1
|
Kim H, Williams SJ, Colombo JS. Characterization of Fibronectin-Adherent, Non-Fibronectin-Adherent, and Explant-Derived Human Dental Pulp Stem Cell Populations. Dent J (Basel) 2025; 13:159. [PMID: 40277489 PMCID: PMC12026221 DOI: 10.3390/dj13040159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/18/2025] [Accepted: 03/28/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Dental pulp stem cells (DPSCs) are of significant interest due to their mesenchymal lineage and relative availability from extracted teeth. This study aims to examine the relationship between fibronectin-adherent, non-fibronectin-adherent, and explant-derived DPSC populations in terms of the population doubling rate in culture and the expression of mesenchymal cell surface markers and their capacity for osteodifferentiation. Methods: Human pulp tissue was removed from healthy extracted human teeth, enzymatically digested prior to seeding onto fibronectin-coated plates, and left to adhere for 20 min, yielding a fibronectin-adherent population. The remaining non-adherent cells were transferred and designated 'non-fibronectin-adherent.' Intact pulp was placed on uncoated plastic for 5 days, with the migrated cells designated 'explant-derived'. DPSCs from these populations were examined in terms of population doubling rates, the expression of CD90, CD44, CD105, and CD73, and the expression of RUNX2, SPP1, and BGLAP after 7 days in osteoinductive media. Results: The fibronectin-adherent cells had the greatest population doubling over time. All populations demonstrated comparable percentages of cells positive for mesenchymal markers, though individual marker expression varied slightly. The explant-derived cells showed increased expression of RUNX2 after 7 days in osteoinductive media, while the treated single-cell-suspension-derived populations showed increased expression of SPP1 mRNA. Conclusions: Fibronectin enrichment resulted in a population with the greatest rate of population doubling over extended culture compared to the other two populations. The proportion of cells positive for all four mesenchymal surface markers was the same between populations. The fibronectin-adherent and non-adherent cultures may have responded more rapidly to osteoinductive media than the explant-derived cells.
Collapse
Affiliation(s)
| | | | - John S. Colombo
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada, Las Vegas, NV 89106, USA; (H.K.); (S.J.W.)
| |
Collapse
|
2
|
Thetchinamoorthy K, Jarczak J, Kieszek P, Wierzbicka D, Ratajczak J, Kucia M, Ratajczak MZ. Very small embryonic-like stem cells (VSELs) on the way for potential applications in regenerative medicine. Front Bioeng Biotechnol 2025; 13:1564964. [PMID: 40124247 PMCID: PMC11926153 DOI: 10.3389/fbioe.2025.1564964] [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: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Evidence has accumulated that adult tissues contain a population of early development stem cells capable of differentiating across germ layers into various types of cells. Our group purified these rare cells, naming them very small embryonic-like stem cells (VSELs). With their broad differentiation potential, VSELs have emerged as a new candidate population for clinical applications. This advancement is now possible due to our recent development of a model for ex vivo expansion of these rare cells. Importantly, no evidence suggests that VSELs, isolated from adult tissues, can form teratomas. In this review paper, we update current research on these cells reported in our laboratory as well as in those of several independent investigators.
Collapse
Affiliation(s)
| | - Justyna Jarczak
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Patrycja Kieszek
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Diana Wierzbicka
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Janina Ratajczak
- Stem Cell Institute at Graham Brown Cancer Center, University of Louisville, Louisville, CO, United States
| | - Magdalena Kucia
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Mariusz Z. Ratajczak
- Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
- Stem Cell Institute at Graham Brown Cancer Center, University of Louisville, Louisville, CO, United States
| |
Collapse
|
3
|
Lica JJ, Jakóbkiewicz-Banecka J, Hellmann A. In Vitro models of leukemia development: the role of very small leukemic stem-like cells in the cellular transformation cascade. Front Cell Dev Biol 2025; 12:1463807. [PMID: 39830209 PMCID: PMC11740207 DOI: 10.3389/fcell.2024.1463807] [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: 07/15/2024] [Accepted: 11/28/2024] [Indexed: 01/22/2025] Open
Abstract
Recent experimental findings indicate that cancer stem cells originate from transformed very small embryonic-like stem cells. This finding represents an essential advancement in uncovering the processes that drive the onset and progression of cancer. In continuously growing cell lines, for the first time, our team's follow-up research on leukemia, lung cancer, and healthy embryonic kidney cells revealed stages that resembles very small precursor stem cells. This review explores the origin of leukemic stem-like cells from very small leukemic stem-like cells establish from transformed very small embryonic-like stem cells. We explore theoretical model of acute myeloid leukemia initiation and progresses through various stages, as well basing the HL60 cell line, present its hierarchical stage development in vitro, highlighting the role of these very small precursor primitive stages. We also discuss the potential implications of further research into these unique cellular stages for advancing leukemia and cancer treatment and prevention.
Collapse
Affiliation(s)
- Jan Jakub Lica
- Department Medical Biology and Genetics, Faculty of Biology, University of Gdansk, Gdansk, Poland
- Department Health Science; Powiśle University, Gdańsk, Poland
| | | | - Andrzej Hellmann
- Department of Hematology and Transplantology, Faculty of Medicine, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
4
|
Jarczak J, Bujko K, Ratajczak MZ, Kucia M. scRNA-seq revealed transcriptional signatures of human umbilical cord primitive stem cells and their germ lineage origin regulated by imprinted genes. Sci Rep 2024; 14:29264. [PMID: 39587190 PMCID: PMC11589151 DOI: 10.1038/s41598-024-79810-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
A population of CD133+lin-CD45- and CD34+lin-CD45- very small embryonic-like stem cells (VSELs) has been identified in postnatal human tissues, including bone marrow (BM), mobilized peripheral blood (mPB) and umbilical cord blood (UCB). Under appropriate conditions, VSELs in vitro and in vivo differentiate into tissue-committed stem cells for all three germ layers. Molecular analysis of adult murine BM-purified VSELs revealed that these rare cells deposited during development in adult tissues (i) express a similar transcriptome as embryonic stem cells, (ii) share several markers characteristic for epiblast and migratory primordial germ cells (PGCs), (iii) highly express a polycomb group protein enhancer of zeste drosophila homolog 2 (Ezh2) and finally (iv) display a unique pattern of imprinting at crucial paternally inherited genes that promotes their quiescence. Here, by employing single-cell RNA sequencing we demonstrate for the first time that purified from UCB human VSELs defined by expression of CD34 or CD133 antigens and lack of lineage markers, including CD45 antigen express similar molecular signature as murine BM-derived VSELs. Specifically, unsupervised clustering revealed numerous subpopulations of VSELs including ones i) annotated to germline compartments, ii) regulated by parental imprinting, iii) responding to early developmental fate decisions, iv) transcription factors involved in differentiation and development, including homeobox family of genes, and v) expressing innate immunity and purinergic signaling genes.
Collapse
Affiliation(s)
- Justyna Jarczak
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland
| | - Kamila Bujko
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland
| | - Mariusz Z Ratajczak
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland
- Stem Cell Institute at Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Magdalena Kucia
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland.
| |
Collapse
|
5
|
Cancedda R, Mastrogiacomo M. The Phoenix of stem cells: pluripotent cells in adult tissues and peripheral blood. Front Bioeng Biotechnol 2024; 12:1414156. [PMID: 39139297 PMCID: PMC11319133 DOI: 10.3389/fbioe.2024.1414156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
Pluripotent stem cells are defined as cells that can generate cells of lineages from all three germ layers, ectoderm, mesoderm, and endoderm. On the contrary, unipotent and multipotent stem cells develop into one or more cell types respectively, but their differentiation is limited to the cells present in the tissue of origin or, at most, from the same germ layer. Multipotent and unipotent stem cells have been isolated from a variety of adult tissues, Instead, the presence in adult tissues of pluripotent stem cells is a very debated issue. In the early embryos, all cells are pluripotent. In mammalians, after birth, pluripotent cells are maintained in the bone-marrow and possibly in gonads. In fact, pluripotent cells were isolated from marrow aspirates and cord blood and from cultured bone-marrow stromal cells (MSCs). Only in few cases, pluripotent cells were isolated from other tissues. In addition to have the potential to differentiate toward lineages derived from all three germ layers, the isolated pluripotent cells shared other properties, including the expression of cell surface stage specific embryonic antigen (SSEA) and of transcription factors active in the early embryos, but they were variously described and named. However, it is likely that they are part of the same cell population and that observed diversities were the results of different isolation and expansion strategies. Adult pluripotent stem cells are quiescent and self-renew at very low rate. They are maintained in that state under the influence of the "niche" inside which they are located. Any tissue damage causes the release in the blood of inflammatory cytokines and molecules that activate the stem cells and their mobilization and homing in the injured tissue. The inflammatory response could also determine the dedifferentiation of mature cells and their reversion to a progenitor stage and at the same time stimulate the progenitors to proliferate and differentiate to replace the damaged cells. In this review we rate articles reporting isolation and characterization of tissue resident pluripotent cells. In the attempt to reconcile observations made by different authors, we propose a unifying picture that could represent a starting point for future experiments.
Collapse
Affiliation(s)
- Ranieri Cancedda
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italy
| | - Maddalena Mastrogiacomo
- Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università Degli Studi di Genova, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
6
|
Lin SL, Lee W, Liu SP, Chang YW, Jeng LB, Shyu WC. Novel Programmed Death Ligand 1-AKT-engineered Mesenchymal Stem Cells Promote Neuroplasticity to Target Stroke Therapy. Mol Neurobiol 2024; 61:3819-3835. [PMID: 38030932 DOI: 10.1007/s12035-023-03779-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023]
Abstract
Although tissue plasminogen activator (t-PA) and endovascular thrombectomy are well-established treatments for acute ischemic stroke, over half of patients with stroke remain disabled for a long time. Thus, a significant unmet need exists to develop an effective strategy for treating acute stroke. We developed a combination of programmed cell death-ligand 1 (PD-L1) and AKT-modified umbilical cord mesenchymal stem cells (UMSC-PD-L1-AKT) implanted through intravenous (IV) and intracarotid (IA) routes to enhance therapeutic efficacy in a murine stroke model for overcoming the hypoxic environment of the ischemic brain, to prolong stem cell survival, and to attenuate systemic inflammation to protect neuroglial cells from ischemic injury. Higher cellular proliferation and survival upon exposure to toxic agents were observed in UMSC-PD-L1-AKT cells than in UMSCs in vitro. Moreover, increased attenuation of CFSE+ cell proliferation and increased survival of primary cortical cells were verified by the interaction with UMSC-PD-L1-AKT. Consistently, dual-route administration (IV + IA) of UMSC-PD-L1-AKT resulted in a significant reduction in infarction volume and improvement of neurological dysfunction in a stroke model. Furthermore, enhancing CD8+CD122+IL-10+ T-regulatory (Treg) cells and reducing CD11b+CD80+ microglial/macrophages and CD3+CD8+TNF-α+ and CD3+CD8+ IFN-α+ cytotoxic T cells induced an anti-inflammatory microenvironment to protect neuroglial cells in the ischemic brain. Collectively, therapeutic intervention using UMSC-PD-L1-AKT could provide a niche for inducing neuroplastic regeneration in brains after stroke.
Collapse
Affiliation(s)
- Syuan-Ling Lin
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Wei Lee
- Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Shih-Ping Liu
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Yi-Wen Chang
- Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan.
| | - Long-Bin Jeng
- Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan.
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan.
| | - Woei-Cherng Shyu
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan.
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan.
- Neuroscience and Brain Disease Center and New Drug Development Center, China Medical University, Taichung, Taiwan.
- Department of Occupational Therapy, Asia University, Taichung, Taiwan.
| |
Collapse
|
7
|
Deng D, Zhang Y, Tang B, Zhang Z. Sources and applications of endothelial seed cells: a review. Stem Cell Res Ther 2024; 15:175. [PMID: 38886767 PMCID: PMC11184868 DOI: 10.1186/s13287-024-03773-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/26/2024] [Indexed: 06/20/2024] Open
Abstract
Endothelial cells (ECs) are widely used as donor cells in tissue engineering, organoid vascularization, and in vitro microvascular model development. ECs are invaluable tools for disease modeling and drug screening in fundamental research. When treating ischemic diseases, EC engraftment facilitates the restoration of damaged blood vessels, enhancing therapeutic outcomes. This article presents a comprehensive overview of the current sources of ECs, which encompass stem/progenitor cells, primary ECs, cell lineage conversion, and ECs derived from other cellular sources, provides insights into their characteristics, potential applications, discusses challenges, and explores strategies to mitigate these issues. The primary aim is to serve as a reference for selecting suitable EC sources for preclinical research and promote the translation of basic research into clinical applications.
Collapse
Affiliation(s)
- Dan Deng
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yu Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Bo Tang
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Zhihui Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
8
|
Aprile D, Patrone D, Peluso G, Galderisi U. Multipotent/pluripotent stem cell populations in stromal tissues and peripheral blood: exploring diversity, potential, and therapeutic applications. Stem Cell Res Ther 2024; 15:139. [PMID: 38735988 PMCID: PMC11089765 DOI: 10.1186/s13287-024-03752-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
The concept of "stemness" incorporates the molecular mechanisms that regulate the unlimited self-regenerative potential typical of undifferentiated primitive cells. These cells possess the unique ability to navigate the cell cycle, transitioning in and out of the quiescent G0 phase, and hold the capacity to generate diverse cell phenotypes. Stem cells, as undifferentiated precursors endow with extraordinary regenerative capabilities, exhibit a heterogeneous and tissue-specific distribution throughout the human body. The identification and characterization of distinct stem cell populations across various tissues have revolutionized our understanding of tissue homeostasis and regeneration. From the hematopoietic to the nervous and musculoskeletal systems, the presence of tissue-specific stem cells underlines the complex adaptability of multicellular organisms. Recent investigations have revealed a diverse cohort of non-hematopoietic stem cells (non-HSC), primarily within bone marrow and other stromal tissue, alongside established hematopoietic stem cells (HSC). Among these non-HSC, a rare subset exhibits pluripotent characteristics. In vitro and in vivo studies have demonstrated the remarkable differentiation potential of these putative stem cells, known by various names including multipotent adult progenitor cells (MAPC), marrow-isolated adult multilineage inducible cells (MIAMI), small blood stem cells (SBSC), very small embryonic-like stem cells (VSELs), and multilineage differentiating stress enduring cells (MUSE). The diverse nomenclatures assigned to these primitive stem cell populations may arise from different origins or varied experimental methodologies. This review aims to present a comprehensive comparison of various subpopulations of multipotent/pluripotent stem cells derived from stromal tissues. By analysing isolation techniques and surface marker expression associated with these populations, we aim to delineate the similarities and distinctions among stromal tissue-derived stem cells. Understanding the nuances of these tissue-specific stem cells is critical for unlocking their therapeutic potential and advancing regenerative medicine. The future of stem cells research should prioritize the standardization of methodologies and collaborative investigations in shared laboratory environments. This approach could mitigate variability in research outcomes and foster scientific partnerships to fully exploit the therapeutic potential of pluripotent stem cells.
Collapse
Affiliation(s)
- Domenico Aprile
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Deanira Patrone
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy
| | - Gianfranco Peluso
- Faculty of Medicine and Surgery, Saint Camillus International, University of Health Sciences, Rome, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, Naples, Italy.
- Genome and Stem Cell Center (GENKÖK), Erciyes University, Kayseri, Turkey.
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine Temple University, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Delcroix GJR, Hackett A, Schiller PC, Temple HT. Characterization of three washing/decellularization procedures for the production of bioactive human micronized neural tissue (hMINT). Cell Tissue Bank 2023; 24:693-703. [PMID: 36854877 DOI: 10.1007/s10561-023-10075-3] [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: 10/25/2022] [Accepted: 01/29/2023] [Indexed: 03/02/2023]
Abstract
BACKGROUND We developed a novel, injectable and decellularized human peripheral nerve-based scaffold, named Micronized Human Neural Tissue (hMINT), designed to be used as a supportive matrix for stem cell transplantation in the context of spinal cord injury (SCI). MATERIALS AND METHODS Human donated sciatic nerves were micronized at liquid nitrogen temperature prior to decellularization using 3 different procedures of various harshness. hMINT were characterized in terms of particle size, DNA, sulfated glycosaminoglycans (sGAG) and growth factors content. To test the biocompatibility and bioactivity of the various preparations, we used a type of mesenchymal stromal cells (MSCs), termed MIAMI cells, which were placed in contact with hMINT to monitor cell attachment by confocal microscopy and gene expression by RT-qPCR in vitro. RESULTS The content of DNA, sGAG and growth factors left in the product after processing was highly dependent on the decellularization procedure used. We demonstrated that hMINT are biocompatible and promoted the attachment and long-term survival of MIAMI cells in vitro. Finally, combination with hMINT increased MIAMI cells mRNA expression of pro-survival and anti-inflammatory factors. Importantly, the strongest bioactivity on MIAMI cells was observed with the hMINT decellularized using the mildest decellularization procedure, therefore emphasizing the importance of achieving an adequate decellularization without losing the hMINT's bioactivity. PERSPECTIVES AND CLINICAL SIGNIFICANCE The capacity of hMINT/stem cells to facilitate protection of injured neural tissue, promote axon re-growth and improve functional recovery will be tested in an animal model of SCI and other neurodegenerative disorders in the future.
Collapse
Affiliation(s)
- Gaëtan J-R Delcroix
- College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA.
| | - Amber Hackett
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Paul C Schiller
- Geriatric Research Education and Clinical Center, Miami VA Healthcare System, Miami, FL, USA
| | | |
Collapse
|
10
|
Kidwai FK, Canalis E, Robey PG. Induced pluripotent stem cell technology in bone biology. Bone 2023; 172:116760. [PMID: 37028583 PMCID: PMC10228209 DOI: 10.1016/j.bone.2023.116760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/09/2023]
Abstract
Technologies on the development and differentiation of human induced pluripotent stem cells (hiPSCs) are rapidly improving, and have been applied to create cell types relevant to the bone field. Differentiation protocols to form bona fide bone-forming cells from iPSCs are available, and can be used to probe details of differentiation and function in depth. When applied to iPSCs bearing disease-causing mutations, the pathogenetic mechanisms of diseases of the skeleton can be elucidated, along with the development of novel therapeutics. These cells can also be used for development of cell therapies for cell and tissue replacement.
Collapse
Affiliation(s)
- Fahad K Kidwai
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, United States of America
| | - Ernesto Canalis
- Center for Skeletal Research, Orthopedic Surgery and Medicine, UConn Musculoskeletal Institute, UConn Health, Farmington, CT 06030-4037, United States of America
| | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, United States of America.
| |
Collapse
|
11
|
Asadpour A, Yahaya BH, Bicknell K, Cottrell GS, Widera D. Uncovering the gray zone: mapping the global landscape of direct-to-consumer businesses offering interventions based on secretomes, extracellular vesicles, and exosomes. Stem Cell Res Ther 2023; 14:111. [PMID: 37138298 PMCID: PMC10156419 DOI: 10.1186/s13287-023-03335-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/10/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND The last decade has seen a significant increase in media attention, industrial growth, and patient interest in stem cell-based interventions. This led to a rise in direct-to-consumer businesses offering stem cell "therapies" for multiple indications with little evidence of safety and efficacy. In parallel, the use of stem cell secretomes as a substitute for stem cell transplantation has become an increasing trend in regenerative medicine with multiple clinical trials currently assessing their efficacy and safety profile. As a result, multiple businesses and private clinics have now started to exploit this situation and are offering secretome-based interventions despite the lack of supporting data. This poses significant risks for the patients and could lead to a credibility crisis in the field. METHODS Internet searches were used to locate clinics marketing and selling interventions based on stem cell secretomes, exosomes, or extracellular vesicles. Data were extracted from websites with a particular focus on the global distribution of the businesses, the cellular source of the secretome, the indication spectrum, and the pricing of the provided services. Lastly, the types of evidence used on the websites of the businesses to market their services were extracted. RESULTS Overall, 114 companies market secretome-based therapies in 28 countries. The vast majority of the interventions are based on allogenic stem cells from undisclosed cellular sources and skin care is the most marketed indication. The price range is USD99-20,000 depending on the indication. CONCLUSIONS The direct-to-consumer industry for secretome-based therapies appears to be primed for growth in the absence of appropriate regulatory frameworks and guidelines. We conclude that such business activity requires tight regulations and monitoring by the respective national regulatory bodies to prevent patients from being conned and more importantly from being put at risk.
Collapse
Affiliation(s)
- Atiyeh Asadpour
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, RG6 6AP, UK
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Sains@Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Katrina Bicknell
- School of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, RG6 6AP, UK
| | - Graeme S Cottrell
- Cellular and Molecular Neuroscience, School of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, RG6 6AP, UK
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, RG6 6AP, UK.
| |
Collapse
|
12
|
Stem cell-nanomedicine system as a theranostic bio-gadolinium agent for targeted neutron capture cancer therapy. Nat Commun 2023; 14:285. [PMID: 36650171 PMCID: PMC9845336 DOI: 10.1038/s41467-023-35935-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
The potential clinical application of gadolinium-neutron capture therapy (Gd-NCT) for glioblastoma multiforme (GBM) treatment has been compromised by the fast clearance and nonspecific biodistribution of gadolinium-based agents. We have developed a stem cell-nanoparticle system (SNS) to actively target GBM for advanced Gd-NCT by magnetizing umbilical cord mesenchymal stem cells (UMSCs) using gadodiamide-concealed magnetic nanoparticles (Gd-FPFNP). Nanoformulated gadodiamide shielded by a dense surface composed of fucoidan and polyvinyl alcohol demonstrates enhanced cellular association and biocompatibility in UMSCs. The SNS preserves the ability of UMSCs to actively penetrate the blood brain barrier and home to GBM and, when magnetically navigates by an external magnetic field, an 8-fold increase in tumor-to-blood ratio is achieved compared with clinical data. In an orthotopic GBM-bearing rat model, using a single dose of irradiation and an ultra-low gadolinium dose (200 μg kg-1), SNS significantly attenuates GBM progression without inducing safety issues, prolonging median survival 2.5-fold compared to free gadodiamide. The SNS is a cell-based delivery system that integrates the strengths of cell therapy and nanotechnology, which provides an alternative strategy for the treatment of brain diseases.
Collapse
|
13
|
Borda M, Aquino JB, Mazzone GL. Cell-based experimental strategies for myelin repair in multiple sclerosis. J Neurosci Res 2023; 101:86-111. [PMID: 36164729 DOI: 10.1002/jnr.25129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/21/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disorder of the central nervous system (CNS), diagnosed at a mean age of 32 years. CNS glia are crucial players in the onset of MS, primarily involving astrocytes and microglia that can cause/allow massive oligodendroglial cells death, without immune cell infiltration. Current therapeutic approaches are aimed at modulating inflammatory reactions during relapsing episodes, but lack the ability to induce very significant repair mechanisms. In this review article, different experimental approaches based mainly on the application of different cell types as therapeutic strategies applied for the induction of myelin repair and/or the amelioration of the disease are discussed. Regarding this issue, different cell sources were applied in various experimental models of MS, with different results, both in significant improvements in remyelination and the reduction of neuroinflammation and glial activation, or in neuroprotection. All cell types tested have advantages and disadvantages, which makes it difficult to choose a better option for therapeutic application in MS. New strategies combining cell-based treatment with other applications would result in further improvements and would be good candidates for MS cell therapy and myelin repair.
Collapse
Affiliation(s)
- Maximiliano Borda
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina
| | - Jorge B Aquino
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina.,CONICET, Comisión Nacional de Investigaciones Científicas y Técnicas
| | - Graciela L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina.,CONICET, Comisión Nacional de Investigaciones Científicas y Técnicas
| |
Collapse
|
14
|
Frederick RM, Sarfani S, Chiu CY, Hayes T, Bettin C, Grear B, Richardson D, Murphy GA. Comparing Rates of Fusion and Time to Fusion in Viable Cellular Allograft and Autograft. Foot Ankle Int 2022; 43:1548-1553. [PMID: 36036537 DOI: 10.1177/10711007221119164] [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] [Indexed: 02/01/2023]
Abstract
BACKGROUND Autograft or allograft frequently are used to enhance bone union in foot and ankle surgery. Viable cellular bone allograft uses viable cells and bone scaffolding in a gel base, but uncertainty remains around allograft's greater efficacy than autograft regarding rates of fusion (ROF) and time to fusion (TTF). METHODS Autograft, viable cellular allograft, and viable cellular allograft with autograft were compared in 199 forefoot, midfoot, and hindfoot arthrodeses performed over a 6-year period. Data collected from electronic medical records and radiographs were analyzed to determine ROF and TTF as well as rates of revision surgery for delayed or nonunion and compared among groups. RESULTS Eighty-seven patients comprised the autograft group, 81 the allograft group, and 31 the combined group. No significant differences were noted in patient demographics among the groups. No statistically significant differences in ROF were noted among the 3 groups, with 86% (75 of 87) fusion in the autograft group, 93% (75 of 81) in the allograft group, and 84% (26 of 31) in the combined group (P = .20). After conducting a multivariate analysis, we found no statistically significant difference for allograft or combined graft on TTF (P = .1379 and .2311, respectively). No significant difference was found in rate of revision surgery for nonunion, which was 1.2% (1 of 81) in the allograft group, 3.4% (3 of 87) in the autograft group, and 6.5% (2 of 31) in the combined group (P = .3). CONCLUSION No significant difference was found in ROF, TTF, or rate of revision surgery when comparing viable cellular allograft to autograft or combined allograft-autograft. Viable cellular allograft may be a reasonable alternative to the gold standard of autograft and should be considered an option in patients undergoing arthrodesis in foot and ankle surgery. LEVEL OF EVIDENCE Level III, therapeutic.
Collapse
Affiliation(s)
- Robert M Frederick
- University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA
| | | | - Chi-Yang Chiu
- University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA
| | - Tristan Hayes
- University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA
| | - Clayton Bettin
- University of Tennessee-Campbell Clinic Department of Orthopaedic Surgery, Memphis, TN, USA
| | - Benjamin Grear
- University of Tennessee-Campbell Clinic Department of Orthopaedic Surgery, Memphis, TN, USA
| | - David Richardson
- University of Tennessee-Campbell Clinic Department of Orthopaedic Surgery, Memphis, TN, USA
| | - G Andrew Murphy
- University of Tennessee-Campbell Clinic Department of Orthopaedic Surgery, Memphis, TN, USA
| |
Collapse
|
15
|
Quesenberry PJ, Wen S, Goldberg LR, Dooner MS. The universal stem cell. Leukemia 2022; 36:2784-2792. [PMID: 36307485 PMCID: PMC9712109 DOI: 10.1038/s41375-022-01715-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/26/2022] [Accepted: 09/22/2022] [Indexed: 11/08/2022]
Abstract
Current dogma is that there exists a hematopoietic pluripotent stem cell, resident in the marrow, which is quiescent, but with tremendous proliferative and differentiative potential. Furthermore, the hematopoietic system is essentially hierarchical with progressive differentiation from the pluripotent stem cells to different classes of hematopoietic cells. However, results summarized here indicate that the marrow pluripotent hematopoietic stem cell is actively cycling and thus continually changing phenotype. As it progresses through cell cycle differentiation potential changes as illustrated by sequential changes in surface expression of B220 and GR-1 epitopes. Further data indicated that the potential of purified hematopoietic stem cells extends to multiple other non-hematopoietic cells. It appears that marrow stem cells will give rise to epithelial pulmonary cells at certain points in cell cycle. Thus, it appears that the marrow "hematopoietic" stem cell is also a stem cell for other non-hematopoietic tissues. These observations give rise to the concept of a universal stem cell. The marrow stem cell is not limited to hematopoiesis and its differentiation potential continually changes as it transits cell cycle. Thus, there is a universal stem cell in the marrow which alters its differentiation potential as it progresses through cell cycle. This potential is expressed when it resides in tissues compatible with its differentiation potential, at a particular point in cell cycle transit, or when it interacts with vesicles from that tissue.
Collapse
Affiliation(s)
- Peter J Quesenberry
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA.
| | - Sicheng Wen
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| | - Laura R Goldberg
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Mark S Dooner
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| |
Collapse
|
16
|
Stem Cells in the Tumor Immune Microenvironment -Part of the Cure or Part of the Disease? Ontogeny and Dichotomy of Stem and Immune Cells has Led to better Understanding. Stem Cell Rev Rep 2022; 18:2549-2565. [PMID: 35841518 DOI: 10.1007/s12015-022-10428-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 10/17/2022]
Abstract
Stem cells are at the basis of tissue homeostasis, hematopoiesis and various regenerative processes. Epigenetic changes in their somatically imprinted genes, prolonged exposure to mutagens/carcinogens or alteration of their niche can lead to the development of an enabling environment for tumor growth and progression. The involvement of stem cells in both health and disease becomes even more compelling with ontogeny as embryonic and extraembryonic stem cells which persist into adulthood in well established and specific niche may have distinct implications in tumorigenesis. Immune surveillance plays an important role in this interplay since the response of immune cells toward the oncogenic process can range from reactivity to placidity and even complicity, being orchestrated by intercellular molecular dialogues with the other key players of the tumor microenvironment. With the current understanding that every developing and adult tissue contains inherent stem and progenitor cells, in this manuscript we review the most relevant interactions carried out between the stem cells, tumor cells and immune cells in a bottom-up incursion through the tumor microenvironment beginning from the perivascular niche and going through the tumoral parenchyma and the related stroma. With the exploitation of various factors that influence the behavior of immune effectors toward stem cells and other resting cells in their niche, new therapeutic strategies to tackle the polarization of immune effectors toward a more immunogenic phenotype may arise.
Collapse
|
17
|
Pattappa G, Reischl F, Jahns J, Schewior R, Lang S, Zellner J, Johnstone B, Docheva D, Angele P. Fibronectin Adherent Cell Populations Derived From Avascular and Vascular Regions of the Meniscus Have Enhanced Clonogenicity and Differentiation Potential Under Physioxia. Front Bioeng Biotechnol 2022; 9:789621. [PMID: 35155405 PMCID: PMC8831898 DOI: 10.3389/fbioe.2021.789621] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
The meniscus is composed of an avascular inner region and vascular outer region. The vascular region has been shown to contain a progenitor population with multilineage differentiation capacity. Strategies facilitating the isolation and propagation of these progenitors can be used to develop cell-based meniscal therapies. Differential adhesion to fibronectin has been used to isolate progenitor populations from cartilage, while low oxygen or physioxia (2% oxygen) enhances the meniscal phenotype. This study aimed to isolate progenitor populations from the avascular and vascular meniscus using differential fibronectin adherence and examine their clonogenicity and differentiation potential under hyperoxia (20% oxygen) and physioxia (2% oxygen). Human vascular and avascular meniscus cells were seeded onto fibronectin-coated dishes for a short period and monitored for colony formation under either hyperoxia or physioxia. Non-fibronectin adherent meniscus cells were also expanded under both oxygen tension. Individual fibronectin adherent colonies were isolated and further expanded, until approximately ten population doublings (passage 3), whereby they underwent chondrogenic, osteogenic, and adipogenic differentiation. Physioxia enhances clonogenicity of vascular and avascular meniscus cells on plastic or fibronectin-coated plates. Combined differential fibronectin adhesion and physioxia isolated a progenitor population from both meniscus regions with trilineage differentiation potential compared to equivalent hyperoxia progenitors. Physioxia isolated progenitors had a significantly enhanced meniscus matrix content without the presence of collagen X. These results demonstrate that combined physioxia and fibronectin adherence can isolate and propagate a meniscus progenitor population that can potentially be used to treat meniscal tears or defects.
Collapse
Affiliation(s)
- Girish Pattappa
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- *Correspondence: Girish Pattappa,
| | - Franziska Reischl
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Judith Jahns
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Ruth Schewior
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Siegmund Lang
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Johannes Zellner
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Sporthopaedicum Regensburg, Regensburg, Germany
| | - Brian Johnstone
- Department of Orthopaedics and Rehabilitation, Oregon Health and Science University, Portland, OR, United States
| | - Denitsa Docheva
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Wurzburg, Wurzburg, Germany
| | - Peter Angele
- Laboratory for Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
- Sporthopaedicum Regensburg, Regensburg, Germany
| |
Collapse
|
18
|
Ye Y, Zhao X, Xu Y, Yu J. Hypoxia-Inducible Non-coding RNAs in Mesenchymal Stem Cell Fate and Regeneration. FRONTIERS IN DENTAL MEDICINE 2021. [DOI: 10.3389/fdmed.2021.799716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can differentiate into multiple cell lines, which makes them an important source of cells for tissue engineering applications. They are defined by the capability to renew themselves and maintain pluripotency. This ability is modulated by the balance between complex cues from cellular microenvironment. Self-renewal and differentiation abilities are regulated by particular microenvironmental signals. Oxygen is considered to be an important part of cell microenvironment, which not only acts as a metabolic substrate but also a signal molecule. It has been proved that MSCs are hypoxic in the physiological environment. Signals from MSCs' microenvironment or niche which means the anatomical location of the MSCs, maintain the final properties of MSCs. Physiological conditions like oxygen tension are deemed to be a significant part of the mesenchymal stem cell niche, and have been proved to be involved in modulating embryonic and adult MSCs. Non-coding RNAs (ncRNAs), which play a key role in cell signal transduction, transcription and translation of genes, have been widely concerned as epigenetic regulators in a great deal of tissues. With the rapid development of bioinformatics analysis tools and high-throughput RNA sequencing technology, more and more evidences show that ncRNAs play a key role in tissue regeneration. It shows potential as a biomarker of MSC differentiation. In this paper, we reviewed the physiological correlation of hypoxia as a unique environmental parameter which is conducive to MSC expansion and maintenance, discussed the correlation of tissue engineering, and summarized the influence of hypoxia related ncRNAs on MSCs' fate and regeneration. This review will provide reference for future research of MSCs' regeneration.
Collapse
|
19
|
Kulus M, Sibiak R, Stefańska K, Zdun M, Wieczorkiewicz M, Piotrowska-Kempisty H, Jaśkowski JM, Bukowska D, Ratajczak K, Zabel M, Mozdziak P, Kempisty B. Mesenchymal Stem/Stromal Cells Derived from Human and Animal Perinatal Tissues-Origins, Characteristics, Signaling Pathways, and Clinical Trials. Cells 2021; 10:cells10123278. [PMID: 34943786 PMCID: PMC8699543 DOI: 10.3390/cells10123278] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/13/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are currently one of the most extensively researched fields due to their promising opportunity for use in regenerative medicine. There are many sources of MSCs, of which cells of perinatal origin appear to be an invaluable pool. Compared to embryonic stem cells, they are devoid of ethical conflicts because they are derived from tissues surrounding the fetus and can be safely recovered from medical waste after delivery. Additionally, perinatal MSCs exhibit better self-renewal and differentiation properties than those derived from adult tissues. It is important to consider the anatomy of perinatal tissues and the general description of MSCs, including their isolation, differentiation, and characterization of different types of perinatal MSCs from both animals and humans (placenta, umbilical cord, amniotic fluid). Ultimately, signaling pathways are essential to consider regarding the clinical applications of MSCs. It is important to consider the origin of these cells, referring to the anatomical structure of the organs of origin, when describing the general and specific characteristics of the different types of MSCs as well as the pathways involved in differentiation.
Collapse
Affiliation(s)
- Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
| | - Rafał Sibiak
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
- Division of Reproduction, Department of Obstetrics, Gynecology, and Gynecologic Oncology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Katarzyna Stefańska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
| | - Maciej Zdun
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
| | - Maria Wieczorkiewicz
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
| | - Hanna Piotrowska-Kempisty
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
| | - Jędrzej M. Jaśkowski
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.M.J.); (D.B.)
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.M.J.); (D.B.)
| | - Kornel Ratajczak
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
| | - Maciej Zabel
- Division of Anatomy and Histology, University of Zielona Gora, 65-046 Zielona Gora, Poland;
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Correspondence:
| |
Collapse
|
20
|
Wartalski K, Gorczyca G, Wiater J, Tabarowski Z, Duda M. Porcine ovarian cortex-derived putative stem cells can differentiate into endothelial cells in vitro. Histochem Cell Biol 2021; 156:349-362. [PMID: 34269874 PMCID: PMC8550686 DOI: 10.1007/s00418-021-02016-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2021] [Indexed: 12/19/2022]
Abstract
Endothelial cells (ECs), the primary component of the vasculature, play a crucial role in neovascularization. However, the number of endogenous ECs is inadequate for both experimental purposes and clinical applications. Porcine ovarian putative stem cells (poPSCs), although not pluripotent, are characterized by great plasticity. Therefore, this study aimed to investigate whether poPSCs have the potential to differentiate into cells of endothelial lineage. poPSCs were immunomagnetically isolated from postnatal pig ovaries based on the presence of SSEA-4 protein. Expression of mesenchymal stem cells (MSCs) markers after pre-culture, both at the level of mRNA: ITGB1, THY, and ENG and corresponding protein: CD29, CD90, and CD105 were significantly higher compared to the control ovarian cortex cells. To differentiate poPSCs into ECs, inducing medium containing vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), insulin-like growth factor (IGF), epidermal growth factor (EGF), ascorbic acid, and heparin was applied. After 14 days, poPSC differentiation into ECs was confirmed by immunofluorescence staining for vascular endothelial cadherin (VECad) and vascular endothelial growth factor receptor-2 (VEGFR-2). Semi-quantitative WB analysis of these proteins confirmed their high abundance. Additionally, qRT-PCR showed that mRNA expression of corresponding marker genes: CDH5, KDR was significantly higher compared with undifferentiated poPSCs. Finally, EC functional status was confirmed by the migration test that revealed that they were capable of positive chemotaxis, while tube formation assay demonstrated their ability to develop capillary networks. In conclusion, our results provided evidence that poPSCs may constitute the MSC population in the ovary and confirmed that they might be a potential source of ECs for tissue engineering.
Collapse
Affiliation(s)
- Kamil Wartalski
- Faculty of Medicine, Department of Histology, Jagiellonian University Medical College, Kopernika 7 Street, 31-034, Krakow, Poland
| | - Gabriela Gorczyca
- Faculty of Biology, Institute of Zoology and Biomedical Research, Department of Endocrinology, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387, Krakow, Poland
| | - Jerzy Wiater
- Faculty of Medicine, Department of Histology, Jagiellonian University Medical College, Kopernika 7 Street, 31-034, Krakow, Poland
| | - Zbigniew Tabarowski
- Faculty of Biology, Institute of Zoology and Biomedical Research, Department of Experimental Hematology, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387, Krakow, Poland
| | - Małgorzata Duda
- Faculty of Biology, Institute of Zoology and Biomedical Research, Department of Endocrinology, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387, Krakow, Poland.
| |
Collapse
|
21
|
Aquino JB, Sierra R, Montaldo LA. Diverse cellular origins of adult blood vascular endothelial cells. Dev Biol 2021; 477:117-132. [PMID: 34048734 DOI: 10.1016/j.ydbio.2021.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022]
Abstract
During embryonic stages, vascular endothelial cells (ECs) originate from the mesoderm, at specific extraembryonic and embryonic regions, through a process called vasculogenesis. In the adult, EC renewal/replacement mostly depend on local resident ECs or endothelial progenitor cells (EPCs). Nevertheless, contribution from circulating ECs/EPCs was also reported. In addition, cells lacking from EC/EPC markers with in vitro extended plasticity were shown to originate endothelial-like cells (ELCs). Most of these cells consist of mesenchymal stromal progenitors, which would eventually get mobilized from the bone marrow after injury. Based on that, current knowledge on different mouse and human bone marrow stromal cell (BM-SC) subpopulations, able to contribute with mesenchymal stromal/stem cells (MSCs), is herein reviewed. Such analyses underline an unexpected heterogeneity among sinusoidal LepR+ stromal/CAR cells. For instance, in a recent report a subgroup of LepR+ stromal/CAR progenitors, which express GLAST and is traced in Wnt1Cre;R26RTom mice, was found to contribute with ELCs in vivo. These GLAST + Wnt1+ BM-SCs were shown to get mobilized to the peripheral blood and to contribute with liver regeneration. Other sources of ELCs, such as adipose, neural and dental pulp tissues, were also published. Finally, mechanisms likely involved in the enhanced cellular plasticity properties of bone marrow/adipose tissue stromal cells, able to originate ELCs, are assessed. In the future, strategies to analyze the in vivo expression profile of stromal cells, with MSC properties, in combination with screening of active genomic regions at the single cell-level, during early postnatal development and/or after injury, will likely help understanding properties of these ELC sources.
Collapse
Affiliation(s)
- Jorge B Aquino
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina.
| | - Romina Sierra
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina
| | - Laura A Montaldo
- CONICET-Universidad Austral, Instituto de Investigaciones en Medicina Traslacional (IIMT), Developmental Biology & Regenerative Medicine Laboratory, Argentina
| |
Collapse
|
22
|
Aprile D, Alessio N, Demirsoy IH, Squillaro T, Peluso G, Di Bernardo G, Galderisi U. MUSE Stem Cells Can Be Isolated from Stromal Compartment of Mouse Bone Marrow, Adipose Tissue, and Ear Connective Tissue: A Comparative Study of Their In Vitro Properties. Cells 2021; 10:761. [PMID: 33808472 PMCID: PMC8065981 DOI: 10.3390/cells10040761] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 01/10/2023] Open
Abstract
The cells present in the stromal compartment of many tissues are a heterogeneous population containing stem cells, progenitor cells, fibroblasts, and other stromal cells. A SSEA3(+) cell subpopulation isolated from human stromal compartments showed stem cell properties. These cells, known as multilineage-differentiating stress-enduring (MUSE) cells, are capable of resisting stress and possess an excellent ability to repair DNA damage. We isolated MUSE cells from different mouse stromal compartments, such as those present in bone marrow, subcutaneous white adipose tissue, and ear connective tissue. These cells showed overlapping in vitro biological properties. The mouse MUSE cells were positive for stemness markers such as SOX2, OCT3/4, and NANOG. They also expressed TERT, the catalytic telomerase subunit. The mouse MUSE cells showed spontaneous commitment to differentiation in meso/ecto/endodermal derivatives. The demonstration that multilineage stem cells can be isolated from an animal model, such as the mouse, could offer a valid alternative to the use of other stem cells for disease studies and envisage of cellular therapies.
Collapse
Affiliation(s)
- Domenico Aprile
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (D.A.); (N.A.); (I.H.D.); (T.S.); (G.D.B.)
| | - Nicola Alessio
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (D.A.); (N.A.); (I.H.D.); (T.S.); (G.D.B.)
| | - Ibrahim H. Demirsoy
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (D.A.); (N.A.); (I.H.D.); (T.S.); (G.D.B.)
| | - Tiziana Squillaro
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (D.A.); (N.A.); (I.H.D.); (T.S.); (G.D.B.)
| | | | - Giovanni Di Bernardo
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (D.A.); (N.A.); (I.H.D.); (T.S.); (G.D.B.)
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli Campania University, 80138 Naples, Italy; (D.A.); (N.A.); (I.H.D.); (T.S.); (G.D.B.)
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
- Genome and Stem Cell Center (GENKOK), Erciyes University, 38280 Kayseri, Turkey
| |
Collapse
|
23
|
Detriche G, Guerin CL, Gendron N, Mirault T, Smadja DM. Do Endothelial Colony-forming Cells Come From Bone Marrow or Vessels/VSELs? Stem Cell Rev Rep 2021; 17:1500-1502. [PMID: 33651335 DOI: 10.1007/s12015-021-10140-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 10/22/2022]
Affiliation(s)
- Grégoire Detriche
- Innovative Therapies in Hemostasis, Université de Paris, INSERM, F-75006, Paris, France.,Vascular Medicine Department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP- CUP), F-75015, Paris, France
| | - Coralie L Guerin
- Innovative Therapies in Hemostasis, Université de Paris, INSERM, F-75006, Paris, France.,Institut Curie, 75006, Paris, France
| | - Nicolas Gendron
- Innovative Therapies in Hemostasis, Université de Paris, INSERM, F-75006, Paris, France.,Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), F-75015, Paris, France
| | - Tristan Mirault
- Vascular Medicine Department, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP- CUP), F-75015, Paris, France.,Université de Paris, PARCC, INSERM, F-75015, Paris, France
| | - David M Smadja
- Innovative Therapies in Hemostasis, Université de Paris, INSERM, F-75006, Paris, France. .,Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), F-75015, Paris, France.
| |
Collapse
|
24
|
Yunir E, Kurniawan F, Rezaprasga E, Wijaya IP, Suroyo I, Matondang S, Irawan C, Soewondo P. Autologous Bone-Marrow vs. Peripheral Blood Mononuclear Cells Therapy for Peripheral Artery Disease in Diabetic Patients. Int J Stem Cells 2021; 14:21-32. [PMID: 33377454 PMCID: PMC7904521 DOI: 10.15283/ijsc20088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 01/09/2023] Open
Abstract
Diabetes mellitus (DM) remains one of the most important risk factors for peripheral artery disease (PAD), with approximately 20% of DM patients older than 40 years old are affected with PAD. The current standard management for severe PAD is endovascular intervention with or without surgical bypass. Unfortunately, up to 40% of patients are unable to undergo these revascularization therapies due to excessive surgical risk or adverse vascular side effects. Stem cell therapy has emerged as a novel therapeutic strategy for these ‘no-option’ patients. Several types of stem cells are utilized for PAD therapy, including bone marrow mononuclear cells (BMMNC) and peripheral blood mononuclear cells (PBMNC). Many studies have reported the safety of BMMNC and PBMNC, as well as its efficacy in reducing ischemic pain, ulcer size, pain-free walking distance, ankle-brachial index (ABI), and transcutaneous oxygen pressure (TcPO2). However, the capacity to establish the efficacy of reducing major amputation rates, amputation free survival, and all-cause mortality is limited, as shown by several randomized placebo-controlled trials. The present literature review will focus on comparing safety and efficacy between BMMNC and PBMNC as cell-based management in diabetic patients with PAD who are not suitable for revascularization therapy.
Collapse
Affiliation(s)
- Em Yunir
- Department of Internal Medicine, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Metabolic Disorder, Cardiovascular, and Aging Cluster, Indonesian Medical Education and Research Institute, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Farid Kurniawan
- Department of Internal Medicine, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Metabolic Disorder, Cardiovascular, and Aging Cluster, Indonesian Medical Education and Research Institute, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Edo Rezaprasga
- Department of Internal Medicine, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Metabolic Disorder, Cardiovascular, and Aging Cluster, Indonesian Medical Education and Research Institute, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Ika Prasetya Wijaya
- Department of Internal Medicine, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Indrati Suroyo
- Department of Radiology, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Sahat Matondang
- Department of Radiology, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Cosphiadi Irawan
- Department of Internal Medicine, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Pradana Soewondo
- Department of Internal Medicine, Dr. Cipto Mangunkusumo National General Hospital, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Metabolic Disorder, Cardiovascular, and Aging Cluster, Indonesian Medical Education and Research Institute, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
25
|
Levoux J, Prola A, Lafuste P, Gervais M, Chevallier N, Koumaiha Z, Kefi K, Braud L, Schmitt A, Yacia A, Schirmann A, Hersant B, Sid-Ahmed M, Ben Larbi S, Komrskova K, Rohlena J, Relaix F, Neuzil J, Rodriguez AM. Platelets Facilitate the Wound-Healing Capability of Mesenchymal Stem Cells by Mitochondrial Transfer and Metabolic Reprogramming. Cell Metab 2021; 33:283-299.e9. [PMID: 33400911 DOI: 10.1016/j.cmet.2020.12.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/31/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Platelets are known to enhance the wound-healing activity of mesenchymal stem cells (MSCs). However, the mechanism by which platelets improve the therapeutic potential of MSCs has not been elucidated. Here, we provide evidence that, upon their activation, platelets transfer respiratory-competent mitochondria to MSCs primarily via dynamin-dependent clathrin-mediated endocytosis. We found that this process enhances the therapeutic efficacy of MSCs following their engraftment in several mouse models of tissue injury, including full-thickness cutaneous wound and dystrophic skeletal muscle. By combining in vitro and in vivo experiments, we demonstrate that platelet-derived mitochondria promote the pro-angiogenic activity of MSCs via their metabolic remodeling. Notably, we show that activation of the de novo fatty acid synthesis pathway is required for increased secretion of pro-angiogenic factors by platelet-preconditioned MSCs. These results reveal a new mechanism by which platelets potentiate MSC properties and underline the importance of testing platelet mitochondria quality prior to their clinical use.
Collapse
Affiliation(s)
- Jennyfer Levoux
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Alexandre Prola
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France; EnvA, IMRB, 94700 Maisons-Alfort, France
| | - Peggy Lafuste
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Marianne Gervais
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Nathalie Chevallier
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France; Etablissement Français du Sang, 94017, Créteil, France
| | - Zeynab Koumaiha
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Kaouthar Kefi
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Laura Braud
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Alain Schmitt
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - Azzedine Yacia
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | | | - Barbara Hersant
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France; AP-HP, Hôpital Henri Mondor, A. Chenevier, Service de chirurgie plastique et maxillo-faciale, Créteil, France
| | - Mounia Sid-Ahmed
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France; AP-HP, Hôpital Henri Mondor, A. Chenevier, Service de chirurgie plastique et maxillo-faciale, Créteil, France
| | - Sabrina Ben Larbi
- Institut NeuroMyoGène, Université Claude Bernard - Lyon 1, University Lyon, CNRS UMR 5310, INSERM U1217, Lyon, France
| | - Katerina Komrskova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Prague, Czech Republic; Department of Zoology, Faculty of Science, Charles University, 128 44 Prague 2, Czech Republic
| | - Jakub Rohlena
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Prague, Czech Republic
| | - Frederic Relaix
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France; EnvA, IMRB, 94700 Maisons-Alfort, France; APHP, Hôpitaux Universitaires Henri Mondor & Centre de Référence des Maladies Neuromusculaires GNMH, 94000, Créteil, France
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Prague, Czech Republic; School of Medical Science, Griffith University, Southport, QLD 4222, Australia
| | | |
Collapse
|
26
|
Distinct Shades of Adipocytes Control the Metabolic Roles of Adipose Tissues: From Their Origins to Their Relevance for Medical Applications. Biomedicines 2021; 9:biomedicines9010040. [PMID: 33466493 PMCID: PMC7824911 DOI: 10.3390/biomedicines9010040] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue resides in specific depots scattered in peripheral or deeper locations all over the body and it enwraps most of the organs. This tissue is always in a dynamic evolution as it must adapt to the metabolic demand and constraints. It exhibits also endocrine functions important to regulate energy homeostasis. This complex organ is composed of depots able to produce opposite functions to monitor energy: the so called white adipose tissue acts to store energy as triglycerides preventing ectopic fat deposition while the brown adipose depots dissipate it. It is composed of many cell types. Different types of adipocytes constitute the mature cells specialized to store or burn energy. Immature adipose progenitors (AP) presenting stem cells properties contribute not only to the maintenance but also to the expansion of this tissue as observed in overweight or obese individuals. They display a high regeneration potential offering a great interest for cell therapy. In this review, we will depict the attributes of the distinct types of adipocytes and their contribution to the function and metabolic features of adipose tissue. We will examine the specific role and properties of distinct depots according to their location. We will consider their cellular heterogeneity to present an updated picture of this sophisticated tissue. We will also introduce new trends pointing out a rational targeting of adipose tissue for medical applications.
Collapse
|
27
|
Hénon P, Lahlil R. CD34+ Stem Cells and Regenerative Medicine. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
28
|
Esmaeilizadeh Z, Mohammadi B, Rajabibazl M, Ghaderian SMH, Omrani MD, Fazeli Z. Expression Analysis of GDNF/RET Signaling Pathway in Human AD-MSCs Grown in HEK 293 Conditioned Medium (HEK293-CM). Cell Biochem Biophys 2020; 78:531-539. [PMID: 32803668 DOI: 10.1007/s12013-020-00936-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: 10/01/2019] [Accepted: 08/05/2020] [Indexed: 10/23/2022]
Abstract
Mesenchymal stem cells have been considered as the suitable source for the repair of kidney lesions. The study and identification of novel approaches could improve the efficiency of these cells in the recovery of kidney. In the present study, the effect of HEK 293 conditioned medium (HEK293-CM) was evaluated on the expression of GDNF/RET signaling pathway and their downstream genes in the human adipose-derived mesenchymal stem cells (AD-MSCs). For this purpose, the human AD-MSCs were cultured in the medium containing HEK293-CM. After the RNA extraction and cDNA synthesis, the expression level of GFRA1, GDNF, SPRY1, ETV4, ETV5, and CRLF1 genes were determined by SYBR Green Real time PCR. The obtained results indicated that the GDNF and GFRA1 expression enhanced in the AD-MSCs following treatment with 10% HEK293-CM-5%FBS as compared to the untreated AD-MSCs. These results were consistent with the decreased expression of SPRY1. The significant increased expression of ETV4, ETV5, and CRLF1 genes also showed that HEK293-CM activated the GDNF/RET signaling pathway in the AD-MSCs (P < 0.05). The obtained data suggested that the treatment with HEK293-CM activated the GDNF/RET signaling pathway in the human AD-MSCs.
Collapse
Affiliation(s)
- Zahra Esmaeilizadeh
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahar Mohammadi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mir Davood Omrani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Fazeli
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Ghoneim MA, Refaie AF, Elbassiouny BL, Gabr MM, Zakaria MM. From Mesenchymal Stromal/Stem Cells to Insulin-Producing Cells: Progress and Challenges. Stem Cell Rev Rep 2020; 16:1156-1172. [PMID: 32880857 PMCID: PMC7667138 DOI: 10.1007/s12015-020-10036-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells (MSCs) are an attractive option for cell therapy for type 1 diabetes mellitus (DM). These cells can be obtained from many sources, but bone marrow and adipose tissue are the most studied. MSCs have distinct advantages since they are nonteratogenic, nonimmunogenic and have immunomodulatory functions. Insulin-producing cells (IPCs) can be generated from MSCs by gene transfection, gene editing or directed differentiation. For directed differentiation, MSCs are usually cultured in a glucose-rich medium with various growth and activation factors. The resulting IPCs can control chemically-induced diabetes in immune-deficient mice. These findings are comparable to those obtained from pluripotent cells. PD-L1 and PD-L2 expression by MSCs is upregulated under inflammatory conditions. Immunomodulation occurs due to the interaction between these ligands and PD-1 receptors on T lymphocytes. If this function is maintained after differentiation, life-long immunosuppression or encapsulation could be avoided. In the clinical setting, two sites can be used for transplantation of IPCs: the subcutaneous tissue and the omentum. A 2-stage procedure is required for the former and a laparoscopic procedure for the latter. For either site, cells should be transplanted within a scaffold, preferably one from fibrin. Several questions remain unanswered. Will the transplanted cells be affected by the antibodies involved in the pathogenesis of type 1 DM? What is the functional longevity of these cells following their transplantation? These issues have to be addressed before clinical translation is attempted. Graphical Abstract Bone marrow MSCs are isolated from the long bone of SD rats. Then they are expanded and through directed differentiation insulin-producing cells are formed. The differentiated cells are loaded onto a collagen scaffold. If one-stage transplantation is planned, a drug delivery system must be incorporated to ensure immediate oxygenation, promote vascularization and provide some growth factors. Some mechanisms involved in the immunomodulatory function of MSCs. These are implemented either by cell to cell contact or by the release of soluble factors. Collectively, these pathways results in an increase in T-regulatory cells.
Collapse
|
30
|
Akakpo W, Schirmann A, Ferretti L, Ben-Naoum K, Carnicelli D, Graziana JP, Hupertan V, Madec FX, Marcelli F, Methorst C, Morel-Journel N, Savareux L, Terrier JE, Faix A, Huyghe E, Yiou R. [Biotherapies for erectile dysfunction and Peyronie's disease: Where are we now?]. Prog Urol 2020; 30:1000-1013. [PMID: 32826194 DOI: 10.1016/j.purol.2020.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/21/2020] [Accepted: 05/05/2020] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Clinical trials of cell therapy for erectile dysfunction (ED) and Peyronie's disease (PD) were recently conducted after preclinical studies. AIMS The aims of this study are to give an update on biotherapy for ED and PD and to describe the regulatory framework for these therapies. MATERIALS AND METHODS A literature review was performed through PubMed and Clinical.trials.gov addressing cell therapy for ED and PD and using following keywords "erectile dysfunction", "Peyronie's disease", "stem cell", and "platelet-rich plasma". RESULTS Preclinical studies in rodent models have shown the potential benefit of cell therapy for ED after radical prostatectomy or caused by metabolic diseases, and PD. The tissues used to obtain the therapeutic product were bone marrow, adipose tissue and blood (PRP, platelet-rich plasma). Mechanism of action was shown to be temporary and mainly paracrine. Four clinical trials were published concerning ED after radical prostatectomy and in diabetic patients and one for PD. Eleven clinical trials including three randomized trials are currently going on. Preclinical and preliminary clinical results suggested the possibility to improve spontaneous erectile function and response to pharmaceutical treatment in initially non-responder patients. This effect is mediated by an improvement of penile vascularization. A reduction of penile curvature without side effect was noted after injections into the plaque of PD patients. Most of these therapeutic strategies using autologous cells were considered as "Advanced Therapy Medicinal Products" with strict regulatory frameworks imposing heavy constraints, in particular in case of "substantial" modification of the cells. The regulatory framework remains unclear and more permissive for PRP and cell therapy processes with extemporaneous preparation/injection and no "substantial" modifications. CONCLUSIONS First results on cell therapy for ED and PD are promising. The regulatory framework can significantly change according to cell preparations and origins leading to various constraints. This regulatory framework is crucial to consider for the choice of the procedure.
Collapse
Affiliation(s)
- W Akakpo
- Service d'urologie, université Pierre et Marie Curie, hôpital universitaire de la Pitié-Salpêtrière, 75013 Paris, France
| | - A Schirmann
- Service d'urologie, hôpitaux universitaires Henri-Mondor, CHU Henri-Mondor, AP-HP, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - L Ferretti
- Service d'urologie, hôpital d'instruction des armées Robert-Picqué, Villenave-d'Ornon, France
| | - K Ben-Naoum
- Service d'urologie, CHU de Nîmes, Nîmes, France
| | - D Carnicelli
- Service d'urologie, CH de Chambery, Chambery, France
| | - J-P Graziana
- Clinique mutualiste de la porte de l'Orient, Lorient, France
| | - V Hupertan
- Cabinet médical Paris Batignolles, Paris, France
| | - F X Madec
- Service d'urologie, CHU de Nantes, Nantes, France
| | - F Marcelli
- Service d'urologie, CHRU de Lille, Lille, France
| | - C Methorst
- Service d'urologie, CH des Quatre Villes, Saint-Cloud, France
| | | | - L Savareux
- Hôpital privé de La Châtaignerie, Beaumont, France
| | | | - A Faix
- Clinique mutualiste Beau-Soleil, Montpellier, France
| | - E Huyghe
- Service d'urologie, CHU de Toulouse, Toulouse, France
| | - R Yiou
- Service d'urologie, hôpitaux universitaires Henri-Mondor, CHU Henri-Mondor, AP-HP, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France.
| |
Collapse
|
31
|
Cismaru CA, Soritau O, Jurj AM, Lajos R, Pop B, Bocean C, Albzour B, Baldasici O, Moldovan C, Neagoe IB. Isolation and Characterization of a Fetal-Maternal Microchimeric Stem Cell Population in Maternal Hair Follicles Long after Parturition. Stem Cell Rev Rep 2020; 15:519-529. [PMID: 31123983 DOI: 10.1007/s12015-019-09885-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fetal-maternal microchimerism describes the acquisition of fetal stem cells (FSC) by the mother during pregnancy and their long-term persistence after parturition. FSC may engraft in a variety of maternal tissues especially if there is organ/tissue injury, but their role and mechanism of persistence still remains elusive. Clinical applications due to their pluripotency, immunomodulatory effects and accessibility make them good candidates for ex-vivo manipulation and autologous therapies. The hair follicles contain a distinctive niche for pluripotent stem cells (PSC). To date, there is no published evidence of fetal microchimerism in the hair follicle. In our study, follicular unit extraction (FUE) technique allowed easy stem cell cultures to be obtained while simple hair follicle removal by pull-out technique failed to generate stem cells in culture. We identified microchimeric fetal stem cells within the primitive population of maternal stem cells isolated from the hair follicles with typical mesenchymal phenotype, expression of PSC genes and differentiation potential towards osteocytes, adypocites and chondrocytes. This is the first study to isolate fetal microchimeric stem cells in adult human hair long after parturition. We presume a sanctuary partition mechanism with PSC of the mother deposited during early embryogenesis could explain their long-term persistence.
Collapse
Affiliation(s)
- Cosmin Andrei Cismaru
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania. .,The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania.
| | - Olga Soritau
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania
| | - Ancuta-Maria Jurj
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Raduly Lajos
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bogdan Pop
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania
| | - Cosmina Bocean
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania
| | - Bogdan Albzour
- Department of Dermatology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Oana Baldasici
- The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania
| | - Cristian Moldovan
- Research Center for Advanced Medicine - Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan Neagoe
- Research Center for Functional Genomics Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania.,Research Center for Advanced Medicine - Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
32
|
Wartalski K, Gorczyca G, Wiater J, Tabarowski Z, Palus-Chramiec K, Setkowicz Z, Duda M. Efficient generation of neural-like cells from porcine ovarian putative stem cells - morphological characterization and evaluation of their electrophysiological properties. Theriogenology 2020; 155:256-268. [PMID: 32810809 DOI: 10.1016/j.theriogenology.2020.05.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/20/2020] [Accepted: 05/23/2020] [Indexed: 12/20/2022]
Abstract
Until recently, the mammalian ovary was considered to consist of fully differentiated tissues, but evidence for the presence of adult stem cells in this organ appeared. The differentiation potential of these cells, referred to as putative stem cells, is not well defined. Porcine ovarian putative stem cells (poPSCs) were immunomagnetically isolated from postnatal pig ovaries based on the presence of the SSEA-4 surface marker protein. First, they were cultured in the undifferentiated state. After the third passage, a novel 7-day culture method inducing their differentiation into neural-like cells by the addition of forskolin (FSK), retinoic acid (RA) and basic fibroblast growth factor (bFGF) to the culture medium was applied. After 7 days, poPSCs successfully differentiated into neural-like cells, as evidenced by neural morphology and the presence of the neuronal markers nestin, NeuN, and GFAP, as confirmed by immunofluorescence, western blot, and real-time PCR. Electrophysiological analysis of potassium and sodium channel activity (patch clamp) confirmed that they indeed differentiated into neurons. The plasticity of poPSCs offers an excellent opportunity, especially in the field of neuroscience, since they can differentiate into neurons or glial cells. Although poPSCs might not be pluripotent cells, they also escape the rigid classification framework of adult stem cells.
Collapse
Affiliation(s)
- Kamil Wartalski
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland; Department of Histology, Jagiellonian University Medical College, Kopernika 7, 31-034, Krakow, Poland
| | - Gabriela Gorczyca
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Jerzy Wiater
- Department of Histology, Jagiellonian University Medical College, Kopernika 7, 31-034, Krakow, Poland; Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Zbigniew Tabarowski
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Zuzanna Setkowicz
- Department of Neuroanatomy, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Małgorzata Duda
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland.
| |
Collapse
|
33
|
Ghamari SH, Abbasi-Kangevari M, Tayebi T, Bahrami S, Niknejad H. The Bottlenecks in Translating Placenta-Derived Amniotic Epithelial and Mesenchymal Stromal Cells Into the Clinic: Current Discrepancies in Marker Reports. Front Bioeng Biotechnol 2020; 8:180. [PMID: 32232037 PMCID: PMC7083014 DOI: 10.3389/fbioe.2020.00180] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/24/2020] [Indexed: 12/23/2022] Open
Abstract
Placenta-derived amniotic cells have prominent features for application in regenerative medicine. However, there are still discrepancies in the characterization of human amniotic epithelial and mesenchymal stromal cells. It seems crucial that the characterization of human amniotic membrane cells be investigated to determine whether there are currently discrepancies in their characterization reports. In addition, possible causes for the witnessed discrepancies need to be addressed toward paving the way for further clinical application and safer practices. The objective of this review is to investigate the marker characterization as well as the potential causes of the discrepancies in the previous reports on placenta-derived amniotic epithelial and mesenchymal stromal cells. The current discrepancies could be potentially due to reasons including passage number and epithelial to mesenchymal transition (EMT), cell heterogeneity, isolation protocols and cross-contamination, the region of cell isolation on placental disk, measuring methods, and gestational age.
Collapse
Affiliation(s)
- Seyyed-Hadi Ghamari
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Abbasi-Kangevari
- Student Research Committee, Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tahereh Tayebi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Vienna, Austria
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Chen K, Huang Y, Singh R, Wang ZZ. Arrhythmogenic risks of stem cell replacement therapy for cardiovascular diseases. J Cell Physiol 2020; 235:6257-6267. [PMID: 31994198 DOI: 10.1002/jcp.29554] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/22/2022]
Abstract
Ischemic heart disease and congestive heart failure are major contributors to high morbidity and mortality. Approximately 1.5 million cases of myocardial infarction occur annually in the United States; the yearly incidence rate is approximately 600 cases per 100,000 people. Although significant progress to improve the survival rate has been made by medications and implantable medical devices, damaged cardiomyocytes are unable to be recovered by current treatment strategies. After almost two decades of research, stem cell therapy has become a very promising approach to generate new cardiomyocytes and enhance the function of the heart. Along with clinical trials with stem cells conducted in cardiac regeneration, concerns regarding safety and potential risks have emerged. One of the contentious issues is the electrical dysfunctions of cardiomyocytes and cardiac arrhythmia after stem cell therapy. In this review, we focus on the cell sources currently used for stem cell therapy and discuss related arrhythmogenic risk.
Collapse
Affiliation(s)
- Kang Chen
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuting Huang
- Department of Medicine, University of Maryland Medical Center Midtown Campus, Baltimore, Maryland
| | - Radhika Singh
- Center for Biotechnology Education, Johns Hopkins University, Baltimore, Maryland
| | - Zack Z Wang
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
35
|
Rossi F, Noren H, Sarria L, Schiller PC, Nathanson L, Beljanski V. Combination therapies enhance immunoregulatory properties of MIAMI cells. Stem Cell Res Ther 2019; 10:395. [PMID: 31852519 PMCID: PMC6921447 DOI: 10.1186/s13287-019-1515-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 11/14/2019] [Accepted: 11/28/2019] [Indexed: 01/08/2023] Open
Abstract
Background Mesenchymal stromal cells (MSCs), adult stromal cells most commonly isolated from bone marrow (BM), are being increasingly utilized in various therapeutic applications including tissue repair via immunomodulation, which is recognized as one of their most relevant mechanism of action. The promise of MSC-based therapies is somewhat hindered by their apparent modest clinical benefits, highlighting the need for approaches that would increase the efficacy of such therapies. Manipulation of cellular stress-response mechanism(s) such as autophagy, a catabolic stress-response mechanism, with small molecules prior to or during MSC injection could improve MSCs’ therapeutic efficacy. Unfortunately, limited information exists on how manipulation of autophagy affects MSCs’ response to inflammation and subsequent immunoregulatory properties. Methods In this study, we exposed BM-MSC precursor cells, “marrow-isolated adult multilineage inducible” (MIAMI) cells, to autophagy modulators tamoxifen (TX) or chloroquine (CQ), together with IFN-γ. Exposed cells then underwent RNA sequencing (RNAseq) to determine the effects of TX or CQ co-treatments on cellular response to IFN-γ at a molecular level. Furthermore, we evaluated their immunoregulatory capacity using activated CD4+ T cells by analyzing T cell activation marker CD25 and the percentage of proliferating T cells after co-culturing the cells with MIAMI cells treated or not with TX or CQ. Results RNAseq data indicate that the co-treatments alter both mRNA and protein levels of key genes responsible for MSCs’ immune-regulatory properties. Interestingly, TX and CQ also altered some of the microRNAs targeting such key genes. In addition, while IFN-γ treatment alone increased the surface expression of PD-L1 and secretion of IDO, this increase was further enhanced with TX. An improvement in MIAMI cells’ ability to decrease the activation and proliferation of T cells was also observed with TX, and to a lesser extent, CQ co-treatments. Conclusion Altogether, this work suggests that both TX and CQ have a potential to enhance MIAMI cells’ immunoregulatory properties. However, this enhancement is more pronounced with TX co-treatment.
Collapse
Affiliation(s)
- Fiorella Rossi
- Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, Davie, FL, 33328, USA
| | - Hunter Noren
- Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, Davie, FL, 33328, USA
| | - Leonor Sarria
- Institute for Neuroimmune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Paul C Schiller
- Department of Orthopaedics, University of Miami Miller School of Medicine, Miami, FL, USA.,Prime Cell Biomedical Inc., Miami, FL, USA
| | - Lubov Nathanson
- Institute for Neuroimmune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Vladimir Beljanski
- Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, 3200 South University Drive, Fort Lauderdale, Davie, FL, 33328, USA.
| |
Collapse
|
36
|
Isolation and Culture of Human Stem Cells from Apical Papilla under Low Oxygen Concentration Highlight Original Properties. Cells 2019; 8:cells8121485. [PMID: 31766521 PMCID: PMC6952825 DOI: 10.3390/cells8121485] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/07/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
Stem cells isolated from the apical papilla of wisdom teeth (SCAPs) are an attractive model for tissue repair due to their availability, high proliferation rate and potential to differentiate in vitro towards mesodermal and neurogenic lineages. Adult stem cells, such as SCAPs, develop in stem cell niches in which the oxygen concentration [O2] is low (3–8% compared with 21% of ambient air). In this work, we evaluate the impact of low [O2] on the physiology of SCAPs isolated and processed in parallel at 21% or 3% O2 without any hyperoxic shock in ambient air during the experiment performed at 3% O2. We demonstrate that SCAPs display a higher proliferation capacity at 3% O2 than in ambient air with elevated expression levels of two cell surface antigens: the alpha-6 integrin subunit (CD49f) and the embryonic stem cell marker (SSEA4). We show that the mesodermal differentiation potential of SCAPs is conserved at early passage in both [O2], but is partly lost at late passage and low [O2], conditions in which SCAPs proliferate efficiently without any sign of apoptosis. Unexpectedly, we show that autophagic flux is active in SCAPs irrespective of [O2] and that this process remains high in cells even after prolonged exposure to 3% O2.
Collapse
|
37
|
Oshchepkova A, Neumestova A, Matveeva V, Artemyeva L, Morozova K, Kiseleva E, Zenkova M, Vlassov V. Cytochalasin-B-Inducible Nanovesicle Mimics of Natural Extracellular Vesicles That Are Capable of Nucleic Acid Transfer. MICROMACHINES 2019; 10:E750. [PMID: 31683842 PMCID: PMC6915531 DOI: 10.3390/mi10110750] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/27/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles provide cell-to-cell communication and have great potential for use as therapeutic carriers. This study was aimed at the development of an extracellular vesicle-based system for nucleic acid delivery. Three types of nanovesicles were assayed as oligonucleotide carriers: mesenchymal stem cell-derived extracellular vesicles and mimics prepared either by cell treatment with cytochalasin B or by vesicle generation from plasma membrane. Nanovesicles were loaded with a DNA oligonucleotide by freezing/thawing, sonication, or permeabilization with saponin. Oligonucleotide delivery was assayed using HEK293 cells. Extracellular vesicles and mimics were characterized by a similar oligonucleotide loading level but different efficiency of oligonucleotide delivery. Cytochalasin-B-inducible nanovesicles exhibited the highest level of oligonucleotide accumulation in HEK293 cells and a loading capacity of 0.44 ± 0.05 pmol/µg. The loaded oligonucleotide was mostly protected from nuclease action.
Collapse
Affiliation(s)
- Anastasiya Oshchepkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Alexandra Neumestova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Vera Matveeva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Lyudmila Artemyeva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Ksenia Morozova
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia.
| | - Elena Kiseleva
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia.
| | - Marina Zenkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Valentin Vlassov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| |
Collapse
|
38
|
A Combinatorial Cell and Drug Delivery Strategy for Huntington's Disease Using Pharmacologically Active Microcarriers and RNAi Neuronally-Committed Mesenchymal Stromal Cells. Pharmaceutics 2019; 11:pharmaceutics11100526. [PMID: 31614758 PMCID: PMC6835496 DOI: 10.3390/pharmaceutics11100526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/15/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023] Open
Abstract
For Huntington's disease (HD) cell-based therapy, the transplanted cells are required to be committed to a neuronal cell lineage, survive and maintain this phenotype to ensure their safe transplantation in the brain. We first investigated the role of RE-1 silencing transcription factor (REST) inhibition using siRNA in the GABAergic differentiation of marrow-isolated adult multilineage inducible (MIAMI) cells, a subpopulation of MSCs. We further combined these cells to laminin-coated poly(lactic-co-glycolic acid) PLGA pharmacologically active microcarriers (PAMs) delivering BDNF in a controlled fashion to stimulate the survival and maintain the differentiation of the cells. The PAMs/cells complexes were then transplanted in an ex vivo model of HD. Using Sonic Hedgehog (SHH) and siREST, we obtained GABAergic progenitors/neuronal-like cells, which were able to secrete HGF, SDF1 VEGFa and BDNF, of importance for HD. GABA-like progenitors adhered to PAMs increased their mRNA expression of NGF/VEGFa as well as their secretion of PIGF-1, which can enhance reparative angiogenesis. In our ex vivo model of HD, they were successfully transplanted while attached to PAMs and were able to survive and maintain this GABAergic neuronal phenotype. Together, our results may pave the way for future research that could improve the success of cell-based therapy for HDs.
Collapse
|
39
|
Madanagopal TT, Franco-Obregón A, Rosa V. Comparative study of xeno-free induction protocols for neural differentiation of human dental pulp stem cells in vitro. Arch Oral Biol 2019; 109:104572. [PMID: 31600663 DOI: 10.1016/j.archoralbio.2019.104572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 09/18/2019] [Accepted: 09/22/2019] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To compare three different xeno-free protocols for neural differentiation of human dental pulp stem cells (DPSC). METHODS DPSC were treated with three different media to induce neural differentiation namely N1 (DMEM for 5 days), N2 (PSC neural induction media for 7 days) and N3 (neural media with B27 supplement, 40 ng/ml bFGF and 20 ng/ml EGF for 21 days). Cell proliferation (MTS assay), morphology, gene (qPCR for NESTIN, VIMENTIN, TUB-3, ENO2, NF-M and NF-H) and protein expression (flow cytometry) of neurogenic markers were assessed at different time points and compared to untreated cells (DMEM supplemented with 10% FBS). Statistical analysis was performed with global significance level of 5%. RESULTS N1 and N2 formulations increased the genetic expression of two out of six genes TUB-3, NF-M and TUB-3, NF-H, respectively, whereas N3 elevated the expression of all genes by the late stage. N3 also stimulated protein expression for NESTIN, TUB-3 and NF-H. Cells treated with both N2 and N3 presented neuron-like morphology, decreased proliferation and expression of stemness genes at protocol end point. CONCLUSION N3 was the most effective formulation in promoting a neurogenic shift in gene and protein expression. Cells provided with the N3 formulation exhibited neuron-like morphology, elaborating axonal-like projections concomitant with cell cycle withdrawal and reduced expression of stemness genes indicating greater commitment to a neurogenic lineage.
Collapse
Affiliation(s)
- Thulasi Thiruvallur Madanagopal
- Faculty of Dentistry, National University of Singapore, Singapore; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Health Innovation & Technology, iHealthtech, National University of Singapore, Singapore
| | - Vinicius Rosa
- Faculty of Dentistry, National University of Singapore, Singapore; National University Centre For Oral Health Singapore, National University Hospital System, Singapore.
| |
Collapse
|
40
|
Lee YH, Lee HT, Chen CL, Chang CH, Hsu CY, Shyu WC. Role of FOXC1 in regulating APSCs self-renewal via STI-1/PrP C signaling. Am J Cancer Res 2019; 9:6443-6465. [PMID: 31588228 PMCID: PMC6771253 DOI: 10.7150/thno.35619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/20/2019] [Indexed: 01/03/2023] Open
Abstract
Forkhead box protein C1 (FOXC1) is known to regulate developmental processes in the skull and brain. Methods: The unique multipotent arachnoid-pia stem cells (APSCs) isolated from human and mouse arachnoid-pia membranes of meninges were grown as 3D spheres and displayed a capacity for self-renewal. Additionally, APSCs also expressed the surface antigens as mesenchymal stem cells. By applying the FOXC1 knockout mice and mouse brain explants, signaling cascade of FOXC1-STI-1-PrPC was investigated to demonstrate the molecular regulatory pathway for APSCs self-renewal. Moreover, APSCs implantation in stroke model was also verified whether neurogenic property of APSCs could repair the ischemic insult of the stroke brain. Results: Activated FOXC1 regulated the proliferation of APSCs in a cell cycle-dependent manner, whereas FOXC1-mediated APSCs self-renewal was abolished in FOXC1 knockout mice (FOXC1-/- mice). Moreover, upregulation of STI-1 regulated by FOXC1 enhanced cell survival and self-renewal of APSCs through autocrine signaling of cellular prion protein (PrPC). Mouse brain explants STI-1 rescues the cortical phenotype in vitro and induces neurogenesis in the FOXC1 -/- mouse brain. Furthermore, administration of APSCs in ischemic brain restored the neuroglial microenvironment and improved neurological dysfunction. Conclusion: We identified a novel role for FOXC1 in the direct regulation of the STI-1-PrPC signaling pathway to promote cell proliferation and self-renewal of APSCs.
Collapse
|
41
|
Deegan AJ, Hendrikson WJ, El Haj AJ, Rouwkema J, Yang Y. Regulation of endothelial cell arrangements within hMSC - HUVEC co-cultured aggregates. Biomed J 2019; 42:166-177. [PMID: 31466710 PMCID: PMC6717755 DOI: 10.1016/j.bj.2019.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 11/25/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background Micro-mass culturing or cellular aggregation is an effective method used to form mineralised bone tissue. Poor core cell viability, however, is often an impeding characteristic of large micro-mass cultures, and equally for large tissue-engineered bone grafts. Because of this, efforts are being made to enhance large graft perfusion, often through pre-vascularisation, which involves the co-culture of endothelial cells and bone cells or stem cells. Methods This study investigated the effects of different aggregation techniques and culture conditions on endothelial cell arrangements in mesenchymal stem cell and human umbilical vein endothelial cell co-cultured aggregates when endothelial cells constituted just 5%. Two different cellular aggregation techniques, i.e. suspension culture aggregation and pellet culture aggregation, were applied alongside two subsequent culturing techniques, i.e. hydrostatic loading and static culturing. Endothelial cell arrangements were assessed under such conditions to indicate potential pre-vascularisation. Results Our study found that the suspension culture aggregates cultured under hydrostatic loading offered the best environment for enhanced endothelial cell regional arrangements, closely followed by the pellet culture aggregates cultured under hydrostatic loading, the suspension culture aggregates cultured under static conditions, and the pellet culture aggregates cultured under static conditions. Conclusions The combination of particular aggregation techniques with dynamic culturing conditions appeared to have a synergistic effect on the cellular arrangements within the co-cultured aggregates.
Collapse
Affiliation(s)
- Anthony J Deegan
- Institute for Science and Technology in Medicine, School of Medicine, Keele University, Stoke-on-Trent, United Kingdom
| | - Wim J Hendrikson
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, AE, the Netherlands
| | - Alicia J El Haj
- Institute for Science and Technology in Medicine, School of Medicine, Keele University, Stoke-on-Trent, United Kingdom
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Ying Yang
- Institute for Science and Technology in Medicine, School of Medicine, Keele University, Stoke-on-Trent, United Kingdom.
| |
Collapse
|
42
|
Otali D, Al Diffalha S, Grizzle WE. Biological, Medical, and Other Tissue Variables Affecting Biospecimen Utilization. Biopreserv Biobank 2019; 17:258-263. [PMID: 31188629 PMCID: PMC6588123 DOI: 10.1089/bio.2018.0094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Bioresources are critical resources that support biomedical research because of their ability to appropriately collect, process, store, and distribute a wide range of high-quality biospecimens that meet the needs of specific investigators. Of note, some biorepositories are concerned by their growing inventories and their low rates of tissue utilization. This review discusses the technical characteristics of biospecimens that can cause morphological and molecular variability and/or limit the usefulness of biospecimens in research. This article also describes current challenges related to biospecimen characteristics that may affect biospecimen utilization. These include inadequate awareness of investigators about the availability of biospecimens with specific morphologic and molecular features, donor variability, preanalytical variables, technical problems inherent with an investigator's request for biospecimens, limited tissue availability from a biorepository based on requested sizes and/or numbers of available biospecimens, effects of times of warm and cold ischemia, damage of tissues during surgery, and molecular changes during storage. To ensure maximal biospecimen utilization of all types of biospecimens requires continual education of investigators from diverse fields, particularly on factors that cause variability in the morphological and molecular characteristics of tissues. The investigators' requests for biospecimens and associated data should be reviewed carefully, including by a bioresource-associated pathologist. Queries arising from the request/application form should be resolved by bioresource personnel directly with the investigator.
Collapse
Affiliation(s)
- Dennis Otali
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Sameer Al Diffalha
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - William E. Grizzle
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama
- The Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
43
|
Pan S, Chen YC, Zhao N, Feng X, Yang DD, Wang Y, Jin ZB. A new subset of small stem cells in bovine bone marrow stromal cell populations. J Cell Biochem 2019; 120:13881-13892. [PMID: 30983000 DOI: 10.1002/jcb.28661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/21/2018] [Accepted: 01/07/2019] [Indexed: 01/23/2023]
Abstract
Bone marrow stromal cells (BMSCs) are a unique population of multipotent cells that exhibit pluripotent properties to a certain extent and are significantly heterogeneous in terms of the cell population. We isolate a small cell subpopulation from bovine BMSCs, bovine small stem cells (bSSCs), and herein characterize their properties. The bSSCs are smaller in size and express nuclear Oct-4 and other pluripotency markers. In addition, when cultured in suspension conditions, bSSCs form three-dimensional spheres and display a strong capability for self-renewal and differentiation into cells from three germ layers. Notably, bSSCs display neural features with Sox1 and Pax6 expression. Using bSSCs as donor nuclear cells for somatic cell nuclear transfer, we further demonstrate that the developmental potential of cloned embryos in vitro is significantly increased. Our study identifies a new bovine bone marrow stromal cell-derived stem cell subtype that could have broad importance for developmental biology as well as great potential for regenerative medicine.
Collapse
Affiliation(s)
- Shaohui Pan
- Laboratory for Stem Cell and Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Sciences, Wenzhou, China
| | - Yu-Chen Chen
- Laboratory for Stem Cell and Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Sciences, Wenzhou, China
| | - Ning Zhao
- Laboratory for Stem Cell and Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Sciences, Wenzhou, China
| | - Xiang Feng
- Laboratory for Stem Cell and Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Sciences, Wenzhou, China
| | - Dan-Dan Yang
- Laboratory for Stem Cell and Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Sciences, Wenzhou, China
| | - Yongshen Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Zi-Bing Jin
- Laboratory for Stem Cell and Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Visual Sciences, Wenzhou, China
| |
Collapse
|
44
|
Robinson PG, Murray IR, West CC, Goudie EB, Yong LY, White TO, LaPrade RF. Reporting of Mesenchymal Stem Cell Preparation Protocols and Composition: A Systematic Review of the Clinical Orthopaedic Literature. Am J Sports Med 2019; 47:991-1000. [PMID: 29554460 DOI: 10.1177/0363546518758667] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are increasingly being used in the treatment of a wide variety of sports-related conditions. Despite this enthusiasm, the biological properties of MSCs and their effects on musculoskeletal tissue healing remain poorly understood. MSC-based strategies encompass cell populations with heterogeneous phenotypes isolated from multiple tissues and using different methods. Therefore, comprehensive reporting of the source, preparation methods, and characteristics of MSC strategies is essential to enable interpretation of results. PURPOSE To perform a systematic review of levels of reporting of key variables in MSC preparation and composition for clinical studies evaluating MSC-based therapies in the treatment of musculoskeletal conditions. STUDY DESIGN Systematic review. METHODS A systematic review of the clinical orthopaedic and sports medicine literature from 2002 to 2017 was performed. The following inclusion criteria were used: human clinical trials, published in the English language, involving the administration of MSC-based therapies for orthopaedic or sports medicine applications. In vitro or ex vivo studies, editorials, letters to the editor, and studies relating to cosmetic, neurological, or dental applications were excluded. RESULTS Of the 1259 studies identified on the initial search, 36 studies were found to satisfy the inclusion criteria for analysis on comprehensive review. Fifty-seven percent of studies evaluated bone marrow-derived MSCs, 41% evaluated adipose-derived MSCs, and 2% evaluated synovium-derived MSCs. Considerable deficiencies in the reporting of key variables, including the details of stem cell processing, culture conditions, and the characteristics of cell populations delivered, were noted. Overall, studies reported only 52% (range, 30%-80%) of variables that may critically influence outcome. No study provided adequate information relating to all of these variables. CONCLUSION All existing clinical studies evaluating MSCs for orthopaedic or sports medicine applications are limited by inadequate reporting of both preparation protocols and composition. Deficient reporting of the variables that may critically influence outcome precludes interpretation, prevents others from reproducing experimental conditions, and makes comparisons across studies difficult. We encourage the adoption of emerging minimum reporting standards for clinical studies evaluating the use of MSCs in orthopaedics.
Collapse
Affiliation(s)
| | - Iain R Murray
- Department of Trauma and Orthopaedics, University of Edinburgh, UK.,Scottish Centre for Regenerative Medicine, University of Edinburgh, UK
| | | | - Ewan B Goudie
- Department of Trauma and Orthopaedics, University of Edinburgh, UK
| | - Li Y Yong
- Scottish Centre for Regenerative Medicine, University of Edinburgh, UK
| | - Timothy O White
- Department of Trauma and Orthopaedics, University of Edinburgh, UK
| | | |
Collapse
|
45
|
Xu QQ, Guo WW, Wang X, Zhai S, Yang S. Acid stimulation-induced semi-pluripotent characteristics in human somatic cells. Acta Otolaryngol 2019; 139:146-152. [PMID: 30734622 DOI: 10.1080/00016489.2018.1484565] [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: 10/27/2022]
Abstract
BACKGROUND Clinical trials of cell-based therapies using induced pluripotent stem (iPS) cells have already been started for several neurological diseases. OBJECTIVE The purpose of the present study was to explore the characteristics and differentiation of somatic cells in vitro undergoing a low pH treatment, so as to provide new therapeutic strategies for treating sensorineural hearing loss. METHODS Somatic cells were treated with low pH solution to alter their characteristics. In addition, a mouse model of the cochlear lesion was constructed using bilirubin. Subsequently, the characteristics and therapeutic effect of somatic cells undergoing low pH treatment were examined by morphology, alkaline phosphatase (AKP) activity, immunofluorescence assay and q-PCR. RESULTS The cells in the experimental group grew better than those in the control group. The AKP activity in the experimental group was higher than that in the control group. The expression of Nanog and Oct4 was both positive in the two groups. When the cells were changed to neurobasal medium, the marker of nestin was positive. CONCLUSION The human somatic cells undergoing a low pH treatment showed the similar characteristics as those of iPS cells, although the functions and therapeutic effect of these altered human somatic cells need to be further studied.
Collapse
Affiliation(s)
- Qing-Qing Xu
- Department of Allergy, Beijing Tongren Hospital, Beijing, China
| | - Wei-Wei Guo
- Department of Otolaryngology, Head & Neck Surgery, Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, China
| | - Xiangdong Wang
- Department of ENT, Beijing Tongren Hospital, Beijing, China
| | - Suoqiang Zhai
- PLA General Hospital, Institute of Otorhinolaryngology, Beijing, China
| | - Shiming Yang
- Department of Otolaryngology, Head and Neck Surgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
46
|
Suman S, Domingues A, Ratajczak J, Ratajczak MZ. Potential Clinical Applications of Stem Cells in Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1201:1-22. [PMID: 31898779 DOI: 10.1007/978-3-030-31206-0_1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The field of regenerative medicine is looking for a pluripotent/multipotent stem cell able to differentiate across germ layers and be safely employed in therapy. Unfortunately, with the exception of hematopoietic stem/progenitor cells (HSPCs) for hematological applications, the current clinical results with stem cells are somewhat disappointing. The potential clinical applications of the more primitive embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have so far been discouraging, as both have exhibited several problems, including genomic instability, a risk of teratoma formation, and the possibility of rejection. Therefore, the only safe stem cells that have so far been employed in regenerative medicine are monopotent stem cells, such as the abovementioned HSPCs or mesenchymal stem cells (MSCs) isolated from postnatal tissues. However, their monopotency, and therefore limited differentiation potential, is a barrier to their broader application in the clinic. Interestingly, results have accumulated indicating that adult tissues contain rare, early-development stem cells known as very small embryonic-like stem cells (VSELs), which can differentiate into cells from more than one germ layer. This chapter addresses different sources of stem cells for potential clinical application and their advantages and problems to be solved.
Collapse
Affiliation(s)
- Suman Suman
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Alison Domingues
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland.
| |
Collapse
|
47
|
Fazeli Z, Faramarzi S, Ahadi A, Omrani MD, Ghaderian SM. Efficiency of mesenchymal stem cells in treatment of urinary incontinence: a systematic review on animal models. Regen Med 2018; 14:69-76. [PMID: 30560712 DOI: 10.2217/rme-2018-0040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM In recent years, the administration of stem cells has been considered a new option for treatment of urinary incontinence (UI). In the present study, the efficiency of mesenchymal stem cell (MSC) transplantation in the treatment of UI was evaluated. METHODS Combinations of the key words 'mesenchymal stem cells', 'MSCs', 'urinary incontinence', 'urethral sphincter' and 'involuntary urination' were searched in PubMed and Science Direct databases. Following application of exclusion criteria to the 1946 papers obtained and review and duplicate articles were removed, 23 articles were considered further. The search was limited to the animal model studies. RESULTS The data obtained from the evaluation of different studies indicated that the injected MSCs play an important role in the neovascularization and the recovery of muscle cells in UI models through the paracrine process. CONCLUSION The obtained data suggested that further trials are needed to be focused on clinical phase of MSC therapy on the patients affected by UI.
Collapse
Affiliation(s)
- Zahra Fazeli
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran
| | - Sepideh Faramarzi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran
| | - Alireza Ahadi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran
| | - Mir D Omrani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran
| | - Sayyed Mh Ghaderian
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran
| |
Collapse
|
48
|
Gounari E, Daniilidis A, Tsagias N, Michopoulou A, Kouzi K, Koliakos G. Isolation of a novel embryonic stem cell cord blood-derived population with in vitro hematopoietic capacity in the presence of Wharton's jelly-derived mesenchymal stromal cells. Cytotherapy 2018; 21:246-259. [PMID: 30522805 DOI: 10.1016/j.jcyt.2018.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/06/2018] [Accepted: 11/13/2018] [Indexed: 01/16/2023]
Abstract
BACKGROUND Recent studies highlight the existence of a population of cord blood (CB)-derived stem cells that bare embryonic features (very small embryonic-like stem cells [VSELs]) as the most primitive CB-stem cell population. In the present study, we present for the first time a novel and high purity isolation method of VSELs with in vitro hematopoietic capacity in the presence of Wharton's jelly-derived mesenchymal stromal cells (WJ-MSCs). METHODS The experimental procedure includes isolation upon gradually increased centrifugation spins and chemotaxis to Stromal cell-derived factor 1a (SDF-1a). Τhis cell population is characterized with flow cytometry, alkaline phosphatase (ALP) staining and qRT-PCR. The functional role of the isolated VSELs is assayed following co-culture with WJ-MSCs or bone marrow-derived mesenchymal stromal cells (BM-MSCs), whereas the stimulation of the quiescent VSEL population is verified via cell cycle analysis. The in vitro hematopoietic capacity is evaluated in methylcellulose cultures and also through induction of erythroid differentiation. RESULTS The final isolated subpopulation is characterized as a small-sized CD45/Lineage-/CXCR4+/CD133+/SSEA-4+cell population, positive in ALP staining and overexpressing the Oct3/4, Nanog and Sox-2 transcription factors. Upon the co-culture with MSCs, a stimulation of the quiescent VSEL population is observed. An impressive increase in the co-expression of the CD34+/CD45+ markers is observed following the co-culture with the WJ-MSCs, which is confirmed by the intense clonogenic ability suggesting in vitro differentiation toward all of the hematopoietic cell lineages and successful differentiation toward erythrocytes. DISCUSSION Conclusively, we propose a novel, rapid and rather simplified isolation method of CB-VSELs, capable of in vitro hematopoiesis.
Collapse
Affiliation(s)
- Eleni Gounari
- Biohellenika Biotechnology Company, Thessaloniki, Greece; Department of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Angelos Daniilidis
- 2nd Department of Obstetrics and Gynecology, Hippokratio General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Anna Michopoulou
- Biohellenika Biotechnology Company, Thessaloniki, Greece; Department of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kokkona Kouzi
- Biohellenika Biotechnology Company, Thessaloniki, Greece; Department of Histology Embryology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George Koliakos
- Biohellenika Biotechnology Company, Thessaloniki, Greece; Department of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
49
|
Magnus MK, Iceman KL, Roukis TS. Living Cryopreserved Bone Allograft as an Adjunct for Hindfoot Arthrodesis. Clin Podiatr Med Surg 2018; 35:295-310. [PMID: 29861013 DOI: 10.1016/j.cpm.2018.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Hindfoot arthrodesis is a frequently performed procedure by foot and ankle surgeons. The relatively high nonunion rate associated with these procedures has led surgeons to use adjunctive bone graft to help augment osseous union. Cellular bone allografts are a specific type of graft that incorporates osteoconductive, osteoinductive, and osteogenic properties while also eliminating the common disadvantages of autografts and traditional allografts. This article discusses the role of cellular bone allografts in hindfoot arthrodesis procedures, a review of current literature, and a comparison of available products.
Collapse
Affiliation(s)
- Mark K Magnus
- Gundersen Medical Foundation, Mail Stop: CO3-006A, 1900 South Avenue, La Crosse, WI 54601, USA
| | - Kelli L Iceman
- Gundersen Medical Foundation, Mail Stop: CO3-006A, 1900 South Avenue, La Crosse, WI 54601, USA
| | - Thomas S Roukis
- Orthopaedic Center, Gundersen Healthcare System, Mail Stop: CO2-006, 1900 South Avenue, La Crosse, WI 54601, USA.
| |
Collapse
|
50
|
Kim Y, Jin HJ, Heo J, Ju H, Lee HY, Kim S, Lee S, Lim J, Jeong SY, Kwon J, Kim M, Choi SJ, Oh W, Yang YS, Hwang HH, Yu HY, Ryu CM, Jeon HB, Shin DM. Small hypoxia-primed mesenchymal stem cells attenuate graft-versus-host disease. Leukemia 2018; 32:2672-2684. [PMID: 29789652 PMCID: PMC6286327 DOI: 10.1038/s41375-018-0151-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/12/2018] [Accepted: 04/16/2018] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are of particular interest for the treatment of immune-related diseases due to their immunosuppressive capacity. Here, we show that Small MSCs primed with Hypoxia and Calcium ions (SHC-MSCs) exhibit enhanced stemness and immunomodulatory functions for treating allogeneic conflicts. Compared with naïve cultured human umbilical cord blood-derived MSCs, SHC-MSCs were resistant to passage-dependent senescence mediated via the monocyte chemoattractant protein-1 and p53/p21 cascade and secreted large amounts of pro-angiogenic and immunomodulatory factors, resulting in suppression of T-cell proliferation. SHC-MSCs showed DNA demethylation in pluripotency, germline, and imprinted genes similarly to very small embryonic-like stem cells, suggesting a potential mutual relationship. Genome-wide DNA methylome and transcriptome analyses indicated that genes related to immune modulation, cell adhesion, and the cell cycle were up-regulated in SHC-MSCs. Particularly, polo-like kinase-1 (PLK1), zinc-finger protein-143, dehydrogenase/reductase-3, and friend-of-GATA2 play a key role in the beneficial effects of SHC-MSCs. Administration of SHC-MSCs or PLK1-overexpressing MSCs significantly ameliorated symptoms of graft-versus-host disease (GVHD) in a humanized mouse model, resulting in significantly improved survival, less weight loss, and reduced histopathologic injuries in GVHD target organs compared with naïve MSC-infused mice. Collectively, our findings suggest that SHC-MSCs can improve the clinical treatment of allogeneic conflicts, including GVHD.
Collapse
Affiliation(s)
- YongHwan Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Department of Physiology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Hye Jin Jin
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seongnam, 13494, Korea
| | - Jinbeom Heo
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Department of Physiology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Hyein Ju
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Department of Physiology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Hye-Yeon Lee
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Department of Physiology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Sujin Kim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Department of Physiology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Seungun Lee
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Department of Physiology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Jisun Lim
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Department of Physiology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Sang Young Jeong
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seongnam, 13494, Korea
| | - JiHye Kwon
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seongnam, 13494, Korea
| | - Miyeon Kim
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seongnam, 13494, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seongnam, 13494, Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seongnam, 13494, Korea
| | - Yoon Sun Yang
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seongnam, 13494, Korea
| | - Hyun Ho Hwang
- King Abdullah University of Science and Technology (KAUST), Thuwal, Jeddah, 23955-6900, Saudi Arabia
| | - Hwan Yeul Yu
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Department of Physiology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Chae-Min Ryu
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea.,Department of Physiology, University of Ulsan College of Medicine, Seoul, 05505, Korea
| | - Hong Bae Jeon
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seongnam, 13494, Korea.
| | - Dong-Myung Shin
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Korea. .,Department of Physiology, University of Ulsan College of Medicine, Seoul, 05505, Korea.
| |
Collapse
|