1
|
Atia GA, Abdal Dayem A, Taher ES, Alghonemy WY, Cho SG, Aldarmahi AA, Haque MA, Alshambky A, Taymour N, Ibrahim AM, Zaghamir DE, Elmorsy EM, Hetta HF, Mohamed ME, Abass KS, Khanday S, Abdeen A. Urine-derived stem cells: a sustainable resource for advancing personalized medicine and dental regeneration. Front Bioeng Biotechnol 2025; 13:1571066. [PMID: 40357329 PMCID: PMC12066649 DOI: 10.3389/fbioe.2025.1571066] [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: 02/05/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Urine-based therapy, an ancient practice, has been utilized across numerous civilizations to address a wide range of ailments. Urine was considered a priceless resource in numerous traditional therapeutic applications due to its reported medicinal capabilities. While the utilization of urine treatment is contentious and lacks significant support from modern healthcare, the discovery of urine-derived stem cells (UDSCs) has introduced a promising avenue for cell-based therapy. UDSCs offer a noninvasive and easily repeatable collection method, making them a practical and viable option for therapeutic applications. Research has shown that UDSCs contribute to organ preservation by promoting revascularization and decreasing inflammatory reactions in many diseases and conditions. This review will outline the contemporary status of UDSCs research and explore their potential applications in both fundamental science and medical practice.
Collapse
Affiliation(s)
- Gamal A. Atia
- Department of Oral Medicine, Periodontology, and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, Seoul, Republic of Korea
| | - Ehab S. Taher
- Department of Basic and Clinical Medical Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
| | - Wafaa Y. Alghonemy
- Department of Basic and Clinical Medical Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, Seoul, Republic of Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Republic of Korea
| | - Ahmed A. Aldarmahi
- Department of Basic Science, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- National Guard- Health Affairs, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
| | - Md Azizul Haque
- Department of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Abeer Alshambky
- Molecular Therapeutics Program, Fox Chase Cancer Center, Temple University, Philadelphia, PA, United States
- Department of Biochemistry, Animal Health Research Institute, Cairo, Egypt
| | - Noha Taymour
- Department of Substitutive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ateya M. Ibrahim
- College of Nursing, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Donia E. Zaghamir
- College of Nursing, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ekramy M. Elmorsy
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Helal F. Hetta
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohamed E. Mohamed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Kasim S. Abass
- Department of Physiology, Biochemistry, and Pharmacology, College of Veterinary Medicine, University of Kirkuk, Kirkuk, Iraq
| | - Shifan Khanday
- Department of Biomedical Sciences, Dubai Medical College for Girls, Dubai Medical University, Dubai, United Arab Emirates
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| |
Collapse
|
2
|
Mićanović D, Stanisavljević S, Li H, Koprivica I, Jonić N, Stojanović I, Savković V, Saksida T. Mesenchymal Stem Cells from Mouse Hair Follicles Inhibit the Development of Type 1 Diabetes. Int J Mol Sci 2024; 25:5974. [PMID: 38892159 PMCID: PMC11172537 DOI: 10.3390/ijms25115974] [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: 04/03/2024] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are known for their immunosuppressive properties. Based on the demonstrated anti-inflammatory effect of mouse MSCs from hair follicles (moMSCORS) in a murine wound closure model, this study evaluates their potential for preventing type 1 diabetes (T1D) in C57BL/6 mice. T1D was induced in C57BL/6 mice by repeated low doses of streptozotocin. moMSCORS were injected intravenously on weekly basis. moMSCORS reduced T1D incidence, the insulitis stage, and preserved insulin production in treated animals. moMSCORS primarily exerted immunomodulatory effects by inhibiting CD4+ T cell proliferation and activation. Ex vivo analysis indicated that moMSCORS modified the cellular immune profile within pancreatic lymph nodes and pancreatic infiltrates by reducing the numbers of M1 pro-inflammatory macrophages and T helper 17 cells and upscaling the immunosuppressive T regulatory cells. The proportion of pathogenic insulin-specific CD4+ T cells was down-scaled in the lymph nodes, likely via soluble factors. The moMSCORS detected in the pancreatic infiltrates of treated mice presumably exerted the observed suppressive effect on CD4+ through direct contact. moMSCORS alleviated T1D symptoms in the mouse, qualifying as a candidate for therapeutic products by multiple advantages: non-invasive sampling by epilation, easy access, permanent availability, scalability, and benefits of auto-transplantation.
Collapse
Affiliation(s)
- Dragica Mićanović
- Department of Immunology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (D.M.); (S.S.); (I.K.); (N.J.); (I.S.); (T.S.)
| | - Suzana Stanisavljević
- Department of Immunology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (D.M.); (S.S.); (I.K.); (N.J.); (I.S.); (T.S.)
| | - Hanluo Li
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China;
- Department of Cranial Maxillofacial Plastic Surgery, University Clinic Leipzig, 04103 Leipzig, Germany
| | - Ivan Koprivica
- Department of Immunology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (D.M.); (S.S.); (I.K.); (N.J.); (I.S.); (T.S.)
| | - Natalija Jonić
- Department of Immunology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (D.M.); (S.S.); (I.K.); (N.J.); (I.S.); (T.S.)
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (D.M.); (S.S.); (I.K.); (N.J.); (I.S.); (T.S.)
| | - Vuk Savković
- Department of Cranial Maxillofacial Plastic Surgery, University Clinic Leipzig, 04103 Leipzig, Germany
| | - Tamara Saksida
- Department of Immunology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia; (D.M.); (S.S.); (I.K.); (N.J.); (I.S.); (T.S.)
| |
Collapse
|
3
|
Huang RL, Li Q, Ma JX, Atala A, Zhang Y. Body fluid-derived stem cells - an untapped stem cell source in genitourinary regeneration. Nat Rev Urol 2023; 20:739-761. [PMID: 37414959 PMCID: PMC11639537 DOI: 10.1038/s41585-023-00787-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 07/08/2023]
Abstract
Somatic stem cells have been obtained from solid organs and tissues, including the bone marrow, placenta, corneal stroma, periosteum, adipose tissue, dental pulp and skeletal muscle. These solid tissue-derived stem cells are often used for tissue repair, disease modelling and new drug development. In the past two decades, stem cells have also been identified in various body fluids, including urine, peripheral blood, umbilical cord blood, amniotic fluid, synovial fluid, breastmilk and menstrual blood. These body fluid-derived stem cells (BFSCs) have stemness properties comparable to those of other adult stem cells and, similarly to tissue-derived stem cells, show cell surface markers, multi-differentiation potential and immunomodulatory effects. However, BFSCs are more easily accessible through non-invasive or minimally invasive approaches than solid tissue-derived stem cells and can be isolated without enzymatic tissue digestion. Additionally, BFSCs have shown good versatility in repairing genitourinary abnormalities in preclinical models through direct differentiation or paracrine mechanisms such as pro-angiogenic, anti-apoptotic, antifibrotic, anti-oxidant and anti-inflammatory effects. However, optimization of protocols is needed to improve the efficacy and safety of BFSC therapy before therapeutic translation.
Collapse
Affiliation(s)
- Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Anthony Atala
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
4
|
Cavaleiro C, Afonso GJM, Oliveira PJ, Valero J, Mota SI, Ferreiro E. Urine-derived stem cells in neurological diseases: current state-of-the-art and future directions. Front Mol Neurosci 2023; 16:1229728. [PMID: 37965041 PMCID: PMC10642248 DOI: 10.3389/fnmol.2023.1229728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2023] Open
Abstract
Stem cells have potential applications in the field of neurological diseases, as they allow for the development of new biological models. These models can improve our understanding of the underlying pathologies and facilitate the screening of new therapeutics in the context of precision medicine. Stem cells have also been applied in clinical tests to repair tissues and improve functional recovery. Nevertheless, although promising, commonly used stem cells display some limitations that curb the scope of their applications, such as the difficulty of obtention. In that regard, urine-derived cells can be reprogrammed into induced pluripotent stem cells (iPSCs). However, their obtaining can be challenging due to the low yield and complexity of the multi-phased and typically expensive differentiation protocols. As an alternative, urine-derived stem cells (UDSCs), included within the population of urine-derived cells, present a mesenchymal-like phenotype and have shown promising properties for similar purposes. Importantly, UDSCs have been differentiated into neuronal-like cells, auspicious for disease modeling, while overcoming some of the shortcomings presented by other stem cells for these purposes. Thus, this review assesses the current state and future perspectives regarding the potential of UDSCs in the ambit of neurological diseases, both for disease modeling and therapeutic applications.
Collapse
Affiliation(s)
- Carla Cavaleiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Coimbra, Portugal
| | - Gonçalo J. M. Afonso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Coimbra, Portugal
| | - Paulo J. Oliveira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Jorge Valero
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Cell Biology and Pathology, University of Salamanca, Salamanca, Spain
| | - Sandra I. Mota
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Elisabete Ferreiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
5
|
Dvorakova J, Wiesnerova L, Chocholata P, Kulda V, Landsmann L, Cedikova M, Kripnerova M, Eberlova L, Babuska V. Human cells with osteogenic potential in bone tissue research. Biomed Eng Online 2023; 22:33. [PMID: 37013601 PMCID: PMC10069154 DOI: 10.1186/s12938-023-01096-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
Bone regeneration after injury or after surgical bone removal due to disease is a serious medical challenge. A variety of materials are being tested to replace a missing bone or tooth. Regeneration requires cells capable of proliferation and differentiation in bone tissue. Although there are many possible human cell types available for use as a model for each phase of this process, no cell type is ideal for each phase. Osteosarcoma cells are preferred for initial adhesion assays due to their easy cultivation and fast proliferation, but they are not suitable for subsequent differentiation testing due to their cancer origin and genetic differences from normal bone tissue. Mesenchymal stem cells are more suitable for biocompatibility testing, because they mimic natural conditions in healthy bone, but they proliferate more slowly, soon undergo senescence, and some subpopulations may exhibit weak osteodifferentiation. Primary human osteoblasts provide relevant results in evaluating the effect of biomaterials on cellular activity; however, their resources are limited for the same reasons, like for mesenchymal stem cells. This review article provides an overview of cell models for biocompatibility testing of materials used in bone tissue research.
Collapse
Affiliation(s)
- Jana Dvorakova
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00, Plzen, Czech Republic
| | - Lucie Wiesnerova
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00, Plzen, Czech Republic
| | - Petra Chocholata
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00, Plzen, Czech Republic
| | - Vlastimil Kulda
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00, Plzen, Czech Republic
| | - Lukas Landsmann
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00, Plzen, Czech Republic
| | - Miroslava Cedikova
- Biomedical Center, Laboratory of Tumor Biology and Immunotherapy, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00, Plzen, Czech Republic
| | - Michaela Kripnerova
- Department of Biology, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00, Plzen, Czech Republic
| | - Lada Eberlova
- Department of Anatomy, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00, Plzen, Czech Republic
| | - Vaclav Babuska
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00, Plzen, Czech Republic.
| |
Collapse
|
6
|
Adelipour M, Lubman DM, Kim J. Potential applications of mesenchymal stem cells and their derived exosomes in regenerative medicine. Expert Opin Biol Ther 2023; 23:491-507. [PMID: 37147781 PMCID: PMC10330313 DOI: 10.1080/14712598.2023.2211203] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/03/2023] [Indexed: 05/07/2023]
Abstract
INTRODUCTION Regenerative medicine involves the replacement of damaged cells, tissues, or organs to restore normal function. Mesenchymal stem cells (MSCs) and exosomes secreted by MSCs have unique advantages that make them a suitable candidate in the field of regenerative medicine. AREAS COVERED This article provides a comprehensive overview of regenerative medicine, focusing on the use of MSCs and their exosomes as potential therapies for replacing damaged cells, tissues, or organs. This article discusses the distinct advantages of both MSCs and their secreted exosomes, including their immunomodulatory effects, lack of immunogenicity, and recruitment to damaged areas. While both MSCs and exosomes have these advantages, MSCs also have the unique ability to self-renew and differentiate. This article also assesses the current challenges associated with the application of MSCs and their secreted exosomes in therapy. We have reviewed proposed solutions for improving MSC or exosome therapy, including ex-vivo preconditioning strategies, genetic modification, and encapsulation. Literature search was conducted using Google Scholar and PubMed databases. EXPERT OPINION Providing insight into the future development of MSC and exosome-based therapies and to encourage the scientific community to focus on the identified gaps, develop appropriate guidelines, and enhance the clinical application of these therapies.
Collapse
Affiliation(s)
- Maryam Adelipour
- Department of Chemistry, Chungnam National University, Daejeon, Republic of Korea
- Department of Biochemistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - David M. Lubman
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jeongkwon Kim
- Department of Chemistry, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
7
|
Huang YZ, He T, Cui J, Jiang YL, Zeng JF, Zhang WQ, Xie HQ. Urine-Derived Stem Cells for Regenerative Medicine: Basic Biology, Applications, and Challenges. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:978-994. [PMID: 35049395 DOI: 10.1089/ten.teb.2021.0142] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Regenerative medicine based on stem cell research has the potential to provide advanced health care for human beings. Recent studies demonstrate that stem cells in human urine can serve as an excellent source of graft cells for regenerative therapy, mainly due to simple, low-cost, and noninvasive cell isolation. These cells, termed human urine-derived stem cells (USCs), are highly expandable and can differentiate into various cell lineages. They share many biological properties with mesenchymal stem cells, such as potent paracrine effects and immunomodulation ability. The advantage of USCs has motivated researchers to explore their applications in regenerative medicine, including genitourinary regeneration, musculoskeletal repair, skin wound healing, and disease treatment. Although USCs have showed many positive outcomes in preclinical studies, and although the possible applications of USCs for animal therapy have been reported, many issues need to be addressed before clinical translation. This article provides a comprehensive review of USC biology and recent advances in their application for tissue regeneration. Challenges in the clinical translation of USC-based therapy are also discussed. Impact statement Recently, stem cells isolated from urine, referred to as urine-derived stem cells (USCs), have gained much interest in the field of regenerative medicine. Many advantages of human USCs have been found for cell-based therapy: (i) the cell isolation procedure is simple and low cost; (ii) they have remarkable proliferation ability, multidifferentiation potential, and paracrine effects; and (iii) they facilitate tissue regeneration in many animal models. With the hope to facilitate the development of USC-based therapy, we describe the current understanding of USC biology, summarize recent advances in their applications, and discuss future challenges in clinical translation.
Collapse
Affiliation(s)
- Yi-Zhou Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Tao He
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China.,Department of Breast Surgery, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, China
| | - Jing Cui
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yan-Lin Jiang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Jun-Feng Zeng
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Wen-Qian Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| |
Collapse
|
8
|
Falzarano MS, Grilli A, Zia S, Fang M, Rossi R, Gualandi F, Rimessi P, El Dani R, Fabris M, Lu Z, Li W, Mongini T, Ricci F, Pegoraro E, Bello L, Barp A, Sansone VA, Hegde M, Roda B, Reschiglian P, Bicciato S, Selvatici R, Ferlini A. RNA-seq in DMD urinary stem cells recognized muscle-related transcription signatures and addressed the identification of atypical mutations by whole-genome sequencing. HGG ADVANCES 2022; 3:100054. [PMID: 35047845 PMCID: PMC8756543 DOI: 10.1016/j.xhgg.2021.100054] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 08/18/2021] [Indexed: 12/13/2022] Open
Abstract
Urinary stem cells (USCs) are a non-invasive, simple, and affordable cell source to study human diseases. Here we show that USCs are a versatile tool for studying Duchenne muscular dystrophy (DMD), since they are able to address RNA signatures and atypical mutation identification. Gene expression profiling of DMD individuals' USCs revealed a profound deregulation of inflammation, muscle development, and metabolic pathways that mirrors the known transcriptional landscape of DMD muscle and worsens following USCs' myogenic transformation. This pathogenic transcription signature was reverted by an exon-skipping corrective approach, suggesting the utility of USCs in monitoring DMD antisense therapy. The full DMD transcript profile performed in USCs from three undiagnosed DMD individuals addressed three splicing abnormalities, which were decrypted and confirmed as pathogenic variations by whole-genome sequencing (WGS). This combined genomic approach allowed the identification of three atypical and complex DMD mutations due to a deep intronic variation and two large inversions, respectively. All three mutations affect DMD gene splicing and cause a lack of dystrophin protein production, and one of these also generates unique fusion genes and transcripts. Further characterization of USCs using a novel cell-sorting technology (Celector) highlighted cell-type variability and the representation of cell-specific DMD isoforms. Our comprehensive approach to USCs unraveled RNA, DNA, and cell-specific features and demonstrated that USCs are a robust tool for studying and diagnosing DMD.
Collapse
Affiliation(s)
- Maria S Falzarano
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Andrea Grilli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena 41121, Italy
| | | | | | - Rachele Rossi
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Francesca Gualandi
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Paola Rimessi
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Reem El Dani
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Marina Fabris
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | | | - Wenyan Li
- BGI-Shenzhen, Shenzhen 518083, China
| | | | | | - Elena Pegoraro
- ERN Neuromuscular Center, Department of Neurosciences, Unit of Neurology, University of Padua, Padua 35122, Italy
| | - Luca Bello
- ERN Neuromuscular Center, Department of Neurosciences, Unit of Neurology, University of Padua, Padua 35122, Italy
| | - Andrea Barp
- The NEMO Clinical Center, Neurorehabilitation Unit, University of Milan, Milan 20162, Italy
| | - Valeria A Sansone
- The NEMO Clinical Center, Neurorehabilitation Unit, University of Milan, Milan 20162, Italy
| | - Madhuri Hegde
- PerkinElmer Genomics, 3950 Shackleford Rd., Ste. 195, Duluth, GA 30096, USA
| | - Barbara Roda
- Stem Sel s.r.l., Bologna 40127, Italy
- Department of Chemistry "G. Ciamician," University of Bologna, Bologna 40126, Italy
| | - Pierluigi Reschiglian
- Stem Sel s.r.l., Bologna 40127, Italy
- Department of Chemistry "G. Ciamician," University of Bologna, Bologna 40126, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena 41121, Italy
| | - Rita Selvatici
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| | - Alessandra Ferlini
- Department of Medical Sciences, Unit of Medical Genetics, University of Ferrara, Ferrara 44121, Italy
| |
Collapse
|
9
|
Zhang W, Hu J, Huang Y, Wu C, Xie H. Urine-derived stem cells: applications in skin, bone and articular cartilage repair. BURNS & TRAUMA 2021; 9:tkab039. [PMID: 34859109 PMCID: PMC8633594 DOI: 10.1093/burnst/tkab039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/18/2021] [Indexed: 02/05/2023]
Abstract
As an emerging type of adult stem cell featuring non-invasive acquisition, urine-derived stem cells (USCs) have shown great potential for applications in tissue engineering and regenerative medicine. With a growing amount of research on the topic, the effectiveness of USCs in various disease models has been shown and the underlying mechanisms have also been explored, though many aspects still remain unclear. In this review, we aim to provide an up-to-date overview of the biological characteristics of USCs and their applications in skin, bone and articular cartilage repair. In addition to the identification procedure of USCs, we also summarize current knowledge of the underlying repair mechanisms and application modes of USCs. Potential concerns and perspectives have also been summarized.
Collapse
Affiliation(s)
- Wenqian Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jungen Hu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yizhou Huang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chenyu Wu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huiqi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
10
|
Falzarano MS, Rossi R, Grilli A, Fang M, Osman H, Sabatelli P, Antoniel M, Lu Z, Li W, Selvatici R, Al-Khalili C, Gualandi F, Bicciato S, Torelli S, Ferlini A. Urine-Derived Stem Cells Express 571 Neuromuscular Disorders Causing Genes, Making Them a Potential in vitro Model for Rare Genetic Diseases. Front Physiol 2021; 12:716471. [PMID: 34744760 PMCID: PMC8565768 DOI: 10.3389/fphys.2021.716471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Neuromuscular disorders (NMDs) are a heterogeneous group of genetic diseases, caused by mutations in genes involved in spinal cord, peripheral nerve, neuromuscular junction, and muscle functions. To advance the knowledge of the pathological mechanisms underlying NMDs and to eventually identify new potential drugs paving the way for personalized medicine, limitations regarding the availability of neuromuscular disease-related biological samples, rarely accessible from patients, are a major challenge. Aim: We characterized urinary stem cells (USCs) by in-depth transcriptome and protein profiling to evaluate whether this easily accessible source of patient-derived cells is suitable to study neuromuscular genetic diseases, focusing especially on those currently involved in clinical trials. Methods: The global transcriptomics of either native or MyoD transformed USCs obtained from control individuals was performed by RNA-seq. The expression of 610 genes belonging to 16 groups of disorders (http://www.musclegenetable.fr/) whose mutations cause neuromuscular diseases, was investigated on the RNA-seq output. In addition, protein expression of 11 genes related to NMDs including COL6A, EMD, LMNA, SMN, UBA1, DYNC1H1, SOD1, C9orf72, DYSF, DAG1, and HTT was analyzed in native USCs by immunofluorescence and/or Western blot (WB). Results: RNA-seq profile of control USCs shows that 571 out of 610 genes known to be involved in NMDs, are expressed in USCs. Interestingly, the expression levels of the majority of NMD genes remain unmodified following USCs MyoD transformation. Most genes involved in the pathogenesis of all 16 groups of NMDs are well represented except for channelopathies and malignant hyperthermia related genes. All tested proteins showed high expression values, suggesting consistency between transcription and protein representation in USCs. Conclusion: Our data suggest that USCs are human cells, obtainable by non-invasive means, which might be used as a patient-specific cell model to study neuromuscular disease-causing genes and that they can be likely adopted for a variety of in vitro functional studies such as mutation characterization, pathway identification, and drug screening.
Collapse
Affiliation(s)
- Maria Sofia Falzarano
- UOL (Unità Operativa Logistica) of Medical Genetics, University of Ferrara, Ferrara, Italy
| | - Rachele Rossi
- UOL (Unità Operativa Logistica) of Medical Genetics, University of Ferrara, Ferrara, Italy.,The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Andrea Grilli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Mingyan Fang
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Hana Osman
- UOL (Unità Operativa Logistica) of Medical Genetics, University of Ferrara, Ferrara, Italy.,Department of Medical Microbiology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Patrizia Sabatelli
- CNR-Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza"- Unit of Bologna, Bologna, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Manuela Antoniel
- CNR-Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza"- Unit of Bologna, Bologna, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Zhiyuan Lu
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Wenyan Li
- Beijing Genomics Institute (BGI)-Shenzhen, Shenzhen, China
| | - Rita Selvatici
- UOL (Unità Operativa Logistica) of Medical Genetics, University of Ferrara, Ferrara, Italy
| | - Cristina Al-Khalili
- Department of Proteomics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Francesca Gualandi
- UOL (Unità Operativa Logistica) of Medical Genetics, University of Ferrara, Ferrara, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Torelli
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| | - Alessandra Ferlini
- UOL (Unità Operativa Logistica) of Medical Genetics, University of Ferrara, Ferrara, Italy.,The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
11
|
Wu R, Soland M, Liu G, Shi Y, Zhang C, Tang Y, Almeida-Porada G, Zhang Y. Functional characterization of the immunomodulatory properties of human urine-derived stem cells. Transl Androl Urol 2021; 10:3566-3578. [PMID: 34733653 PMCID: PMC8511544 DOI: 10.21037/tau-21-506] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/02/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Urine-derived stem cells (USCs) have been widely researched as a novel cell source for stem cell therapy, but their immunomodulatory characteristics remain to be investigated. This study aimed to characterize the immunomodulatory properties of human USCs. METHODS Human USCs were isolated from fresh voiding urine samples from healthy male donors and expanded. Their cell surface markers were characterized by flow cytometry analysis and the telomerase activities for several USCs clones were determined. The immunosuppressive potential of USCs was evaluated by the performing the mixed lymphocyte reaction (MLR) [co-culture with peripheral blood mononuclear cells (PBMNCs)] and natural killer cells (NK) cytotoxicity assay. USCs cytokines release profile was determined by using human cytokine proteome array. RESULTS USCs exhibited high cell surface expression of embryonic/mesenchymal stem cells (MSCs) markers CD29, CD44, CD54, CD73, CD90, CD146, and CD166, while lacked expression of hematopoietic stem cell markers CD11, CD14, CD19, CD31, CD34, CD45, B cell marker CD79, and co-stimulatory factors CD80 and CD86, thus, exhibiting the phenotype of MSCs. MLR indicated that USCs significantly inhibited the proliferation of PBMNCs, as compared to that of the human smooth muscle cells (SMCs). In cell cytotoxicity assays, NK cells displayed less cytotoxicity against USCs than against bone marrow mesenchymal stem cells (BMSCs) and SMCs. Furthermore, upon PBMNCs stimulation, USCs secreted higher levels of immunomodulatory cytokines, including IL-6, IL-8, MCP-1, RANTES, GROα, and GM-CSF, compared to those of BMSCs, especially when directly contact mix-culture with PBMNCs. CONCLUSIONS USCs secreted immunoregulatory cytokines and possessed immunomodulatory properties, comparable to those of BMSCs.
Collapse
Affiliation(s)
- Rongpei Wu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Melisa Soland
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA
| | - Guihua Liu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA
- Reproductive Medical Center, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingai Shi
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chi Zhang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yiming Tang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA
| |
Collapse
|
12
|
Ghori FF, Wahid M. Induced pluripotent stem cells from urine of Duchenne muscular dystrophy patients. Pediatr Int 2021; 63:1038-1047. [PMID: 33599058 DOI: 10.1111/ped.14655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/23/2021] [Accepted: 02/08/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND The most common muscular dystrophy, Duchenne muscular dystrophy (DMD), is a lethal, X-linked disorder with no widespread cure. Worldwide, in vitro studies involving new, mutation-specific cures and regenerative therapies are employing disease-specific patient-specific cells. However, these may not be completely relevant for Pakistani children because of the human genome diversities and geographic variation in mutation type and frequency. Therefore, this study aimed to generate DMD induced pluripotent stem cells (iPSCs) from the urine of Pakistani children with DMD, to serve as a precious source of differentiated cells, such as Pakistani DMD-cardiomyocytes, for future disease-modelling, drug testing, and gene therapy. METHODS Urine-derived cells (UDCs) isolated from mid-stream urine underwent molecular characterization and cellular reprogramming towards iPSCs using the episomal vector system followed by molecular profiling of the iPSCs. RESULTS Colonies of elongated and spindle-shaped or rounded rice-grain like UDCs were spotted 4-7 days after plating and expanded rapidly with a second passage at 2-3 weeks. Multicolor flow cytometry confirmed the expression of mesenchymal stem-cell markers. The reprogramed iPSCs consisted of colonies of round, tightly-packed cells with large nuclei that were positively fluorescent for the pluripotency markers octamer binding transcription factor-4 (OCT-4), tumour resistance antigen 1-60 (TRA-1-60), and stage specific embryonic 4 antigen (SSEA-4), but not for the negative pluripotency marker SSEA-1. To the best of our knowledge, this was the first time DMD-iPSCs have been generated for Pakistani children. CONCLUSION This integration-free, feeder-free, efficient, and reproducible reprogramming method employed UDCs. Urine is a low-cost, non-invasive, painless, and repeatable source of rapidly expandable cells from children and morbid individuals for obtaining autologous cells for drug-assays and disease-modelling, suitable for DMD and other debilitating diseases.
Collapse
Affiliation(s)
- Fareeha Faizan Ghori
- Stem Cells and Regenerative Medicine Research Group, Dow Research Institute of Biotechnology and Biomedical Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Mohsin Wahid
- Stem Cells and Regenerative Medicine Research Group, Dow Research Institute of Biotechnology and Biomedical Sciences, Dow University of Health Sciences, Karachi, Pakistan.,Department of Pathology, Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
13
|
Shi T, Cheung M. Urine-derived induced pluripotent/neural stem cells for modeling neurological diseases. Cell Biosci 2021; 11:85. [PMID: 33985584 PMCID: PMC8117626 DOI: 10.1186/s13578-021-00594-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/29/2021] [Indexed: 01/05/2023] Open
Abstract
Neurological diseases are mainly modeled using rodents through gene editing, surgery or injury approaches. However, differences between humans and rodents in terms of genetics, neural development, and physiology pose limitations on studying disease pathogenesis in rodent models for neuroscience research. In the past decade, the generation of induced pluripotent stem cells (iPSCs) and induced neural stem cells (iNSCs) by reprogramming somatic cells offers a powerful alternative for modeling neurological diseases and for testing regenerative medicines. Among the different somatic cell types, urine-derived stem cells (USCs) are an ideal cell source for iPSC and iNSC reprogramming, as USCs are highly proliferative, multipotent, epithelial in nature, and easier to reprogram than skin fibroblasts. In addition, the use of USCs represents a simple, low-cost and non-invasive procedure for generating iPSCs/iNSCs. This review describes the cellular and molecular properties of USCs, their differentiation potency, different reprogramming methods for the generation of iPSCs/iNSCs, and their potential applications in modeling neurological diseases.
Collapse
Affiliation(s)
- Tianyuan Shi
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Martin Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
14
|
Burdeyron P, Giraud S, Hauet T, Steichen C. Urine-derived stem/progenitor cells: A focus on their characterization and potential. World J Stem Cells 2020; 12:1080-1096. [PMID: 33178393 PMCID: PMC7596444 DOI: 10.4252/wjsc.v12.i10.1080] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/26/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Cell therapy, i.e., the use of cells to repair an affected tissue or organ, is at the forefront of regenerative and personalized medicine. Among the multiple cell types that have been used for this purpose [including adult stem cells such as mesenchymal stem cells or pluripotent stem cells], urine-derived stem cells (USCs) have aroused interest in the past years. USCs display classical features of mesenchymal stem cells such as differentiation capacity and immunomodulation. Importantly, they have the main advantage of being isolable from one sample of voided urine with a cheap and unpainful procedure, which is broadly applicable, whereas most adult stem cell types require invasive procedure. Moreover, USCs can be differentiated into renal cell types. This is of high interest for renal cell therapy-based regenerative approaches. This review will firstly describe the isolation and characterization of USCs. We will specifically present USC phenotype, which is not an object of consensus in the literature, as well as detail their differentiation capacity. In the second part of this review, we will present and discuss the main applications of USCs. These include use as a substrate to generate human induced pluripotent stem cells, but we will deeply focus on the use of USCs for cell therapy approaches with a detailed analysis depending on the targeted organ or system. Importantly, we will also focus on the applications that rely on the use of USC-derived products such as microvesicles including exosomes, which is a strategy being increasingly employed. In the last section, we will discuss the remaining barriers and challenges in the field of USC-based regenerative medicine.
Collapse
Affiliation(s)
- Perrine Burdeyron
- INSERM U1082 IRTOMIT, CHU de Poitiers, Poitiers 86021, France
- Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers 86021, France
| | - Sébastien Giraud
- INSERM U1082 IRTOMIT, CHU de Poitiers, Poitiers 86021, France
- Service de Biochimie, CHU de Poitiers, Poitiers 86021, France
| | - Thierry Hauet
- INSERM U1082 IRTOMIT, CHU de Poitiers, Poitiers 86021, France
- Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers 86021, France
- Service de Biochimie, CHU de Poitiers, Poitiers 86021, France
| | - Clara Steichen
- INSERM U1082 IRTOMIT, CHU de Poitiers, Poitiers 86021, France
- Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers 86021, France.
| |
Collapse
|
15
|
Jena D, Kharche SD, Singh SP, Rani S, Dige MS, Ranjan R, Singh SK, Kumar H. Growth and proliferation of caprine bone marrow mesenchymal stem cells on different culture media. Tissue Cell 2020; 67:101446. [PMID: 33099198 DOI: 10.1016/j.tice.2020.101446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 01/26/2023]
Abstract
The growth and proliferation of mesenchymal stem cells are very sensitive in in vitro and a number of factors like media play a significant role in that context. In this study we assessed effect of different media on growth and proliferation of bone marrow derived mesenchymal stem cells (BMMSCs). The BMMSCs were isolated from caprine bone marrow and were subjected to magnetic activated cell sorting against CD90+, CD105+, CD271+and CD34- along with FC blocker. After characterisation, 2 × 104 cells were seeded in 12 well culture plates in four different media viz. MesenCult, MesenPRO, StemPro and complete DMEM (15 % FBS) to study their growth kinetic for 6 days from passage 0 (P0) to passage 3 (P3). The population doubling time (PDT) was derived from growth curve using logarithmic formula. The results showed that the BMMSCs growth and proliferation was highest in MesenCult media in P0 which varied significantly (p < 0.05) from rest of media and from P1 to P3, it was MesenPRO which yielded maximum cells (p < 0.05). The PDT was also in line with growth curve findings. In conclusion, the MesenPRO media had higher growth and proliferation rate from P1 to P3 although MesenCult had higher cell numbers in P0. In conclusion, the use of MesenPRO media could be a better option than conventional media when mesenchymal stem cells are used in clinical applications and other therapeutic purposes taking consideration to its higher growth and proliferation rate. And MesenCult would be a great option to harvest MSCs from P0.
Collapse
Affiliation(s)
- Dayanidhi Jena
- Dept. of Veterinary Clinical Complex, Faculty of Veterinary & Animal Sciences, Banaras Hindu University, Varanasi, U.P., 231001, India
| | - Suresh Dinkar Kharche
- ICAR-Central Institute for Research on Goats (CIRG), Makhdoom, Farah, 281122, Mathura, U.P., India.
| | - Shiva Pratap Singh
- ICAR-Central Institute for Research on Goats (CIRG), Makhdoom, Farah, 281122, Mathura, U.P., India
| | - Sonam Rani
- ICAR-Central Institute for Research on Goats (CIRG), Makhdoom, Farah, 281122, Mathura, U.P., India
| | - Mahesh Shivanand Dige
- ICAR-Central Institute for Research on Goats (CIRG), Makhdoom, Farah, 281122, Mathura, U.P., India
| | - Ravi Ranjan
- ICAR-Central Institute for Research on Goats (CIRG), Makhdoom, Farah, 281122, Mathura, U.P., India
| | - Sanjay Kumar Singh
- ICAR-Indian Veterinary Research Institute, Bareilly, U.P., 243122, India
| | - Harendra Kumar
- ICAR-Indian Veterinary Research Institute, Bareilly, U.P., 243122, India
| |
Collapse
|
16
|
Zhou C, Wu XR, Liu HS, Liu XH, Liu GH, Zheng XB, Hu T, Liang ZX, He XW, Wu XJ, Smith LC, Zhang Y, Lan P. Immunomodulatory Effect of Urine-derived Stem Cells on Inflammatory Bowel Diseases via Downregulating Th1/Th17 Immune Responses in a PGE2-dependent Manner. J Crohns Colitis 2020; 14:654-668. [PMID: 31841595 DOI: 10.1093/ecco-jcc/jjz200] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Despite the therapeutic promise of stem cell therapy in the treatment of inflammatory bowel diseases [IBD], most donor cell populations have to be obtained via invasive approaches and often remain insufficiently validated. Urine-derived stem cells [USC] were recently shown to have regenerative properties and can be harvested in a safe, low-cost, and noninvasive way. This study aims to evaluate the immunomodulatory effect of USC and their efficacy in the management of IBD. METHODS Human USC were isolated and expanded from the urine of healthy male adult volunteers [n = 3, age range 24-30 years]. USC were characterised by cell surface marker expression profile and multipotent differentiation. The in vitro immunomodulatory effect of USC was evaluated by co-culturing with human CD4+ T cells upon stimulation with phytohaemagglutinin [PHA]. The proliferation of CD4+ T was measured by fluorescence-activated cell sorting [FACS]. Cytokine array and quantitative real-time polymerase chain reaction [RT-PCR] were applied to examine cytokine levels. In vivo therapeutic value of USC was assessed using a murine colitis model induced by dextran sulphate sodium [DSS] or 2, 4, 6-trinitrobenzene sulphonic acid [TNBS]. The immunomodulatory effect of USC and bone marrow-derived mesenchymal stem cells [BMSC] was compared when co-cultured with CD4+ T cells. The therapeutic efficacy of USC and BMSC on IBD was compared when administered in an acute DSS model in vivo. RESULTS USC were positive for mesenchymal stem cell markers but were negative for haematopoietic stem cell markers. These cells differentiated into osteo-, adipo-, and chondrogenic cell lineages. Similar to BMSC, the proliferation of CD4+ T cells was significantly inhibited when co-cultured with USC, as a consequence of Th1/Th17 immune response inhibition. Systemic administration of USC significantly ameliorated the clinical and histopathological severity of colitis and increased the survival rate in both acute and chronic murine colitis models. Moreover, implantation of USC led to downregulation of the Th1/Th17 immune responses in a PGE2-dependent manner. CONCLUSIONS This study demonstrated that implantation of USC reduces inflammation in an IBD rodent model via downregulation of Th1/Th17 immune responses, indicating that USC therapy serves as a potential cell-based therapeutic candidate treatment for IBD.
Collapse
Affiliation(s)
- Chi Zhou
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xian-Rui Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Hua-Shan Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xuan-Hui Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Gui-Hua Liu
- Reproductive Centre, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiao-Bin Zheng
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tuo Hu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhen-Xing Liang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xiao-Wen He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Jian Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Leona C Smith
- Emergency Medicine MD, PGY1, Albert B Chandler Hospital, Lexington, KY, USA
| | - Yuanyuan Zhang
- Wake Forest Institute of Regenerative Medicine, Wake Forest University, Winston Salem, NC, USA
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
17
|
Urine-Derived Stem Cells: Applications in Regenerative and Predictive Medicine. Cells 2020; 9:cells9030573. [PMID: 32121221 PMCID: PMC7140531 DOI: 10.3390/cells9030573] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/17/2020] [Accepted: 02/25/2020] [Indexed: 12/14/2022] Open
Abstract
Despite being a biological waste, human urine contains a small population of cells with self-renewal capacity and differentiation potential into several cell types. Being derived from the convoluted tubules of nephron, renal pelvis, ureters, bladder and urethra, urine-derived stem cells (UDSC) have a similar phenotype to mesenchymal stroma cells (MSC) and can be reprogrammed into iPSC (induced pluripotent stem cells). Having simple, safer, low-cost and noninvasive collection procedures, the interest in UDSC has been growing in the last decade. With great potential in regenerative medicine applications, UDSC can also be used as biological models for pharmacology and toxicology tests. This review describes UDSC biological characteristics and differentiation potential and their possible use, including the potential of UDSC-derived iPSC to be used in drug discovery and toxicology, as well as in regenerative medicine. Being a new cellular platform amenable to noninvasive collection for disease stratification and personalized therapy could be a future application for UDSC.
Collapse
|
18
|
Liu G, David BT, Trawczynski M, Fessler RG. Advances in Pluripotent Stem Cells: History, Mechanisms, Technologies, and Applications. Stem Cell Rev Rep 2020; 16:3-32. [PMID: 31760627 PMCID: PMC6987053 DOI: 10.1007/s12015-019-09935-x] [Citation(s) in RCA: 301] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past 20 years, and particularly in the last decade, significant developmental milestones have driven basic, translational, and clinical advances in the field of stem cell and regenerative medicine. In this article, we provide a systemic overview of the major recent discoveries in this exciting and rapidly developing field. We begin by discussing experimental advances in the generation and differentiation of pluripotent stem cells (PSCs), next moving to the maintenance of stem cells in different culture types, and finishing with a discussion of three-dimensional (3D) cell technology and future stem cell applications. Specifically, we highlight the following crucial domains: 1) sources of pluripotent cells; 2) next-generation in vivo direct reprogramming technology; 3) cell types derived from PSCs and the influence of genetic memory; 4) induction of pluripotency with genomic modifications; 5) construction of vectors with reprogramming factor combinations; 6) enhancing pluripotency with small molecules and genetic signaling pathways; 7) induction of cell reprogramming by RNA signaling; 8) induction and enhancement of pluripotency with chemicals; 9) maintenance of pluripotency and genomic stability in induced pluripotent stem cells (iPSCs); 10) feeder-free and xenon-free culture environments; 11) biomaterial applications in stem cell biology; 12) three-dimensional (3D) cell technology; 13) 3D bioprinting; 14) downstream stem cell applications; and 15) current ethical issues in stem cell and regenerative medicine. This review, encompassing the fundamental concepts of regenerative medicine, is intended to provide a comprehensive portrait of important progress in stem cell research and development. Innovative technologies and real-world applications are emphasized for readers interested in the exciting, promising, and challenging field of stem cells and those seeking guidance in planning future research direction.
Collapse
Affiliation(s)
- Gele Liu
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA.
| | - Brian T David
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA
| | - Matthew Trawczynski
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA
| | - Richard G Fessler
- Department of Neurosurgery, Rush University Medical College, 1725 W. Harrison St., Suite 855, Chicago, IL, 60612, USA
| |
Collapse
|
19
|
Identification and characterization of two morphologically distinct stem cell subpopulations from human urine samples. SCIENCE CHINA-LIFE SCIENCES 2019; 63:712-723. [PMID: 31515730 DOI: 10.1007/s11427-018-9543-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/06/2019] [Indexed: 02/05/2023]
Abstract
Urine-derived stem cells (USCs) have shown potentials for the treatment of skeletal and urological disorders. Based on published literature and our own data, USCs consist of heterogeneous populations of cells. In this paper, we identify and characterize two morphologically distinct subpopulations of USCs from human urine samples, named as spindle-shaped USCs (SS-USCs) and rice-shaped USCs (RS-USCs) respectively. The two subpopulations showed similar clone-forming efficiency, while SS-USCs featured faster proliferation, higher motility, and greater potential for osteogenic and adipogenic differentiation, RS-USCs showed greater potential for chondrogenic differentiation. POU5F1 was strongly expressed in both subpopulations, but MYC was weakly expressed. Both subpopulations showed similar patterns of CD24, CD29, CD34, CD44, CD73, CD90 and CD105 expression, while a higher percentage of RS-USCs were positive for CD133. SS-USCs were positive for VIM, weakly positive for SLC12A1 and UMOD, and negative for KRT18, NPHS1, AQP1 and AQP2, indicating a renal mesenchyme origin; while RS-USCs are positive for VIM, partially positive for KRT18, NPHS1, AQP1, SLC12A1 and UMOD, and negative for AQP2, indicating a nephron tubule origin. The above results can facilitate understanding of the biological characteristics of subpopulations of USCs, and provide a basis for further research and applications of such cells.
Collapse
|
20
|
Falzarano MS, Ferlini A. Urinary Stem Cells as Tools to Study Genetic Disease: Overview of the Literature. J Clin Med 2019; 8:jcm8050627. [PMID: 31071994 PMCID: PMC6572423 DOI: 10.3390/jcm8050627] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 02/07/2023] Open
Abstract
Urine specimens represent a novel and non-invasive approach to isolate patient-specific stem cells by easy and low-cost procedures, replacing the traditional sources (muscle/skin biopsy/adipose tissue) obtained with invasive and time-consuming methods. Urine-derived stem cells (USCs) can be used in a broad field of applications, such as regenerative medicine, cell therapy, diagnostic testing, disease modelling and drug screening. USCs are a good source of cells for generating induced pluripotent stem cells (iPSCs) and importantly, they can also be directly converted into specific cell lines. In this review, we show the features of USCs and their use as a promising in vitro model to study genetic diseases.
Collapse
Affiliation(s)
- Maria Sofia Falzarano
- UOL (Unita` Operativa Logistica) of Medical Genetics, University of Ferrara, 44121 Ferrara, Italy.
| | - Alessandra Ferlini
- UOL (Unita` Operativa Logistica) of Medical Genetics, University of Ferrara, 44121 Ferrara, Italy.
- Neuromuscular Unit, Great Ormond Street Hospital, University College London, Bloomsbury, London WC1E 6BT, UK.
| |
Collapse
|
21
|
Xing F, Liu G, Duan X, Xiang Z. [The application of urine derived stem cells in regeneration of musculoskeletal system]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:1477-1482. [PMID: 30417628 PMCID: PMC8414118 DOI: 10.7507/1002-1892.201804024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 10/13/2018] [Indexed: 02/05/2023]
Abstract
Objective To review the application of urine derived stem cells (USCs) in regeneration of musculoskeletal system. Methods The original literature about USCs in the regeneration of musculoskeletal system was extensively reviewed and analyzed. Results The source of USCs is noninvasive and extensive. USCs express MSCs surface markers with stable proliferative and multi-directional differentiation capabilities, and are widely used in bone, skin, nerve, and other skeletal and muscle system regeneration fields and show a certain repair capacity. Conclusion USCs from non-invasive sources have a wide application prospect in the regeneration of musculoskeletal system, but the definite biological mechanism of its repair needs further study.
Collapse
Affiliation(s)
- Fei Xing
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Guoming Liu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Xin Duan
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Zhou Xiang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041,
| |
Collapse
|
22
|
Chen H, Li J, Yan H. The transplantation of human urine stem cells combined with chondroitinase ABC promotes brain-derived neurotrophic factor and nerve growth factor following spinal cord injury in rats. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3858-3866. [PMID: 31949773 PMCID: PMC6962799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/04/2018] [Indexed: 06/10/2023]
Abstract
Cells based on therapies are currently gaining momentum in neural tissue engineering to treat spinal cord injury (SCI). The present study aimed to evaluate the effects of the concomitant use of human urine stem cells (hUSCs) and chondroitinase ABC (ChABC) on functional improvement and to explore the expressions of brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF). The SCI model was induced by a falling heavy object. hUSCs were cultured and transplanted into the impaired spinal cord with ChABC administration. The Basso, Beattie and Bresnahan (BBB) scores were valued, and real time PCR, immunofluorescence and Western blot were used to detect the expression of BDNF and NGF. We found that rats receiving both hUSCs and ChABC treatment demonstrated the best functional recovery. In addition, the mRNA and protein expressions of the BDNF and NGF expressions were found to be effectively higher in the combined treatment group than these in the other groups. In conclusion, hUSCs transplantation combined with ChABC administration promotes motor functional recovery in SCI rats, which may be associated with BDNF and NGF regulation.
Collapse
Affiliation(s)
- Hemu Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Anhui Medical University China
| | - Jian Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Anhui Medical University China
| | - Han Yan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Anhui Medical University China
| |
Collapse
|
23
|
Pavathuparambil Abdul Manaph N, Al-Hawaas M, Bobrovskaya L, Coates PT, Zhou XF. Urine-derived cells for human cell therapy. Stem Cell Res Ther 2018; 9:189. [PMID: 29996911 PMCID: PMC6042455 DOI: 10.1186/s13287-018-0932-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Desirable cells for human cell therapy would be ones that can be generated by simple isolation and culture techniques using a donor sample obtained by non-invasive methods. To date, the different donor-specific cells that can be isolated from blood, skin, and hair require invasive methods for sample isolation and incorporate complex and costly reagents to culture. These cells also take considerable time for their in-vitro isolation and expansion. Previous studies suggest that donor-derived cells, namely urine stem cells and renal cells, may be isolated from human urine samples using a cost-effective and simple method of isolation, incorporating not such complex reagents. Moreover, the isolated cells, particularly urine stem cells, are superior to conventional stem cell sources in terms of favourable gene profile and inherent multipotent potential. Transdifferentiation or differentiation of human urine-derived cells can generate desirable cells for regenerative therapy. In this review, we intended to discuss the characteristics and therapeutic applications of urine-derived cells for human cell therapy. Conclusively, with detailed study and optimisation, urine-derived cells have a prospective future to generate functional lineage-specific cells for patients from a clinical translation point of view.
Collapse
Affiliation(s)
- Nimshitha Pavathuparambil Abdul Manaph
- Central Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, 5000 South Australia
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, 5000 South Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, 5000 South Australia
| | - Mohammed Al-Hawaas
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, 5000 South Australia
| | - Larisa Bobrovskaya
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, 5000 South Australia
| | - Patrick T. Coates
- Central Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, 5000 South Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, 5000 South Australia
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, 5000 South Australia
| |
Collapse
|
24
|
Knuth CA, Kiernan CH, Palomares Cabeza V, Lehmann J, Witte-Bouma J, Ten Berge D, Brama PA, Wolvius EB, Strabbing EM, Koudstaal MJ, Narcisi R, Farrell E. Isolating Pediatric Mesenchymal Stem Cells with Enhanced Expansion and Differentiation Capabilities. Tissue Eng Part C Methods 2018; 24:313-321. [PMID: 29631483 DOI: 10.1089/ten.tec.2018.0031] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cells/marrow stromal cells (MSCs) are attractive for applications ranging from research and development to use in clinical therapeutics. However, the most commonly studied MSCs, adult bone marrow MSCs (A-MSCs), are limited by significant donor variation resulting in inconsistent expansion rates and multilineage differentiation capabilities. We have recently obtained permission to isolate pediatric MSCs (P-MSCs) from surplus iliac crest bone chips. Here, we developed a simple and easily replicable isolation protocol yielding P-MSCs, which adhere to MSC defining guidelines. After confirming immunophenotypic marker expression, we compared expansion rates, senescence, morphology, and trilineage differentiation of P-MSCs to A-MSCs for multiple donors. We found P-MSCs have faster in vitro replication, consistently show significantly lower senescence, and are capable of more reproducible multilineage differentiation than A-MSCs. We, therefore, believe P-MSCs are a promising candidate for use in research applications and potentially as part of an allogeneic therapeutic treatment.
Collapse
Affiliation(s)
- Callie An Knuth
- 1 Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| | - Caoimhe H Kiernan
- 1 Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| | - Virginia Palomares Cabeza
- 1 Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands .,2 Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands .,3 School of Veterinary Medicine, Veterinary Science Centre, University College Dublin , Dublin, Ireland
| | - Johannes Lehmann
- 4 Department of Cell Biology, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands .,5 Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| | - Janneke Witte-Bouma
- 1 Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| | - Derk Ten Berge
- 4 Department of Cell Biology, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| | - Pieter A Brama
- 3 School of Veterinary Medicine, Veterinary Science Centre, University College Dublin , Dublin, Ireland
| | - Eppo B Wolvius
- 1 Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| | - Elske M Strabbing
- 1 Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| | - Maarten J Koudstaal
- 1 Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| | - Roberto Narcisi
- 6 Department of Orthopedics, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| | - Eric Farrell
- 1 Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus MC, University Medical Center Rotterdam , Rotterdam, The Netherlands
| |
Collapse
|
25
|
Bacakova L, Zarubova J, Travnickova M, Musilkova J, Pajorova J, Slepicka P, Kasalkova NS, Svorcik V, Kolska Z, Motarjemi H, Molitor M. Stem cells: their source, potency and use in regenerative therapies with focus on adipose-derived stem cells - a review. Biotechnol Adv 2018; 36:1111-1126. [PMID: 29563048 DOI: 10.1016/j.biotechadv.2018.03.011] [Citation(s) in RCA: 371] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/12/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023]
Abstract
Stem cells can be defined as units of biological organization that are responsible for the development and the regeneration of organ and tissue systems. They are able to renew their populations and to differentiate into multiple cell lineages. Therefore, these cells have great potential in advanced tissue engineering and cell therapies. When seeded on synthetic or nature-derived scaffolds in vitro, stem cells can be differentiated towards the desired phenotype by an appropriate composition, by an appropriate architecture, and by appropriate physicochemical and mechanical properties of the scaffolds, particularly if the scaffold properties are combined with a suitable composition of cell culture media, and with suitable mechanical, electrical or magnetic stimulation. For cell therapy, stem cells can be injected directly into damaged tissues and organs in vivo. Since the regenerative effect of stem cells is based mainly on the autocrine production of growth factors, immunomodulators and other bioactive molecules stored in extracellular vesicles, these structures can be isolated and used instead of cells for a novel therapeutic approach called "stem cell-based cell-free therapy". There are four main sources of stem cells, i.e. embryonic tissues, fetal tissues, adult tissues and differentiated somatic cells after they have been genetically reprogrammed, which are referred to as induced pluripotent stem cells (iPSCs). Although adult stem cells have lower potency than the other three stem cell types, i.e. they are capable of differentiating into only a limited quantity of specific cell types, these cells are able to overcome the ethical and legal issues accompanying the application of embryonic and fetal stem cells and the mutational effects associated with iPSCs. Moreover, adult stem cells can be used in autogenous form. These cells are present in practically all tissues in the organism. However, adipose tissue seems to be the most advantageous tissue from which to isolate them, because of its abundancy, its subcutaneous location, and the need for less invasive techniques. Adipose tissue-derived stem cells (ASCs) are therefore considered highly promising in present-day regenerative medicine.
Collapse
Affiliation(s)
- Lucie Bacakova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic.
| | - Jana Zarubova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Martina Travnickova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Julia Pajorova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, 4-Krc, Czech Republic
| | - Petr Slepicka
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Nikola Slepickova Kasalkova
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Vaclav Svorcik
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technicka 5, 166 28 Prague, 6-Dejvice, Czech Republic
| | - Zdenka Kolska
- Faculty of Science, J.E. Purkyne University, Ceske mladeze 8, 400 96 Usti nad Labem, Czech Republic
| | - Hooman Motarjemi
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| | - Martin Molitor
- Clinic of Plastic Surgery, Faculty Hospital Na Bulovce, Budinova 67/2, 180 81 Prague, 8-Liben, Czech Republic
| |
Collapse
|
26
|
SNEV hPrp19/hPso4 Regulates Adipogenesis of Human Adipose Stromal Cells. Stem Cell Reports 2016; 8:21-29. [PMID: 28041875 PMCID: PMC5233435 DOI: 10.1016/j.stemcr.2016.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/01/2016] [Accepted: 12/01/2016] [Indexed: 11/26/2022] Open
Abstract
Aging is accompanied by loss of subcutaneous adipose tissue. This may be due to reduced differentiation capacity or deficiency in DNA damage repair (DDR) factors. Here we investigated the role of SNEVhPrp19/hPso4, which was implicated in DDR and senescence evasion, in adipogenic differentiation of human adipose stromal cells (hASCs). We showed that SNEV is induced during adipogenesis and localized both in the nucleus and in the cytoplasm. Knockdown of SNEV perturbed adipogenic differentiation and led to accumulation of DNA damage in hASCs upon oxidative stress. In addition, we demonstrated that SNEV is required for fat deposition in Caenorhabditis elegans. Consequently, we tested other DDR factors and found that WRN is also required for adipogenesis in both models. These results demonstrate that SNEV regulates adipogenesis in hASCs and indicate that DDR capacity in general might be a pre-requisite for this process.
SNEV is required for adipogenic differentiation of human adipose stromal cells SNEV modulates pro- and anti-adipogenic signaling pathways SNEV regulates DNA repair capacity of human adipose stromal cells SNEV modulates organismal fat deposition in Caenorhabditis elegans
Collapse
|