDM was induced in 24 male adult rats, divided into four groups: control, CM group, PBM group (80 Hz, 0.2 J/cm2, 890 nm), and rats receiving both CM and PBM. Clinical and laboratory evaluations were conducted on days 4, 8, and 16, and euthanasia was performed using CO2 on day 16. Tensiometrical and stereological examinations were carried out using two wound samples from each rat. RESULTS Across all evaluated factors, including wound closure ratio, microbiological, tensiometrical, and stereological parameters, similar patterns were observed. The outcomes of CM + PBM, PBM, and CM treatments were significantly superior in all evaluated parameters compared to the control group (p = 0.000 for all). Both PBM and CM + PBM treatments showed better tensiometrical and stereological results than CM alone (almost all, p = 0.000), and CM + PBM outperformed PBM alone in almost all aspects (p = 0.000). Microbiologically, both CM + PBM and PBM exhibited fewer colony-forming units (CFU) than CM alone (both, p = 0.000). CONCLUSION PBM, CM, and CM + PBM interventions substantially enhanced the maturation stage of the wound healing process in IIDHWM of TIDM rats by mitigating the inflammatory response and reducing CFU count. Moreover, these treatments promoted new tissue formation in the wound bed and improved wound strength. Notably, the combined effects of CM + PBM surpassed the individual effects of CM and PBM. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s40200-023-01285-3.
Collapse
Affiliation(s)
- Robabeh Asadi
- Department of Paramedicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Atarodalsadat Mostafavinia
- Department of Anatomical Sciences and Cognitive Neuroscience, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Houssein Ahmadi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnaz Ahrabi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Omidi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | - Zhaleh Mohsenifar
- Department of Pathology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC of Louisville, Louisville, USA
| | - Mohammad Bayat
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC of Louisville, Louisville, USA
| |
Collapse
|
20
|
Isildar B, Ozkan S, Koyuturk M. Therapeutic Potential of Mesenchymal Stem Cell‐Derived Conditioned Medium for Diabetes Mellitus and Related Complications. ADVANCED THERAPEUTICS 2023; 6. [DOI: 10.1002/adtp.202300216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Indexed: 01/06/2025]
Abstract
AbstractDiabetes mellitus (DM) is one of the most life‐threatening metabolic disorders, with 9% of the global prevalence, and it is estimated to be rising to 12.2% in 2045. Currently, there is no definitive treatment for DM. Although life‐saving, insulin administration to control blood sugar is not a cure for DM and is insufficient to prevent DM‐related complications such as nephropathy, neuropathy, or retinopathy. For this reason, studies are continuing to develop treatments that will provide β‐cell regeneration while suppressing autoimmunity. Mesenchymal stem cells (MSCs) are multipotent stem cells with a high proliferation capacity, immunosuppression, and immunomodulation ability. MSCs have gained therapeutic importance with these properties besides their differentiation ability. The immunosuppressive and immunomodulatory properties of the cells arise from the soluble and insoluble factors they secrete into the extracellular environment. Therefore, the culture medium where these cells grow has therapeutic value and is named conditioned medium (CM). In this context, CM obtained from MSCs can provide a similar therapeutic effect with fewer safety concerns. Furthermore, preconditioning of MSCs can improve the effectiveness of these cells and associated cellular products. So, this review summarizes the recent advances in MSC‐derived CMs and their therapeutic potential for DM and related complications.
Collapse
Affiliation(s)
- Basak Isildar
- Balikesir University Faculty of Medicine Histology and Embryology Department Balikesir 10185 Turkey
| | - Serbay Ozkan
- Izmir Katip Celebi University Faculty of Medicine Histology and Embryology Department Izmir 35620 Turkey
| | - Meral Koyuturk
- Istanbul University‐Cerrahpasa Cerrahpasa Faculty of Medicine Histology and Embryology Department Istanbul 34098 Turkey
| |
Collapse
|
21
|
Sohrabi K, Ahmadi H, Amini A, Ahrabi B, Mostafavinia A, Omidi H, Mirzaei M, Fadaei Fathabady F, Fridoni M, Rahmannia M, Chien S, Bayat M. Promising improvement in infected Wound Healing in Type two Diabetic rats by Combined effects of conditioned medium of human adipose-derived stem cells plus Photobiomodulation. Lab Anim Res 2023; 39:29. [PMID: 37964303 PMCID: PMC10648630 DOI: 10.1186/s42826-023-00178-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND We aimed to examine the accompanying and solo impacts of conditioned medium of human adipose-derived stem cells (h-ASC-COM) and photobiomodulation (PBM) on the maturation stage of an ischemic infected delayed-healing wound model (IIDHWM) of rats with type 2 diabetes (TIIDM). RESULTS Outcomes of the wound closure ratio (WCR) results, tensiometrical microbiological, and stereological assessment followed almost identical patterns. While the outcomes of h-ASC-COM + PBM, PBM only, and h-ASC-COM only regimes were significantly better for all evaluated methods than those of group 1(all, p < 0.001), PBM alone and h-ASC-COM + PBM therapy achieved superior results than h-ASC-COM only (ranged from p = 0.05 to p < 0.001). In terms of tensiometrical and stereological examinations, the results of h-ASC-COM + PBM experienced better results than the PBM only (all, p < 0.001). CONCLUSIONS h-ASC-COM + PBM, PBM, and h-ASC-COM cures expressively accelerated the maturation stage in the wound healing process of IIDHWM with MRSA in TIIDM rats by diminishing the inflammatory reaction, and the microbial flora of MRSA; and increasing wound strength, WCR, number of fibroblasts, and new blood vessels. While the h-ASC-COM + PBM and PBM were more suitable than the effect of h-ASC-COM, the results of h-ASC-COM + PBM were superior to PBM only.
Collapse
Affiliation(s)
- Kaysan Sohrabi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Houssein Ahmadi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnaz Ahrabi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atarodalsadat Mostafavinia
- Department of Anatomical Sciences and Cognitive Neuroscience, School of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Omidi
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mansooreh Mirzaei
- Department of Anatomy, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Fadaei Fathabady
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadjavad Fridoni
- Department of Biology and Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Maryam Rahmannia
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville and Noveratech LLC, Louisville, KY, USA
| | - Mohammad Bayat
- Price Institute of Surgical Research, University of Louisville and Noveratech LLC, Louisville, KY, USA.
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Acuto S, Lo Iacono M, Baiamonte E, Lo Re R, Maggio A, Cavalieri V. An optimized procedure for preparation of conditioned medium from Wharton's jelly mesenchymal stromal cells isolated from umbilical cord. Front Mol Biosci 2023; 10:1273814. [PMID: 37854039 PMCID: PMC10580810 DOI: 10.3389/fmolb.2023.1273814] [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: 08/07/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023] Open
Abstract
Cell-free therapy based on conditioned medium derived from mesenchymal stromal cells (MSCs) has gained attention in the field of protective and regenerative medicine. However, the exact composition and properties of MSC-derived conditioned media can vary greatly depending on multiple parameters, which hamper standardization. In this study, we have optimized a procedure for preparation of conditioned medium starting from efficient isolation, propagation and characterization of MSCs from human umbilical cord, using a culture medium supplemented with human platelet lysate as an alternative source to fetal bovine serum. Our procedure successfully maximizes the yield of viable MSCs that maintain canonical key features. Importantly, under these conditions, the compositional profile and biological effects elicited by the conditioned medium preparations derived from these MSC populations do not depend on donor individuality. Moreover, approximately 120 L of conditioned medium could be obtained from a single umbilical cord, which provides a suitable framework to produce industrial amounts of toxic-free conditioned medium with predictable composition.
Collapse
Affiliation(s)
- Santina Acuto
- Campus of Haematology Franco e Piera Cutino, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Melania Lo Iacono
- Campus of Haematology Franco e Piera Cutino, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Elena Baiamonte
- Campus of Haematology Franco e Piera Cutino, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Rosa Lo Re
- Campus of Haematology Franco e Piera Cutino, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Aurelio Maggio
- Campus of Haematology Franco e Piera Cutino, Villa Sofia-Cervello Hospital, Palermo, Italy
| | - Vincenzo Cavalieri
- Laboratory of Molecular Biology, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STeBiCeF), University of Palermo, Palermo, Italy
| |
Collapse
|
23
|
Ortega-Cuartiella A. Therapeutic Potential of Adipose-Derived Stem Cells and Their Secretome in Reversible Alopecias: A Systematic Review. Int J Trichology 2023; 15:173-182. [PMID: 39170092 PMCID: PMC11335044 DOI: 10.4103/ijt.ijt_3_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 10/19/2021] [Indexed: 08/23/2024] Open
Abstract
Androgenic alopecia (AGA) and alopecia areata (AA) are two highly prevalent conditions, affecting both men and women of a wide range of ages, which strongly impact their quality of life and self-esteem. Both pathologies are deemed to be reversible, although conventional therapies have shown limited scope and efficacy. New therapeutic approaches, focusing on the degenerative changes that take place in the hair follicle, are needed to achieve better outcomes. For instance, adipose-derived stem cells (ADSC), abundant and easy to obtain, hold great potential in follicular regeneration. ADSCs can be isolated as stromal vascular fraction (SVF) by the enzymatic digestion of the lipoaspirate or as nanofat by the mechanical breakdown of adipocytes. In addition, commercial preparations of the conditioned medium of the ADSCs secretome (ADSC-conditionate medium [CM]) have entered the market as an appealing alternative because of their comparatively lower cost and accessibility. A search was conducted, crossing relevant terms, on PubMed Central and Google Scholar. Criteria for inclusion were studies in the past 10 years on humans with AGA or AA, where either SVF, nanofat, or ADSC-CM was tested as the main treatment. Eleven publications qualified: two studied nanofat, three, ADSC-CM, and six, SVF, either individually or in combination with other therapies. Only one randomized controlled trial (RCT) was found and classified as evidence 2b according to the Sackett scale. The rest were case-control studies or case series with small samples and no control, graded as evidence 3b and 4. A meta-analysis could not be conducted due to the heterogenicity of the study designs. Given the evidence obtained, Level D NICE recommendation was established. However, we consider that the positive findings are sufficiently consistent to support the elaboration of further RCTs that share criteria and methods.
Collapse
Affiliation(s)
- Alexis Ortega-Cuartiella
- Ad Astra Clinic® Medical Director and Founder, Cl. Doctor Roux 67, Bajo. Barcelona, Spain, International Society for Stem Cell Applications: Platinum Member, Real Instituto Alfonso XIII: Academician
| |
Collapse
|
24
|
Chouaib B, Haack-Sørensen M, Chaubron F, Cuisinier F, Collart-Dutilleul PY. Towards the Standardization of Mesenchymal Stem Cell Secretome-Derived Product Manufacturing for Tissue Regeneration. Int J Mol Sci 2023; 24:12594. [PMID: 37628774 PMCID: PMC10454619 DOI: 10.3390/ijms241612594] [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: 06/07/2023] [Revised: 07/29/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Mesenchymal stem cell secretome or conditioned medium (MSC-CM) is a combination of biomolecules and growth factors in cell culture growth medium, secreted by mesenchymal stem cells (MSCs), and the starting point of several derived products. MSC-CM and its derivatives could be applied after injuries and could mediate most of the beneficial regenerative effects of MSCs without the possible side effects of using MSCs themselves. However, before the clinical application of these promising biopharmaceuticals, several issues such as manufacturing protocols and quality control must be addressed. This review aims to underline the influence of the procedure for conditioned medium production on the quality of the secretome and its derivatives and highlights the questions considering cell sources and donors, cell expansion, cell passage number and confluency, conditioning period, cell culture medium, microenvironment cues, and secretome-derived product purification. A high degree of variability in MSC secretomes is revealed based on these parameters, confirming the need to standardize and optimize protocols. Understanding how bioprocessing and manufacturing conditions interact to determine the quantity, quality, and profile of MSC-CM is essential to the development of good manufacturing practice (GMP)-compliant procedures suitable for replacing mesenchymal stem cells in regenerative medicine.
Collapse
Affiliation(s)
- Batoul Chouaib
- LBN, University of Montpellier, 34000 Montpellier, France; (B.C.); (F.C.)
- Human Health Department, IRSN, French Institute for Radiological Protection and Nuclear Safety, SERAMED, LRMed, 92262 Fontenay-aux-Roses, France
| | - Mandana Haack-Sørensen
- Cardiology Stem Cell Centre 9302, Rigshospitalet University of Copenhagen, Henrik Harpestrengsvej 4C, 2100 Copenhagen, Denmark
| | - Franck Chaubron
- Institut Clinident BioPharma, Biopôle Clermont-Limagne, 63360 Saint Beauzire, France;
| | - Frederic Cuisinier
- LBN, University of Montpellier, 34000 Montpellier, France; (B.C.); (F.C.)
- Faculty of Dentistry, University of Montpellier, 34000 Montpellier, France
- Service Odontologie, CHU Montpellier, 34000 Montpellier, France
| | - Pierre-Yves Collart-Dutilleul
- LBN, University of Montpellier, 34000 Montpellier, France; (B.C.); (F.C.)
- Faculty of Dentistry, University of Montpellier, 34000 Montpellier, France
- Service Odontologie, CHU Montpellier, 34000 Montpellier, France
| |
Collapse
|
25
|
Muhammad Firdaus FI, Nashihah AK, Mohd Fauzi MB, Manira M, Aminuddin S, Lokanathan Y. Application of Conditioned Medium for In Vitro Modeling and Repair of Respiratory Tissue. APPLIED SCIENCES 2023; 13:5862. [DOI: 10.3390/app13105862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Background: The idea of exploring respiratory therapy in vitro predominantly guided by cell-secreted substances has gained ground in recent years. A conditioned medium (CM) consists of protein milieu that contains a diverse spectrum of cytokines, chemokines, angiogenic agents, and growth factors. This review evaluated the efficacy of using CM collected in an in vitro respiratory epithelial model. Methods: Twenty-six papers were included in this review: twenty-one cellular response studies on respiratory secretome application and five studies involving animal research. Results: The CM produced by differentiated cells from respiratory and non-respiratory systems, such as mesenchymal stem cells (MSC), exhibited the similar overall effect of improving proliferation and regeneration. Not only could differentiated cells from respiratory tissues increase proliferation, migration, and attachment, but the CM was also able to protect the respiratory epithelium against cytotoxicity. Most non-respiratory tissue CM was used as a treatment model to determine the effects of the therapy, while only one study used particle-based CM and reported decreased epithelial cell tight junctions, which harmed the epithelial barrier. Conclusion: As it resolves the challenges related to cell development and wound healing while simultaneously generally reducing the danger of immunological compatibility and tumorigenicity, CM might be a potential regenerative therapy in numerous respiratory illnesses. However, additional research is required to justify using CM in respiratory epithelium clinical practice.
Collapse
Affiliation(s)
- Fairuz Izan Muhammad Firdaus
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Ab. Karim Nashihah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mh. Busra Mohd Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Maarof Manira
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Saim Aminuddin
- Graduate School of Medicine, KPJ Healthcare University College, Kota Seriemas, Nilai 71800, Malaysia
- KPJ Ampang Puteri Specialist Hospital, Ampang 68000, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
26
|
Ra K, Park SC, Lee BC. Female Reproductive Aging and Oxidative Stress: Mesenchymal Stem Cell Conditioned Medium as a Promising Antioxidant. Int J Mol Sci 2023; 24:ijms24055053. [PMID: 36902477 PMCID: PMC10002910 DOI: 10.3390/ijms24055053] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/16/2023] [Accepted: 03/05/2023] [Indexed: 03/09/2023] Open
Abstract
The recent tendency to delay pregnancy has increased the incidence of age-related infertility, as female reproductive competence decreases with aging. Along with aging, a lowered capacity of antioxidant defense causes a loss of normal function in the ovaries and uterus due to oxidative damage. Therefore, advancements have been made in assisted reproduction to resolve infertility caused by reproductive aging and oxidative stress, following an emphasis on their use. The application of mesenchymal stem cells (MSCs) with intensive antioxidative properties has been extensively validated as a regenerative therapy, and proceeding from original cell therapy, the therapeutic effects of stem cell conditioned medium (CM) containing paracrine factors secreted during cell culture have been reported to be as effective as that of direct treatment of source cells. In this review, we summarized the current understanding of female reproductive aging and oxidative stress and present MSC-CM, which could be developed as a promising antioxidant intervention for assisted reproductive technology.
Collapse
Affiliation(s)
- Kihae Ra
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Se Chang Park
- Laboratory of Aquatic Biomedicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
- Correspondence: (S.C.P.); (B.C.L.)
| | - Byeong Chun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
- Correspondence: (S.C.P.); (B.C.L.)
| |
Collapse
|
27
|
Kim W, Kim G. Hybrid cell constructs consisting of bioprinted cell-spheroids. Bioeng Transl Med 2023; 8:e10397. [PMID: 36925682 PMCID: PMC10013803 DOI: 10.1002/btm2.10397] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/18/2022] [Accepted: 08/16/2022] [Indexed: 11/11/2022] Open
Abstract
Bioprinted cell constructs have been investigated for regeneration of various tissues. However, poor cell-cell interactions have limited their utility. Although cell-spheroids offer an alternative for efficient cell-cell interactions, they complicate bioprinting. Here, we introduce a new cell-printing process, fabricating cell-spheroids and cell-loaded constructs together without preparation of cell-spheroids in advance. Cells in mineral oil droplets self-assembled to form cell-spheroids due to the oil-aqueous interaction, exhibiting similar biological functions to the conventionally prepared cell-spheroids. By controlling printing parameters, spheroid diameter and location could be manipulated. To demonstrate the feasibility of this process, we fabricated hybrid cell constructs, consisting of endothelial cell-spheroids and stem cells loaded decellularized extracellular matrix/β-tricalcium phosphate struts for regenerating vascularized bone. The hybrid cell constructs exhibited strong angiogenic/osteogenic activities as a result of increased secretion of signaling molecules and synergistic crosstalk between the cells.
Collapse
Affiliation(s)
- WonJin Kim
- Department of Biomechatronic Engineering, College of Biotechnology and BioengineeringSungkyunkwan University (SKKU)SuwonSouth Korea
| | - GeunHyung Kim
- Department of Biomechatronic Engineering, College of Biotechnology and BioengineeringSungkyunkwan University (SKKU)SuwonSouth Korea
- Biomedical Institute for Convergence at SKKU (BICS)Sungkyunkwan UniversitySuwonSouth Korea
| |
Collapse
|
28
|
Joshi JM, Muttigi MS, Upadhya R, Seetharam RN. An overview of the current advances in the treatment of inflammatory diseases using mesenchymal stromal cell secretome. Immunopharmacol Immunotoxicol 2023:1-11. [PMID: 36786742 DOI: 10.1080/08923973.2023.2180388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
The growing interest in mesenchymal stromal cell (MSC) therapy has been leading to the utilization of its therapeutic properties in a variety of inflammatory diseases. The clinical translation of the related research from bench to bedside is cumbersome due to some obvious limitations of cell therapy. It is evident from the literature that the MSC secretome components mediate their wide range of functions. Cell-free therapy using MSC secretome is being considered as an emerging and promising area of biotherapeutics. The secretome mainly consists of bioactive factors, free nucleic acids, and extracellular vesicles. Constituents of the secretome are greatly influenced by the cell's microenvironment. The broad array of immunomodulatory properties of MSCs are now being employed to target inflammatory diseases. This review focuses on the emerging MSC secretome therapies for various inflammatory diseases. The mechanism of action of the various anti-inflammatory factors is discussed. The potential of MSC secretome as a viable anti-inflammatory therapy is deliberated.
Collapse
Affiliation(s)
- Jahnavy Madhukar Joshi
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Manjunatha S Muttigi
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Raghavendra Upadhya
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Raviraja N Seetharam
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
29
|
Abd El-Baset SA, Mazen NF, Abdul-Maksoud RS, Kattaia AAA. The therapeutic prospect of zinc oxide nanoparticles in experimentally induced diabetic nephropathy. Tissue Barriers 2023; 11:2069966. [PMID: 35504734 PMCID: PMC9870014 DOI: 10.1080/21688370.2022.2069966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Diabetic nephropathy (DN) is the most frequent cause of end-stage renal failure. Zinc oxide nanoparticles (ZnO-NPs) are promising antidiabetic agents. Our aim was to evaluate the prospective efficacy of ZnO-NPs in treating DN in streptozotocin-induced diabetic rats. Rats were randomly dispersed into three sets: control group, DN group and DN + ZnO-NPs group. ZnO-NPs were given at a dose of 10 mg/kg/day by oral gavage for 4 weeks. Urine and blood samples were processed for biochemical analyses. Kidney samples were managed for light and electron microscopy studies. Immune histochemical staining of P53, aquaporin11 (AQP11) and mechanistic target of rapamycin (mTOR) were performed. Gene analyses of nephrin, podocin, beclin-1, LC3 and p62 were done. Administration of ZnO-NPs ameliorated the functional and histopathological alterations of the kidney in a rat model of diabetic nephropathy. ZnO-NPs retained the constancy of the glomerular filtration barrier and restored almost normal renal structure. This was confirmed by upregulation of mRNA expression of podocyte markers (nephrin and podocin) and AQP11 immune histochemical expression in the renal tubules. The beneficial outcomes of ZnO-NPs might be attributed to activation of autophagy through inhibiting mTOR signaling pathway. ZnO-NPs enhanced beclin-1 and LC3 mRNA expressions and reduced p62 mRNA expression. ZnO-NPs also exerted anti-apoptotic potential (evidenced by the decrease in p53 immune expression), anti-inflammatory and anti-oxidant effect [endorsed by suppression of serum cyclooxygenase-2 (COX-2) enzyme activity, tissue nuclear factor kappa beta (NF-κB) level and blood hypoxia-inducible factors (HIF-1α) level]. These results may point the way to an effective therapy of DN.Abbreviations: AQP11 Aquaporin11; BUN: Blood urea nitrogen; COX-2: Cyclooxygenase-2; DAB: 3, 3'-diaminobenzidine; DM: Diabetes mellitus; DN: Diabetic nephropathy; ELISA: Enzyme-linked immunosorbent assay; H&E: Hematoxylin & eosin; HIF-1α: Hypoxia-inducible factors; iNOS: inducible nitric oxide synthase; LC3: Microtubule-associated protein 1 light chain 3; mTOR: Mechanistic target of rapamycin; NF-κB: Nuclear factor kappa beta; NPs: Nanoparticles; PAS: Periodic acid Schiff; PCR: Polymerase chain reaction; PGE2: Prostaglandin E2; ROS: Reactive oxygen species; STZ: Streptozotocin; X ± SEM: Mean ± standard error of means; Zn: Zinc; ZnO-NPs: Zinc oxide nanoparticles.
Collapse
Affiliation(s)
- Samia A. Abd El-Baset
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, ZagazigEgypt
| | - Nehad F. Mazen
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, ZagazigEgypt
| | - Rehab S. Abdul-Maksoud
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, ZagazigEgypt
| | - Asmaa A. A. Kattaia
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, ZagazigEgypt,CONTACT Asmaa A. A. Kattaia ; ; Faculty of Medicine, Zagazig University, Zagazig, Asharquia, Egypt, Postal code: 44519
| |
Collapse
|
30
|
Secretome of human umbilical cord mesenchymal stem cell maintains skin homeostasis by regulating multiple skin physiological function. Cell Tissue Res 2023; 391:111-125. [PMID: 36241740 DOI: 10.1007/s00441-022-03697-8] [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: 04/08/2022] [Accepted: 10/05/2022] [Indexed: 01/18/2023]
Abstract
Skin is the largest organ in the body and the first defense to resist various diseases and external stimuli that easily cause infection and inflammation. Aseptic inflammation, barrier damage, and foreign aid pressure induce the destruction and damage to the skin microenvironment. Subsequently, it destroys the skin's physiological function, leading to the maintenance and circulation of steady-state imbalance and aggravating the process of skin disorders. Our study evaluated the therapeutic potential of the secretome of human umbilical cord mesenchymal stem cells (UC-CM) for dermatological diseases in adult human skin cells, ex vivo skin tissue, and a 3D skin model. Our data suggested several advantages of UC-CM due to (1) their low cytotoxicity and sensitization properties; (2) their anti-inflammatory capacity for treating inflammatory chronic cutaneous diseases; (3) their enhanced capacity of the skin barrier for treating abnormal barrier metabolism; and (4) their positive impact on restoring skin homeostasis due to effective regulation ability of skin physiological function including cell apoptosis, detoxification, and anti-aging. We thus envisage that the possibility of harnessing the therapeutic potential of UC-CM might benefit patients suffering from inflammatory skin disorders such as atopic dermatitis, acne, and psoriasis.
Collapse
|
31
|
Isildar B, Ozkan S, Ercin M, Gezginci-Oktayoglu S, Oncul M, Koyuturk M. 2D and 3D cultured human umbilical cord-derived mesenchymal stem cell-conditioned medium has a dual effect in type 1 diabetes model in rats: immunomodulation and beta-cell regeneration. Inflamm Regen 2022; 42:55. [PMID: 36451229 PMCID: PMC9710085 DOI: 10.1186/s41232-022-00241-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a T-cell-mediated autoimmune disease characterized by the irreversible destruction of insulin-producing β-cells in pancreatic islets. Helper and cytotoxic T-cells and cytokine production, which is impaired by this process, take a synergetic role in β-cell destruction, and hyperglycemia develops due to insulin deficiency in the body. Mesenchymal stem cells (MSCs) appear like an excellent therapeutic tool for autoimmune diseases with pluripotent, regenerative, and immunosuppressive properties. Paracrine factors released from MSCs play a role in immunomodulation by increasing angiogenesis and proliferation and suppressing apoptosis. In this context, the study aims to investigate the therapeutic effects of MSC's secretomes by conditioned medium (CM) obtained from human umbilical cord-derived MSCs cultured in 2-dimensional (2D) and 3-dimensional (3D) environments in the T1D model. METHODS First, MSCs were isolated from the human umbilical cord, and the cells were characterized. Then, two different CMs were prepared by culturing MSCs in 2D and 3D environments. The CM contents were analyzed in terms of total protein, IL-4, IL-10, IL-17, and IFN-λ. In vivo studies were performed in Sprague-Dawley-type rats with an autoimmune T1D model, and twelve doses of CM were administered intraperitoneally for 4 weeks within the framework of a particular treatment model. In order to evaluate immunomodulation, the Treg population was determined in lymphocytes isolated from the spleen after sacrification, and IL-4, IL-10, IL-17, and IFN-λ cytokines were analyzed in serum. Finally, β-cell regeneration was evaluated immunohistochemically by labeling Pdx1, Nkx6.1, and insulin markers, which are critical for the formation of β-cells. RESULTS Total protein and IL-4 levels were higher in 3D-CM compared to 2D-CM. In vivo results showed that CMs induce the Treg population and regulate cytokine release. When the immunohistochemical results were evaluated together, it was determined that CM application significantly increased the rate of β-cells in the islets. This increase was at the highest level in the 3D-CM applied group. CONCLUSION The dual therapeutic effect of MSC-CM on immunomodulation and homeostasis/regeneration of β-cells in the T1D model has been demonstrated. Furthermore, this effect could be improved by using 3D scaffolds for culturing MSCs while preparing CM.
Collapse
Affiliation(s)
- Basak Isildar
- grid.506076.20000 0004 1797 5496Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Serbay Ozkan
- grid.506076.20000 0004 1797 5496Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Merve Ercin
- grid.9601.e0000 0001 2166 6619Department of Biology, Molecular Biology Section, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Selda Gezginci-Oktayoglu
- grid.9601.e0000 0001 2166 6619Department of Biology, Molecular Biology Section, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Mahmut Oncul
- grid.506076.20000 0004 1797 5496Department of Gynecology and Obstetrics, Cerrahpasa Faculty of Medicine, Istanbul University- Cerrahpasa, Istanbul, Turkey
| | - Meral Koyuturk
- grid.506076.20000 0004 1797 5496Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
32
|
The Proangiogenic Potential of Rat Adipose-Derived Stromal Cells with and without Cell-Sheet Induction: A Comparative Study. Stem Cells Int 2022; 2022:2601764. [PMID: 36248258 PMCID: PMC9556194 DOI: 10.1155/2022/2601764] [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: 04/18/2022] [Revised: 07/31/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
A functional vasculature for survival remains a challenge for tissue regeneration, which is indispensable for oxygen and nutrient supply. Utilizing mesenchymal stromal cells (MSCs) to alleviate tissue ischemia and repair dysfunctional or damaged endothelium is a promising strategy. Compared to other populations of MSCs, adipose-derived stromal cells (ASCs) possess a more significant proangiogenic potential and are abundantly available. Cell sheet technology has recently been widely utilized in bone engineering. Compared to conventional methods of seeding seed cell suspension onto biological scaffolds, cell sheet technology prevents cell loss and preserves the extracellular matrix (ECM). Nevertheless, the proangiogenic potential of ASC sheets remains unknown. In this study, rat ASC sheets were constructed, and their macro- and microstructures were examined. In addition, we investigated the effects of ASCs and ASC sheets on the biological properties and angiogenic capacity of endothelial cells (ECs). The results demonstrated that the ASC sheets gradually thickened as the number of cells and ECM increased over time and that the cells were in an active state of secretion. Similar to ASC-CM, the conditioned medium (CM) of ASC sheets could significantly enhance the proliferative capacity of ECs. ASC sheet-CM has significant advantages over ASC-CM in promoting the migration and angiogenesis of ECs, where the exosomes secreted by ASC sheets play an essential role. Therefore, using ASC sheets for therapeutic tissue and organ regeneration angiogenesis may be a valuable strategy.
Collapse
|
33
|
Zhang X, Jin X, Li Y, Xu M, Yao Y, Liu K, Ma C, Zhang Y, Ru J, He Y, Gao J. Macrophage-mediated extracellular matrix remodeling after fat grafting in nude mice. FASEB J 2022; 36:e22550. [PMID: 36098482 DOI: 10.1096/fj.202200037r] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 08/12/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022]
Abstract
Clinical unpredictability and variability following fat grafting remain non-negligible problems due to the unknown mechanism of grafted fat retention. The role of the extracellular matrix (ECM), which renders cells with structural and biochemical support, has been ignored. This study aimed to clarify the ECM remodeling process, related cellular events, and the spatiotemporal relationship between ECM remodeling and adipocyte survival and adipogenesis after fat grafting. Labeled Coleman fat by the matrix-tracing technique was grafted in nude mice. The ECM remodeling process and cellular events were assessed in vivo. The related cytokines were evaluated by qRT-PCR. An in vitro cell migration assay was performed to verify the chemotactic effect of M2-like macrophages on fibroblasts. The results demonstrated that in the periphery, most of the adipocytes of the graft survived or regenerated, and the graft-derived ECM was gradually replaced by the newly-formed ECM. In the central parts, most adipocytes in the grafts died shortly after, and a small part of the graft-derived and newly-formed ECM was expressed with irregular morphology. Adipose ECM remodeling is associated with increased infiltration of macrophages and fibroblasts, as well as up-regulated expression of cytokines in the adipose tissue. To sum up, our results describe the various preservation mode of fat grafts after transplantation and underscore the importance of macrophage-mediated ECM remodeling in graft preservation after fat grafting. The appreciation and manipulation of underlying mechanisms that are operant in this setting stand to explore new therapeutic approaches and improve clinical outcomes of fat grafting.
Collapse
Affiliation(s)
- Xiangdong Zhang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxuan Jin
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yibao Li
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mimi Xu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yao Yao
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kaiyang Liu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chijuan Ma
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuchen Zhang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiangjiang Ru
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yunfan He
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianhua Gao
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
34
|
Saeed Y, Liu X. Mesenchymal stem cells to treat female infertility; future perspective and challenges: A review. Int J Reprod Biomed 2022; 20:709-722. [PMID: 36340664 PMCID: PMC9619121 DOI: 10.18502/ijrm.v20i9.12061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/10/2021] [Accepted: 01/15/2022] [Indexed: 11/19/2022] Open
Abstract
Infertility negatively impacts the overall health and social life of affected individuals and couples. Female infertility is their inability to perceive pregnancy. To date, polycystic ovary syndrome, primary ovarian insufficiency, fallopian tube obstruction, endometriosis, and intrauterine synechiae have been identified as the primary causes of infertility in women. However, despite the mutual efforts of clinicians and research scientists, the development of an effective treatment modality has met little success in combating female infertility. Intriguingly, significant research has demonstrated mesenchymal stem cells as an optimal source for treating infertility disorders. Therefore, here we attempted to capsulize to date available studies to summarize the therapeutic potential of mesenchymal stem cells in combating infertility in women by focusing on the underlying mechanism through which stem cells can reduce the effects of ovarian disorders. Furthermore, we also discussed the preclinical and clinical application of stem cell therapy, their limitation, and the future perspective to minimize these limitations.
Collapse
Affiliation(s)
- Yasmeen Saeed
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan City, Guangdong Province, China
| | - Xiaocui Liu
- Guangdong VitaLife Biotechnology Co., LTD, Foshan, Guangdong, China
| |
Collapse
|
35
|
Fuentes P, Torres MJ, Arancibia R, Aulestia F, Vergara M, Carrión F, Osses N, Altamirano C. Dynamic Culture of Mesenchymal Stromal/Stem Cell Spheroids and Secretion of Paracrine Factors. Front Bioeng Biotechnol 2022; 10:916229. [PMID: 36046670 PMCID: PMC9421039 DOI: 10.3389/fbioe.2022.916229] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, conditioned medium (CM) obtained from the culture of mesenchymal stromal/stem cells (MSCs) has been shown to effectively promote tissue repair and modulate the immune response in vitro and in different animal models, with potential for application in regenerative medicine. Using CM offers multiple advantages over the implantation of MSCs themselves: 1) simpler storage, transport, and preservation requirements, 2) avoidance of the inherent risks of cell transplantation, and 3) potential application as a ready-to-go biologic product. For these reasons, a large amount of MSCs research has focused on the characterization of the obtained CM, including soluble trophic factors and vesicles, preconditioning strategies for enhancing paracrine secretion, such as hypoxia, a three-dimensional (3D) environment, and biochemical stimuli, and potential clinical applications. In vitro preconditioning strategies can increase the viability, proliferation, and paracrine properties of MSCs and therefore improve the therapeutic potential of the cells and their derived products. Specifically, dynamic cultivation conditions, such as fluid flow and 3D aggregate culture, substantially impact cellular behaviour. Increased levels of growth factors and cytokines were observed in 3D cultures of MSC grown on orbital or rotatory shaking platforms, in stirred systems, such as spinner flasks or stirred tank reactors, and in microgravity bioreactors. However, only a few studies have established dynamic culture conditions and protocols for 3D aggregate cultivation of MSCs as a scalable and reproducible strategy for CM production. This review summarizes significant advances into the upstream processing, mainly the dynamic generation and cultivation of MSC aggregates, for de CM manufacture and focuses on the standardization of the soluble factor production.
Collapse
Affiliation(s)
- Paloma Fuentes
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - María José Torres
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Rodrigo Arancibia
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Cellus Biomédica, Parque Tecnológico de León, León, Spain
| | - Francisco Aulestia
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Cellus Biomédica, Parque Tecnológico de León, León, Spain
| | - Mauricio Vergara
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Flavio Carrión
- Cellus Medicina Regenerativa S.A., Santiago, Chile
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - Nelson Osses
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Facultad de Ingeniería, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- CREAS, Centro Regional de Estudios en Alimentos Saludables, Valparaíso, Chile
- *Correspondence: Claudia Altamirano,
| |
Collapse
|
36
|
Yano F, Takeda T, Kurokawa T, Tsubaki T, Chijimatsu R, Inoue K, Tsuji S, Tanaka S, Saito T. Effects of conditioned medium obtained from human adipose-derived stem cells on skin inflammation. Regen Ther 2022; 20:72-77. [PMID: 35509265 PMCID: PMC9034017 DOI: 10.1016/j.reth.2022.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/10/2022] [Accepted: 03/31/2022] [Indexed: 11/25/2022] Open
|
37
|
Maurya DK, Bandekar M, Sandur SK. Soluble factors secreted by human Wharton’s jelly mesenchymal stromal/stem cells exhibit therapeutic radioprotection: A mechanistic study with integrating network biology. World J Stem Cells 2022; 14:347-361. [PMID: 35722198 PMCID: PMC9157603 DOI: 10.4252/wjsc.v14.i5.347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/25/2022] [Accepted: 05/08/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human Wharton’s jelly-derived mesenchymal stromal/stem cells (hWJ-MSCs) have gained considerable attention in their applications in cell-based therapy due to several advantages offered by them. Recently, we reported that hWJ-MSCs and their conditioned medium have significant therapeutic radioprotective potential. This finding raised an obvious question to identify unique features of hWJ-MSCs over other sources of stem cells for a better understanding of its radioprotective mechanism.
AIM To understand the radioprotective mechanism of soluble factors secreted by hWJ-MSCs and identification of their unique genes.
METHODS Propidium iodide staining, endogenous spleen colony-forming assay, and survival study were carried out for radioprotection studies. Homeostasis-driven proliferation assay was performed for in vivo lymphocyte proliferation. Analysis of RNAseq data was performed to find the unique genes of WJ-MSCs by comparing them with bone marrow mesenchymal stem cells, embryonic stem cells, and human fibroblasts. Gene enrichment analysis and protein-protein interaction network were used for pathway analysis.
RESULTS Co-culture of irradiated murine splenic lymphocytes with WJ-MSCs offered significant radioprotection to lymphocytes. WJ-MSC transplantation increased the homeostasis-driven proliferation of the lymphocytes. Neutralization of WJ-MSC conditioned medium with granulocyte-colony stimulating factor antibody abolished therapeutic radioprotection. Transcriptome analysis showed that WJ-MSCs share several common genes with bone marrow MSCs and embryonic stem cells and express high levels of unique genes such as interleukin (IL)1-α, IL1-β, IL-6, CXCL3, CXCL5, CXCL8, CXCL2, CCL2, FLT-1, and IL-33. It was also observed that WJ-MSCs preferentially modulate several cellular pathways and processes that handle the repair and regeneration of damaged tissues compared to stem cells from other sources. Cytokine-based network analysis showed that most of the radiosensitive tissues have a more complex network for the elevated cytokines.
CONCLUSION Systemic infusion of WJ-MSC conditioned media will have significant potential for treating accidental radiation exposed victims.
Collapse
Affiliation(s)
- Dharmendra Kumar Maurya
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Mayuri Bandekar
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India
- University of Mumbai, Kalina, Mumbai 400098, India
| | - Santosh Kumar Sandur
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
38
|
Wang Y, Liu J, Yu B, Jin Y, Li J, Ma X, Yu J, Niu J, Liang X. Umbilical cord-derived mesenchymal stem cell conditioned medium reverses neuronal oxidative injury by inhibition of TRPM2 activation and the JNK signaling pathway. Mol Biol Rep 2022; 49:7337-7345. [PMID: 35585377 PMCID: PMC9304044 DOI: 10.1007/s11033-022-07524-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022]
Abstract
Background The mechanism by which MSC-CM protects neuronal cells against ischemic injury remains to be elucidated. In this study, we aimed to clarify the protective effect of umbilical cord-derived mesenchymal stem cell conditioned medium (UC-MSC-CM) on neuronal oxidative injury and its potential mechanism. Methods and Results Neuronal oxidative damage was mimicked by H2O2 treatment of the HT22 cell line. The numbers of cleaved-Caspase-3-positive cells and protein expression of Caspase-9 induced by H2O2 treatment were decreased by UC-MSC-CM treatment. Furthermore, SOD protein expression was increased in the MSC-CM group compared with that in the H2O2 group. The H2O2-induced TRPM2-like currents in HT22 cells were attenuated by MSC-CM treatment. In addition, H2O2 treatment downregulated the expression of p-JNK protein in HT22 cells, and this the downward trend was reversed by incubation with MSC-CM. Conclusions UC-MSC-CM protects neurons against oxidative injury, possibly by inhibiting activation of TRPM2 and the JNK signaling pathway.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China
| | - Jiaxin Liu
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China
| | - Baocong Yu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 750004, Yinchuan, China
| | - Yiran Jin
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China
| | - Jiahui Li
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China
| | - Xiaona Ma
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China
| | - Jianqiang Yu
- School of Pharmacology, Ningxia Medical University, 750004, Yinchuan, China.
| | - Jianguo Niu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, 750004, Yinchuan, China.
| | - Xueyun Liang
- Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, 750001, Yinchuan, China.
| |
Collapse
|
39
|
Wang D, Wen JY, Wu D, Ying ZY, Wen ZM, Peng HQ, Geng C, Feng YB, Sui ZG, Lv HY, Wu J, Xu B. LPS-pretreated MSC-conditioned medium optimized with 10-kDa filter attenuates the injury of H9c2 cardiomyocytes in a model of hypoxia/reoxygenation. Can J Physiol Pharmacol 2022; 100:651-664. [PMID: 35533248 DOI: 10.1139/cjpp-2021-0745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mesenchymal stem cell-derived conditioned medium (MSC-CM) improves cardiac function, which is partly attributed to released paracrine factors. Since such cardioprotection is moderate and transient, it's essential to optimize MSC-CM effective components to alleviate myocardial injury. To optimize MSC-CM, MSCs were treated with or without lipopolysaccharides (LPSs) for 48 h (serum-free), and the supernatant was collected. Then, LPS-CM (MSC stimulated by LPS) was further treated with LPS remover (LPS Re-CM) or was concentrated with a 10-kDa cutoff filter (10 kDa-CM). ELISA showed that all pretreatments increased levels of VEGF, HGF, and IGF except LPS remover; 10 kDa-CM was superior to other-CM. CCK-8 displayed that viability of injured H9c2 cells enhanced with the increase of MSC-CM concentration. We also found 10 kDa-CM significantly alleviated H9c2 hypoxia/reoxygenation (H/R) injury, as evidenced by increased Bcl-2/Bax ratio, decreased the levels of LDH and cTn. TEM, TUNEL, and H&E staining confirmed 10 kDa-CM inhibited H/R-induced H9c2 morphological changes. Proteomic analysis identified 41 differentially expressed proteins in 10 kDa-CM, among which anti-inflammation, pro-angiogenesis, and anti-apoptosis were related to cardiac protection. This study indicates that 10 kDa-CM protects H9c2 cardiomyocytes from H/R injury by preserving most of the protective factors, such as VEGF, HGF, and IGF, in MSC-CM.
Collapse
Affiliation(s)
- Dan Wang
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China.,Ordos Central Hospital, 586048, Department of Pharmacy, Ordos, Inner Mongolia, China;
| | - Jing-Yi Wen
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China;
| | - Di Wu
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China;
| | - Zi-Yue Ying
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China;
| | - Zhi-Min Wen
- The Second Affiliated Hospital of Dalian Medical University, Department of Clinical Laboratory, Dalian, Liaoning, China;
| | - Hui-Qian Peng
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China;
| | - Cong Geng
- The Second Affiliated Hospital of Dalian Medical University, Department of Clinical Laboratory, Dalian, Liaoning, China;
| | - Yuan-Bo Feng
- KU Leuven University Hospitals Leuven, 60182, Leuven, Flanders, Belgium;
| | - Zhi-Gang Sui
- Chinese Academy of Science, Dalian, Liaoning, China;
| | - Hui-Yi Lv
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China;
| | - Jun Wu
- The Second Affiliated Hospital of Dalian Medical University, Department of Echocardiography, Dalian, Liaoning, China;
| | - Bing Xu
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China, 116023;
| |
Collapse
|
40
|
Zhang Y, Zhang X, Jin X, Zhang P, Liu K, Yao Y, Ru J, Li Y, Xu M, Lu F, He Y, Gao J. Adipose Collagen Fragment: A Novel Adipose-Derived Extracellular Matrix Concentrate for Skin Filling. Aesthet Surg J 2022; 42:NP337-NP350. [PMID: 36413201 DOI: 10.1093/asj/sjab386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Skin filler is an option for treating skin aging and wrinkles; however, currently used fillers are limited by poor biocompatibility, rapid degradation, and possible hypersensitivity reactions. Autologous adipose tissue-derived products have been recognized as promising options for skin rejuvenation. OBJECTIVES This study aimed to develop a novel adipose-derived product for skin filling. METHODS Adipose collagen fragment (ACF) was prepared through pulverization, filtration, and centrifugation. The macrography, structure, types of collagen, and cell viability of ACF were evaluated by immunostaining, western blotting, and cell culture assays. ACF, nanofat, and phosphate-buffered saline (9 spots/side, 0.01 mL/spot) were intradermally injected in the dorsal skin of 36 female BALB/c nude mice; the skin filling capacity and the collagen remodeling process were then investigated. Twenty-one female patients with fine rhytides in the infraorbital areas were enrolled and received clinical applications of ACF treatment. Therapeutic effects and patients' satisfaction scores were recorded. RESULTS The mean [standard deviation] yield of ACF from 50 mL of Coleman fat was 4.91 [0.25] mL. ACF contained nonviable cells and high levels of collagen I, collagen IV, and laminin. Fibroblasts and procollagen significantly increased in ACF and ACF-treated dermis (P < 0.05). Overall, 85.7% of patients were satisfied with the therapy results, and no infections, injection site nodules, or other unwanted side effects were observed. CONCLUSIONS ACF significantly improved dermal thickness and collagen synthesis and may serve as a potential autologous skin filler.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Xiangdong Zhang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Xiaoxuan Jin
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Pan Zhang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Kaiyang Liu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Yao Yao
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Jiangjiang Ru
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Yibao Li
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Mimi Xu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Yunfan He
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| | - Jianhua Gao
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, China
| |
Collapse
|
41
|
Später T, Assunção M, Lit KK, Gong G, Wang X, Chen YY, Rao Y, Li Y, Yiu CHK, Laschke MW, Menger MD, Wang D, Tuan RS, Khoo KH, Raghunath M, Guo J, Blocki A. Engineering microparticles based on solidified stem cell secretome with an augmented pro-angiogenic factor portfolio for therapeutic angiogenesis. Bioact Mater 2022; 17:526-541. [PMID: 35846945 PMCID: PMC9270501 DOI: 10.1016/j.bioactmat.2022.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/22/2022] [Accepted: 03/07/2022] [Indexed: 11/17/2022] Open
Abstract
Tissue (re)vascularization strategies face various challenges, as therapeutic cells do not survive long enough in situ, while the administration of pro-angiogenic factors is hampered by fast clearance and insufficient ability to emulate complex spatiotemporal signaling. Here, we propose to address these limitations by engineering a functional biomaterial capable of capturing and concentrating the pro-angiogenic activities of mesenchymal stem cells (MSCs). In particular, dextran sulfate, a high molecular weight sulfated glucose polymer, supplemented to MSC cultures, interacts with MSC-derived extracellular matrix (ECM) components and facilitates their co-assembly and accumulation in the pericellular space. Upon decellularization, the resulting dextran sulfate-ECM hybrid material can be processed into MIcroparticles of SOlidified Secretome (MIPSOS). The insoluble format of MIPSOS protects protein components from degradation, while facilitating their sustained release. Proteomic analysis demonstrates that MIPSOS are highly enriched in pro-angiogenic factors, resulting in an enhanced pro-angiogenic bioactivity when compared to naïve MSC-derived ECM (cECM). Consequently, intravital microscopy of full-thickness skin wounds treated with MIPSOS demonstrates accelerated revascularization and healing, far superior to the therapeutic potential of cECM. Hence, the microparticle-based solidified stem cell secretome provides a promising platform to address major limitations of current therapeutic angiogenesis approaches.
Dextran sulfate assembles with mesenchymal stem cell secretome. As a result, microparticles of solidified stem cell secretome (MIPSOS) are formed. The insoluble MIPSOS format protects proteins from premature degradation. MIPSOS are enriched in pro-angiogenic factors and exhibit gradual release kinetics. MIPSOS demonstrate superior pro-angiogenic properties and thus therapeutic potential.
Collapse
Affiliation(s)
- Thomas Später
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Saar, Germany
| | - Marisa Assunção
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Kwok Keung Lit
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Guidong Gong
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- Bioproducts Institute, Departments of Chemical and Biological Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Xiaoling Wang
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yi-Yun Chen
- Academia Sinica Common Mass Spectrometry Facilities for Proteomics and Protein Modification Analysis, and Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, China
| | - Ying Rao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Yucong Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Shun Hing Institute of Advanced Engineering (SHIAE), Faculty of Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Chi Him Kendrick Yiu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Matthias W. Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Saar, Germany
| | - Michael D. Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Saar, Germany
| | - Dan Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong Special Administrative Region of China
| | - Rocky S. Tuan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Kay-Hooi Khoo
- Academia Sinica Common Mass Spectrometry Facilities for Proteomics and Protein Modification Analysis, and Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, China
| | - Michael Raghunath
- Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Wädenswil, Switzerland
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- Bioproducts Institute, Departments of Chemical and Biological Engineering, The University of British Columbia, Vancouver, BC, Canada
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- Corresponding author. BMI Center for Biomass Materials and Nanointerfaces, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China.
| | - Anna Blocki
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong Special Administrative Region of China
- Corresponding author. School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong Special Administrative Region of China.
| |
Collapse
|
42
|
Arifka M, Wilar G, Elamin KM, Wathoni N. Polymeric Hydrogels as Mesenchymal Stem Cell Secretome Delivery System in Biomedical Applications. Polymers (Basel) 2022; 14:polym14061218. [PMID: 35335547 PMCID: PMC8955913 DOI: 10.3390/polym14061218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 01/27/2023] Open
Abstract
Secretomes of mesenchymal stem cells (MSCs) have been successfully studied in preclinical models for several biomedical applications, including tissue engineering, drug delivery, and cancer therapy. Hydrogels are known to imitate a three-dimensional extracellular matrix to offer a friendly environment for stem cells; therefore, hydrogels can be used as scaffolds for tissue construction, to control the distribution of bioactive compounds in tissues, and as a secretome-producing MSC culture media. The administration of a polymeric hydrogel-based MSC secretome has been shown to overcome the fast clearance of the target tissue. In vitro studies confirm the bioactivity of the secretome encapsulated in the gel, allowing for a controlled and sustained release process. The findings reveal that the feasibility of polymeric hydrogels as MSC -secretome delivery systems had a positive influence on the pace of tissue and organ regeneration, as well as an enhanced secretome production. In this review, we discuss the widely used polymeric hydrogels and their advantages as MSC secretome delivery systems in biomedical applications.
Collapse
Affiliation(s)
- Mia Arifka
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia;
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia;
| | - Khaled M. Elamin
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan;
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia;
- Correspondence: ; Tel.: +62-22-842-888-888
| |
Collapse
|
43
|
Zhang Q, Liu X, Piao C, Jiao Z, Ma Y, Wang Y, Liu T, Xu J, Wang H. Effect of conditioned medium from adipose derived mesenchymal stem cells on endoplasmic reticulum stress and lipid metabolism after hepatic ischemia reperfusion injury and hepatectomy in swine. Life Sci 2022; 289:120212. [PMID: 34896163 DOI: 10.1016/j.lfs.2021.120212] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
AIMS Hepatic ischemia reperfusion injury (HIRI) is associated with liver failure after liver transplantation and hepatectomy. Thus, this study aims to explore the effect of conditioned medium from adipose derived stem cells (ADSC-CM) on endoplasmic reticulum stress (ERS) and lipid metabolism after HIRI combined with hepatectomy in miniature pigs. MAIN METHODS A model of HIRI combined with hepatectomy in miniature pigs was established. The expression of ERS-related proteins and lipid metabolism related genes, as well as triglyceride (TG), total cholesterol (TC), high density lipoprotein (HDL), very low density lipoprotein (VLDL) and acetyl-CoA carboxylase 1 (ACC1) level were measured in liver tissues. KEY FINDINGS Both ADSCs and ADSC-CM could improve the damage in the ultrastructure of hepatocytes. ADSC-CM significantly decreased the protein expression of GRP78, ATF6, XBP1, p-eIF2α, ATF4, p-JNK and CHOP. Oil red O staining revealed that the degree of hepatocyte steatosis was also significantly reduced after treatment with ADSC-CM. In addition, ADSC-CM remarkably decreased TG, TC, HDL and ACC1 level in liver tissues, while enhanced VLDL content. Finally, SREBP1, SCAP, FASN, ACC1, HMGCR and HMGCS1 mRNA expression was also markedly downregulated in liver tissues. SIGNIFICANCE Injection of ADSC-CM into the hepatic parenchymal could represent a novel cell-free therapeutic approach to improve HIRI combined with hepatectomy injury. The inhibition of ERS and the improvement of lipid metabolism in the hepatocytes might be a potential mechanism used by ADSC-CM to prevent liver injury from HIRI combined with hepatectomy.
Collapse
Affiliation(s)
- Qianzhen Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, PR China
| | - Xiaoning Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Chenxi Piao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Zhihui Jiao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, PR China
| | - Yajun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yue Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Tao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jiayuan Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hongbin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
44
|
Cui L, Saeed Y, Li H, Yang J. Regenerative medicine and traumatic brain injury: from stem cell to cell-free therapeutic strategies. Regen Med 2021; 17:37-53. [PMID: 34905963 DOI: 10.2217/rme-2021-0069] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious health concern, yet there is a lack of standardized treatment to combat its long-lasting effects. The objective of the present study was to provide an overview of the limitation of conventional stem-cell therapy in the treatment of TBI and to discuss the application of novel acellular therapies and their advanced strategies to enhance the efficacy of stem cells derived therapies in the light of published study data. Moreover, we also discussed the factor to optimize the therapeutic efficiency of stem cell-derived acellular therapy by overcoming the challenges for its clinical translation. Hence, we concluded that acellular therapy possesses the potential to bring a breakthrough in the field of regenerative medicine to treat TBI.
Collapse
Affiliation(s)
- Lianxu Cui
- Department of Neurosurgery, The First People's Hospital of Foshan, 81 North Lingnan Road, Foshan, Guangdong, 528300, PR China
| | - Yasmeen Saeed
- Guangdong VitaLife Biotechnology Co., LTD, 61 Xiannan Road, Nanhai District, Foshan, Guangdong, 528200, PR China
| | - Haomin Li
- Department of Neurosurgery, The First People's Hospital of Foshan, 81 North Lingnan Road, Foshan, Guangdong, 528300, PR China
| | - Jingli Yang
- School of medicine, Foshan University, 18 Jiangwan Road, Foshan, Guangdong, 528000, PR China
| |
Collapse
|
45
|
Amiri F, Kiani AA, Bahadori M, Roudkenar MH. Co-culture of mesenchymal stem cell spheres with hematopoietic stem cells under hypoxia: a cost-effective method to maintain self-renewal and homing marker expression. Mol Biol Rep 2021; 49:931-941. [PMID: 34741711 DOI: 10.1007/s11033-021-06912-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/29/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Hematopoietic stem cell (HSC) transplantation is considered a possible treatment option capable of curing various diseases. The aim of this study was the co-culturing of mesenchymal stem cell (MSC) spheres with HSCs under hypoxic condition to enhance the proliferation, self-renewal, stemness, and homing capacities of HSCs. METHODS AND RESULTS HSCs were expanded after being subjected to different conditions including cytokines without feeder (Cyto), co-culturing with adherent MSCs (MSC), co-culturing with adherent MSCs + hypoxia (MSC + Hyp), co-culturing with MSCs spheres (Sph-MSC), co-culturing with MSCs spheres + hypoxia (Sph-MSC + Hyp), co-culturing with MSC spheres + cytokines (Sph-MSC + Cyto). After 10 days, total nucleated cell (TNC) and CD34+/CD38- cell counts, colony-forming unit assay (CFU), long-term culture initiating cell (LTC-IC), the expression of endothelial protein C receptor (EPCR), nucleostemin (NS), nuclear factor I/X (Nfix) CXCR4, and VLA-4 were evaluated. The TNC, CD34+/CD38- cell count, CFU, and LTC-IC were higher in the Sph-MSC + Hyp and Sph-MSC + Cyto groups as compared with those of the MSC + Hyp group (P < 0.001). The expanded HSCs co-cultured with MSC spheres in combination with hypoxia expressed more EPCR, CXCR4, VLA-4, NS, and Nfix mRNA. The protein expression was also more up-regulated in the Sph-MSC + Cyto and Sph-MSC + Hyp groups. CONCLUSION Co-culturing HSCs with MSC spheres under hypoxic condition not only leads to higher cellular yield but also increases the expression of self-renewal and homing genes. Therefore, we suggest this approach as a simple and non-expensive strategy that might improve the transplantation efficiency of HSCs.
Collapse
Affiliation(s)
- Fatemeh Amiri
- Department of Medical Laboratory Sciences, School of Para Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Asghar Kiani
- Department of Hematology and Blood Transfusion, Lorestan University of Medical Sciences, Khorramabad, Lorestan, Iran
| | - Marzie Bahadori
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mehryar Habibi Roudkenar
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran. .,Burn and Regenerative Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
46
|
Merkhan MM, Shephard MT, Forsyth NR. Physoxia alters human mesenchymal stem cell secretome. J Tissue Eng 2021; 12:20417314211056132. [PMID: 34733464 PMCID: PMC8558798 DOI: 10.1177/20417314211056132] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
The human mesenchymal stem cell (hMSC) secretome has pleiotropic effects which underpin their therapeutic potential. hMSC serum-free conditioned media (SFCM) has been determined to contain a variety of cytokines with roles in regeneration and suppression of inflammation. Physiological oxygen (physoxia) has been demonstrated to impact upon a number of facets of hMSC biology and we hypothesized that the secretome would be similarly modified. We tested a range of oxygen conditions; 21% O2 (air oxygen (AO)), 2% O2 (intermittent hypoxia (IH)) and 2% O2 Workstation (physoxia (P)) to evaluate their effect on hMSC secretome profiles. Total protein content of secretome was upregulated in IH and P (>3 fold vs AO) and IH (>1 fold vs P). Focused cytokine profiling indicated global upregulation in IH of all 31 biomolecules tested in comparison to AO and P with basic-nerve growth factor (bNGF) and granulocyte colony-stimulating factor (GCSF) (>3 fold vs AO) and bNGF and Rantes (>3 fold vs P) of note. Similarly, upregulation of interferon gamma-induced protein 10 (IP10) was noted in P (>3 fold vs AO). Interleukin-2 (IL2) and Rantes (in AO and P) and adiponectin, IL17a, and epidermal growth factor (EGF) (in AO only) were entirely absent or below detection limits. Quantitative analysis validated the pattern of IH-induced upregulation in vascular endothelial growth factor (VEGF), placental growth factor-1 (PIGF1), Tumor necrosis factor alpha (TNFa), IL2, IL4, and IL10 when compared to AO and P. In summary, modulation of environmental oxygen alters both secretome concentration and composition. This consideration will likely impact on delivering improved mechanistic understanding and potency effects of hMSC-based therapeutics.
Collapse
Affiliation(s)
- Marwan M Merkhan
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK.,College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Matthew T Shephard
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK
| | - Nicholas R Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK
| |
Collapse
|
47
|
Zocchi ML, Facchin F, Pagani A, Bonino C, Sbarbati A, Conti G, Vindigni V, Bassetto F. New perspectives in regenerative medicine and surgery: the bioactive composite therapies (BACTs). EUROPEAN JOURNAL OF PLASTIC SURGERY 2021; 45:1-25. [PMID: 34728900 PMCID: PMC8554210 DOI: 10.1007/s00238-021-01874-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/06/2021] [Indexed: 12/26/2022]
Abstract
Regenerative medicine and surgery is a rapidly expanding branch of translational research in tissue engineering, cellular and molecular biology. To date, the methods to improve cell intake, survival, and isolation need to comply with a complex and still unclear regulatory frame, becoming everyday more restrictive and often limiting the effectiveness and outcome of the therapeutic choices. Thus, the authors developed a novel 360° regenerative strategy based on the synergic action of several new components called the bioactive composite therapies (BACTs) to improve grafted cells intake, and survival in total compliance with the legal and ethical limits of the current regulatory frame. The rationale at the origin of this new technology is based on the evidence that cells need supportive substrate to survive in vitro and this observation, applying the concept of translational medicine, is true also in vivo. Bioactive composite mixtures (BACMs) are tailor-made bioactive mixtures containing several bioactive components that support cells' survival and induce a regenerative response in vivo by stimulating the recipient site to act as an in situ real bioreactor. Many different tissues have been used in the past for the isolation of cells, molecules, and growth factors, but the adipose tissue and its stromal vascular fraction (SVF) remains the most valuable, abundant, safe, and reliable source of regenerative components and particularly of adipose-derived stems cells (ADSCs). The role of plastic surgeons as the historical experts in all the most advanced techniques for harvesting, manipulating, and grafting adipose tissue is fundamental in this constant process of expansion of regenerative procedures. In this article, we analyze the main causes of cell death and the strategies for preventing it, and we present all the technical steps for preparing the main components of BACMs and the different mixing modalities to obtain the most efficient regenerative action on different clinical and pathological conditions. The second section of this work is dedicated to the logical and sequential evolution from simple bioactive composite grafts (BACGs) that distinguished our initial approach to regenerative medicine, to BACTs where many other fundamental technical steps are analyzed and integrated for supporting and enhancing the most efficient regenerative activity. Level of Evidence: Not gradable.
Collapse
Affiliation(s)
- Michele L Zocchi
- Plastic and Reconstructive Surgery Unit, University of Padua, Padua, Italy.,Remix Institute for Regenerative Surgery, Turin, Italy
| | - Federico Facchin
- Plastic and Reconstructive Surgery Unit, University of Padua, Padua, Italy
| | - Andrea Pagani
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich, Germany
| | - Claudia Bonino
- Department of Rheumatology and Immune Diseases, Humanitas Gradenigo Hospital, Turin, Italy
| | - Andrea Sbarbati
- Institute of Human Anatomy, University of Verona, Verona, Italy
| | - Giamaica Conti
- Institute of Human Anatomy, University of Verona, Verona, Italy
| | - Vincenzo Vindigni
- Plastic and Reconstructive Surgery Unit, University of Padua, Padua, Italy
| | - Franco Bassetto
- Plastic and Reconstructive Surgery Unit, University of Padua, Padua, Italy
| |
Collapse
|
48
|
Damayanti RH, Rusdiana T, Wathoni N. Mesenchymal Stem Cell Secretome for Dermatology Application: A Review. Clin Cosmet Investig Dermatol 2021; 14:1401-1412. [PMID: 34675575 PMCID: PMC8502696 DOI: 10.2147/ccid.s331044] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/21/2021] [Indexed: 12/25/2022]
Abstract
Secretome, also known as conditioned medium, is a secreted molecule from mesenchymal stem cells (MSCs) that has a variety of biological activities that can be used in various therapies, especially on the skin applications. A lack of conventional therapies makes secretome as a promising alternative therapy. The presence of growth factors, cytokines, and extracellular vesicles including microvesicles and exosomes in secretome has been widely reported, which serves in improving the proliferation and migration of cells to help in skin regeneration. Therefore, we were able to optimize the use of this secretome in a well-needed special review related to its work in addressing various skin problems. So, in this article, we discussed the benefits and biological activity of secretome on the skin application. This review was compiled based on the approval of several sites, such as Scopus, PubMed, Science Direct, and Google Scholar with the terms "MSC secretome for skin," "secretome for skin," "secretome dermatology," "secretome conditioned medium for skin," "secretome conditioned medium for skin wound," "secretome conditioned medium for aging," "secretome conditioned medium for hair growth," and "secretome conditioned medium for psoriasis." A total of 215 articles were collected for selection, of which 90 articles were used. Based on the results, it was concluded that secretome has a variety of useful activities to regenerate and repair tissue damage that have not been used on the skin, such as for wound healing, photoprotection, promotion of hair growth, psoriasis treatment, and other application as antimicrobial.
Collapse
Affiliation(s)
- Restu Harisma Damayanti
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45353, Indonesia
| | - Taofik Rusdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45353, Indonesia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45353, Indonesia
| |
Collapse
|
49
|
Mesenchymal Stem Cells in the Treatment of COVID-19, a Promising Future. Cells 2021; 10:cells10102588. [PMID: 34685567 PMCID: PMC8533906 DOI: 10.3390/cells10102588] [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: 08/18/2021] [Revised: 09/11/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in virtually all tissues; they have a potent self-renewal capacity and can differentiate into multiple cell types. They also affect the ambient tissue by the paracrine secretion of numerous factors in vivo, including the induction of other stem cells’ differentiation. In vitro, the culture media supernatant is named secretome and contains soluble molecules and extracellular vesicles that retain potent biological function in tissue regeneration. MSCs are considered safe for human treatment; their use does not involve ethical issues, as embryonic stem cells do not require genetic manipulation as induced pluripotent stem cells, and after intravenous injection, they are mainly found in the lugs. Therefore, these cells are currently being tested in various preclinical and clinical trials for several diseases, including COVID-19. Several affected COVID-19 patients develop induced acute respiratory distress syndrome (ARDS) associated with an uncontrolled inflammatory response. This condition causes extensive damage to the lungs and may leave serious post-COVID-19 sequelae. As the disease may cause systemic alterations, such as thromboembolism and compromised renal and cardiac function, the intravenous injection of MSCs may be a therapeutic alternative against multiple pathological manifestations. In this work, we reviewed the literature about MSCs biology, focusing on their function in pulmonary regeneration and their use in COVID-19 treatment.
Collapse
|
50
|
Tang Y, Li J, Wang W, Chen B, Chen J, Shen Z, Hou J, Mei Y, Liu S, Zhang L, Li Z, Lu S. Platelet extracellular vesicles enhance the proangiogenic potential of adipose-derived stem cells in vivo and in vitro. Stem Cell Res Ther 2021; 12:497. [PMID: 34503551 PMCID: PMC8427862 DOI: 10.1186/s13287-021-02561-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Adipose-derived mesenchymal stem cells (ADSC)-based therapy is an outstanding treatment strategy for ischaemic disease. However, the therapeutic efficacy of this strategy is not ideal due to the poor paracrine function and low survival rate of ADSCs in target regions. Platelet extracellular vesicles (PEVs) are nanoparticles derived from activated platelets that can participate in communication between cells. Accumulating evidence indicates that PEVs can regulate the biological functions of several cell lines. In the present study, we aimed to investigate whether PEVs can modulate the proangiogenic potential of ADSCs in vitro and in vivo. METHODS PEVs were identified using scanning electron microscope (SEM), flow cytometry (FCM) and nanoparticle tracking analysis (NTA). The CCK8 assay was performed to detect proliferation of cells. Transwell and wound healing assays were performed to verify migration capacity of cells. AnnexinV-FITC/PI apoptosis kit and live/dead assay were performed to assess ADSCs apoptosis under Cocl2-induced hypoxia condition. The underlying mechanisms by which PEVs affected ADSCs were explored using real time-PCR(RT-PCR) and Western blot. In addition, matrigel plug assays were conducted and mouse hindlimb ischaemic models were established to investigate the proangiogenic potential of PEV-treated ADSCs in vivo. RESULTS We demonstrated that ADSC could internalize PEVs, which lead to a series of biological reactions. In vitro, dose-dependent effects of PEVs on ADSC proliferation, migration and antiapoptotic capacity were observed. Western blotting results suggested that multiple proteins such as ERK, AKT, FAK, Src and PLCγ1 kinase may contribute to these changes. Furthermore, PEVs induced upregulation of several growth factors expression in ADSCs and amplified the proliferation, migration and tube formation of HUVECs induced by ADSC conditioned medium (CM). In in vivo experiments, compared with control ADSCs, the injection of PEV-treated ADSCs resulted in more vascularization in matrigel plugs, attenuated tissue degeneration and increased blood flow and capillary density in ischaemic hindlimb tissues. CONCLUSION Our data demonstrated that PEVs could enhance the proangiogenic potential of ADSCs in mouse hindlimb ischaemia. The major mechanisms of this effect included the promotion of ADSC proliferation, migration, anti-apoptosis ability and paracrine secretion.
Collapse
Affiliation(s)
- Yanan Tang
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jiayan Li
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Weiyi Wang
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Bingyi Chen
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jinxing Chen
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Zekun Shen
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jiaxuan Hou
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yifan Mei
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Shuang Liu
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Liwei Zhang
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Zongjin Li
- Nankai University School of Medicine, 94 Weijin Road, Tianjin, 300071, China.
| | - Shaoying Lu
- Vascular Surgery Department, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|