1
|
Hejazian SM, Hejazian SS, Mostafavi SM, Hosseiniyan SM, Montazersaheb S, Ardalan M, Zununi Vahed S, Barzegari A. Targeting cellular senescence in kidney diseases and aging: A focus on mesenchymal stem cells and their paracrine factors. Cell Commun Signal 2024; 22:609. [PMID: 39696575 DOI: 10.1186/s12964-024-01968-1] [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: 09/15/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Cellular senescence is a phenomenon distinguished by the halting of cellular division, typically triggered by DNA injury or numerous stress-inducing factors. Cellular senescence is implicated in various pathological and physiological processes and is a hallmark of aging. The presence of accumulated senescent cells, whether transiently (acute senescence) or persistently (chronic senescence) plays a dual role in various conditions such as natural kidney aging and different kidney disorders. Elevations in senescent cells and senescence-associated secretory phenotype (SASP) levels correlate with decreased kidney function, kidney ailments, and age-related conditions. Strategies involving senotherapeutic agents like senolytics, senomorphics, and senoinflammation have been devised to specifically target senescent cells. Mesenchymal stem cells (MSCs) and their secreted factors may also offer alternative approaches for anti-senescence interventions. The MSC-derived secretome compromises significant therapeutic benefits in kidney diseases by facilitating tissue repair via anti-inflammatory, anti-fibrosis, anti-apoptotic, and pro-angiogenesis effects, thereby improving kidney function and mitigating disease progression. Moreover, by promoting the clearance of senescent cells or modulating their secretory profiles, MSCs could potentially reverse some age-related declines in kidney function.This review article intends to shed light on the present discoveries concerning the role of cellular senescence in kidney aging and diseases. Furthermore, it outlines the role of senotherapeutics utilized to alleviate kidney damage and aging. It also highlights the possible impact of MSCs secretome on mitigating kidney injury and prolonging lifespan across various models of kidney diseases as a novel senotherapy.
Collapse
Affiliation(s)
| | - Seyyed Sina Hejazian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyedeh Mina Mostafavi
- Ayatollah Taleghani Hospital, Research Development Unit, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Park K, Gao WW, Zheng J, Oh KT, Kim IY, You S. Hydrogel-Mediated Local Delivery of Induced Nephron Progenitor Cell-Sourced Molecules as a Cell-Free Approach for Acute Kidney Injury. Int J Mol Sci 2024; 25:10615. [PMID: 39408943 PMCID: PMC11477367 DOI: 10.3390/ijms251910615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Acute kidney injury (AKI) constitutes a severe condition characterized by a sudden decrease in kidney function. Utilizing lineage-restricted stem/progenitor cells, directly reprogrammed from somatic cells, is a promising therapeutic option in personalized medicine for serious and incurable diseases such as AKI. The present study describes the therapeutic potential of induced nephron progenitor cell-sourced molecules (iNPC-SMs) as a cell-free strategy against cisplatin (CP)-induced nephrotoxicity, employing hyaluronic acid (HA) hydrogel-mediated local delivery to minimize systemic leakage and degradation. iNPC-SMs exhibited anti-apoptotic effects on HK-2 cells by inhibiting CP-induced ROS generation. Additionally, the localized biodistribution facilitated by hydrogel-mediated iNPC-SM delivery contributed to enhanced renal function, anti-inflammatory response, and renal regeneration in AKI mice. This study could serve as a 'proof of concept' for injectable hydrogel-mediated iNPC-SM delivery in AKI and as a model for further exploration of the development of cell-free regenerative medicine strategies.
Collapse
Affiliation(s)
- Kyoungmin Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.P.); (W.-W.G.); (J.Z.); (K.T.O.)
| | - Wei-Wei Gao
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.P.); (W.-W.G.); (J.Z.); (K.T.O.)
| | - Jie Zheng
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.P.); (W.-W.G.); (J.Z.); (K.T.O.)
| | - Kyung Taek Oh
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.P.); (W.-W.G.); (J.Z.); (K.T.O.)
| | - In-Yong Kim
- Catholic High-Performance Cell Therapy Center & Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seungkwon You
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (K.P.); (W.-W.G.); (J.Z.); (K.T.O.)
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
3
|
Tsuji K, Nakanoh H, Fukushima K, Kitamura S, Wada J. MicroRNAs as Biomarkers and Therapeutic Targets for Acute Kidney Injury. Diagnostics (Basel) 2023; 13:2893. [PMID: 37761260 PMCID: PMC10529274 DOI: 10.3390/diagnostics13182893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Acute kidney injury (AKI) is a clinical syndrome where a rapid decrease in kidney function and/or urine output is observed, which may result in the imbalance of water, electrolytes and acid base. It is associated with poor prognosis and prolonged hospitalization. Therefore, an early diagnosis and treatment to avoid the severe AKI stage are important. While several biomarkers, such as urinary L-FABP and NGAL, can be clinically useful, there is still no gold standard for the early detection of AKI and there are limited therapeutic options against AKI. miRNAs are non-coding and single-stranded RNAs that silence their target genes in the post-transcriptional process and are involved in a wide range of biological processes. Recent accumulated evidence has revealed that miRNAs may be potential biomarkers and therapeutic targets for AKI. In this review article, we summarize the current knowledge about miRNAs as promising biomarkers and potential therapeutic targets for AKI, as well as the challenges in their clinical use.
Collapse
Affiliation(s)
- Kenji Tsuji
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hiroyuki Nakanoh
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Kazuhiko Fukushima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Program in Membrane Biology, Center for Systems Biology, Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Shinji Kitamura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Department of Nursing Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama 719-1197, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
4
|
Tsuji K, Kitamura S, Wada J. Potential Strategies for Kidney Regeneration With Stem Cells: An Overview. Front Cell Dev Biol 2022; 10:892356. [PMID: 35586342 PMCID: PMC9108336 DOI: 10.3389/fcell.2022.892356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/19/2022] [Indexed: 11/28/2022] Open
Abstract
Kidney diseases are a major health problem worldwide. Despite advances in drug therapies, they are only capable of slowing the progression of kidney diseases. Accordingly, potential kidney regeneration strategies with stem cells have begun to be explored. There are two different directions for regenerative strategies, de novo whole kidney fabrication with stem cells, and stem cell therapy. De novo whole kidney strategies include: 1) decellularized scaffold technology, 2) 3D bioprinting based on engineering technology, 3) kidney organoid fabrication, 4) blastocyst complementation with chimeric technology, and 5) the organogenic niche method. Meanwhile, stem cell therapy strategies include 1) injection of stem cells, including mesenchymal stem cells, nephron progenitor cells, adult kidney stem cells and multi-lineage differentiating stress enduring cells, and 2) injection of protective factors secreted from these stem cells, including growth factors, chemokines, and extracellular vesicles containing microRNAs, mRNAs and proteins. Over the past few decades, there have been remarkable step-by-step developments in these strategies. Here, we review the current advances in the potential strategies for kidney regeneration using stem cells, along with their challenges for possible clinical use in the future.
Collapse
|
5
|
German OL, Vallese-Maurizi H, Soto TB, Rotstein NP, Politi LE. Retina stem cells, hopes and obstacles. World J Stem Cells 2021; 13:1446-1479. [PMID: 34786153 PMCID: PMC8567457 DOI: 10.4252/wjsc.v13.i10.1446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/14/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023] Open
Abstract
Retinal degeneration is a major contributor to visual dysfunction worldwide. Although it comprises several eye diseases, loss of retinal pigment epithelial (RPE) and photoreceptor cells are the major contributors to their pathogenesis. Early therapies included diverse treatments, such as provision of anti-vascular endothelial growth factor and many survival and trophic factors that, in some cases, slow down the progression of the degeneration, but do not effectively prevent it. The finding of stem cells (SC) in the eye has led to the proposal of cell replacement strategies for retina degeneration. Therapies using different types of SC, such as retinal progenitor cells (RPCs), embryonic SC, pluripotent SCs (PSCs), induced PSCs (iPSCs), and mesenchymal stromal cells, capable of self-renewal and of differentiating into multiple cell types, have gained ample support. Numerous preclinical studies have assessed transplantation of SC in animal models, with encouraging results. The aim of this work is to revise the different preclinical and clinical approaches, analyzing the SC type used, their efficacy, safety, cell attachment and integration, absence of tumor formation and immunorejection, in order to establish which were the most relevant and successful. In addition, we examine the questions and concerns still open in the field. The data demonstrate the existence of two main approaches, aimed at replacing either RPE cells or photoreceptors. Emerging evidence suggests that RPCs and iPSC are the best candidates, presenting no ethical concerns and a low risk of immunorejection. Clinical trials have already supported the safety and efficacy of SC treatments. Serious concerns are pending, such as the risk of tumor formation, lack of attachment or integration of transplanted cells into host retinas, immunorejection, cell death, and also ethical. However, the amazing progress in the field in the last few years makes it possible to envisage safe and effective treatments to restore vision loss in a near future.
Collapse
Affiliation(s)
- Olga L German
- Department of Biology, Biochemistry and Pharmacy, Universidad Nacional del Sur, Bahia blanca 8000, Buenos Aires, Argentina
- Department of Biology, Biochemistry and Pharmacy, Universidad Nacional del Sur, and Neurobiology Department, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) Conicet, Bahía Blanca 8000, Buenos Aires, Argentina
| | - Harmonie Vallese-Maurizi
- Department of Biology, Biochemistry and Pharmacy, Universidad Nacional del Sur, Bahia blanca 8000, Buenos Aires, Argentina
- Department of Biology, Biochemistry and Pharmacy, Universidad Nacional del Sur, and Neurobiology Department, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) Conicet, Bahía Blanca 8000, Buenos Aires, Argentina
| | - Tamara B Soto
- Department of Biology, Biochemistry and Pharmacy, Universidad Nacional del Sur, and Neurobiology Department, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) Conicet, Bahía Blanca 8000, Buenos Aires, Argentina
| | - Nora P Rotstein
- Department of Biology, Biochemistry and Pharmacy, Universidad Nacional del Sur, Bahia blanca 8000, Buenos Aires, Argentina
- Department of Biology, Biochemistry and Pharmacy, Universidad Nacional del Sur, and Neurobiology Department, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) Conicet, Bahía Blanca 8000, Buenos Aires, Argentina
| | - Luis Enrique Politi
- Department of Biology, Biochemistry and Pharmacy, Universidad Nacional del Sur, and Neurobiology Department, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB) Conicet, Bahía Blanca 8000, Buenos Aires, Argentina
| |
Collapse
|
6
|
Habib R. Multifaceted roles of Toll-like receptors in acute kidney injury. Heliyon 2021; 7:e06441. [PMID: 33732942 PMCID: PMC7944035 DOI: 10.1016/j.heliyon.2021.e06441] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/08/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern recognition receptors (PRRs) in the first line defense system of our bodies; they are widely expressed on leukocytes and kidney epithelial cells. Infections due to pathogens or danger signals from injured tissues often activate several TLRs and these receptors mediate their signal transduction through the activation of transcription factors that regulate the expression of cytokine interleukin-1β (IL-1β), type I interferons (IFNs), and nuclear factor kappa light chain enhancer of activated B cells (NF-κB) dependent cytokines and chemokines. Acute kidney injury (AKI) involves early Toll-like receptors driven immunopathology, while resolution of inflammation is needed for rapid regeneration of injured tubular cells. Despite their well known function in the progression of inflammation; interestingly, activation of TLRs also has been implicated in renal epithelial repair through the induction of certain interleukins and improvement in autophagy mechanism. Studies have found that although the blockade of TLRs during the early injury phase of renal tissues prevented tubular necrosis, suppression of interleukins production and impaired kidney regeneration due to their blockade has been observed during the healing phase of tissue. Taken together, these results suggest that the two danger response programs of renal cells i.e. renal inflammation and regeneration may link at the level of TLRs. This review aims to emphasize on the role of TLRs signaling in different acute kidney injury phases. Understanding of these pathways may turn out to be effective as therapeutic option for kidney diseases.
Collapse
Affiliation(s)
- Rakhshinda Habib
- Dow Research Institute of Biotechnology and Biomedical Sciences, Dow University of Health Sciences, Karachi, 74200, Pakistan
| |
Collapse
|
7
|
Mohammadi B, Esmaeilizadeh Z, Rajabibazl M, Ghaderian SMH, Omrani MD, Fazeli Z. Preconditioning of human adipose tissue-derived mesenchymal stem cells with HEK293-coditioned media can influence on the expression of BMP2, BMP6 and BMP11: Potential application in the treatment of renal lesions. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
8
|
Zinger A, Brozovich A, Pasto A, Sushnitha M, Martinez JO, Evangelopoulos M, Boada C, Tasciotti E, Taraballi F. Bioinspired Extracellular Vesicles: Lessons Learned From Nature for Biomedicine and Bioengineering. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2172. [PMID: 33143238 PMCID: PMC7693812 DOI: 10.3390/nano10112172] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/23/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022]
Abstract
Efficient communication is essential in all layers of the biological chain. Cells exchange information using a variety of signaling moieties, such as small molecules, proteins, and nucleic acids. Cells carefully package these messages into lipid complexes, collectively named extracellular vesicles (EVs). In this work, we discuss the nature of these cell carriers, categorize them by their origin, explore their role in the homeostasis of healthy tissues, and examine how they regulate the pathophysiology of several diseases. This review will also address the limitations of using EVs for clinical applications and discuss novel methods to engineer nanoparticles to mimic the structure, function, and features of EVs. Using lessons learned from nature and understanding how cells use EVs to communicate across distant sites, we can develop a better understanding of how to tailor the fundamental features of drug delivery carriers to encapsulate various cargos and target specific sites for biomedicine and bioengineering.
Collapse
Affiliation(s)
- Assaf Zinger
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ava Brozovich
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Texas A&M College of Medicine, Bryan, TX 77807, USA
| | - Anna Pasto
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Italy
| | - Manuela Sushnitha
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Jonathan O. Martinez
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Michael Evangelopoulos
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Christian Boada
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ennio Tasciotti
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Biotechnology Program, San Raffaele University, Via di Val Cannuta, 247, 00166 Roma RM, Italy
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
9
|
Tsuji K, Kitamura S, Sang Y, Fukushima K, Wada J. Adult kidney stem/progenitor cells contribute to regeneration through the secretion of trophic factors. Stem Cell Res 2020; 46:101865. [PMID: 32505897 DOI: 10.1016/j.scr.2020.101865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 05/12/2020] [Accepted: 05/22/2020] [Indexed: 12/20/2022] Open
Abstract
Adult kidney stem cells are known to have important roles in renal regeneration after acute kidney injury. Although trophic factors from tissue stem cells have been reported to promote the regeneration of other organs, there is limited number of evidence of this phenomenon in the kidneys. Here, we explored the effects of secreted factors from kidney stem cells. We intraperitoneally administered culture supernatant obtained from adult rat kidney stem/progenitor cells into rat kidney ischemia/reperfusion injury models, and the treatment significantly ameliorated renal tubulointerstitial injury, suppressed tubular cell apoptosis, diminished inflammation and promoted the proliferation of both residual renal cells and immature cells. In vitro, treatment with culture supernatant from kidney stem cells significantly promoted cell proliferation and suppressed cisplatin-induced cell apoptosis in both normal rat kidney cells and kidney stem cells. In addition, treatment with culture supernatant increased the expression of nestin in normal rat kidney cells, suggesting the dedifferentiation of tubular cells into stem-like cells. Analysis of the culture supernatant revealed that it contained a variety of growth factors. Taken together, the results suggest that these factors together lead to renal regeneration. In conclusion, adult kidney stem cells contribute to renal regeneration indirectly through the secretion of regenerative factors.
Collapse
Affiliation(s)
- Kenji Tsuji
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.
| | - Shinji Kitamura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.
| | - Yizhen Sang
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiko Fukushima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
10
|
Saberi K, Pasbakhsh P, Omidi A, Borhani-Haghighi M, Nekoonam S, Omidi N, Ghasemi S, Kashani IR. Melatonin preconditioning of bone marrow-derived mesenchymal stem cells promotes their engraftment and improves renal regeneration in a rat model of chronic kidney disease. J Mol Histol 2019; 50:129-140. [PMID: 30671880 DOI: 10.1007/s10735-019-09812-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/06/2019] [Indexed: 12/26/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BMMSCs) transplantation has shown to be effective in treating chronic kidney disease. However, the effectiveness of this strategy is constrained by low homing and survival rate of transplanted cells in the injured organs. Therefore, developing strategies to improve homing and cell survival rate and therapeutic potential in cell-based therapies seems necessary. The purpose of this study is to evaluate the effect of pretreating BMMSCs with melatonin (MT) on the prosurvival and renoprotective of transplanted cells into the irreversible model of unilateral ureteral obstruction. Adult male Sprague-Dawley rats were randomized into four groups: Sham, UUO, UUO + BMMSCs, and UUO + BMMSCs + MT. The results of our study demonstrated that preconditioning with MT enhanced homing of BMMSCs into the injured kidney. MT reduced the number of TUNEL positive transplanted cells in the UUO + BMMSCs + MT group. The UUO + BMMSCs + MT group showed lower expressions of TGF-β1, α-SMA and TNF-α at both gene and protein levels but higher expression of E-cadherin compared with the UUO + BMMSCs group. In addition, MT preconditioned BMMSCs ameliorated basement membrane disruption and histological status of injured renal tubules and also reduced fibrosis in damaged kidneys. In conclusion, our results show that stem cells pretreated by MT may represent a feasible approach for improving the beneficial effects of stem cell therapy and significantly enhance their survival after transplantation to the injured kidney.
Collapse
Affiliation(s)
- Kamran Saberi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ameneh Omidi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Shafa Neuroscience Research Center, KhatamAlanbia Hospital, Tehran, Iran
| | - Saeid Nekoonam
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Negar Omidi
- Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sodabeh Ghasemi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Oliveira M, Lira R, Freire T, Luna C, Martins M, Almeida A, Carvalho S, Cortez E, Stumbo AC, Thole A, Carvalho L. Bone marrow mononuclear cell transplantation rescues the glomerular filtration barrier and epithelial cellular junctions in a renovascular hypertension model. Exp Physiol 2019; 104:740-754. [DOI: 10.1113/ep087330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/19/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Mariana Oliveira
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Rafaelle Lira
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Thiago Freire
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Camila Luna
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcela Martins
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Aline Almeida
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Simone Carvalho
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Erika Cortez
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Carolina Stumbo
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Alessandra Thole
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lais Carvalho
- Laboratory of Stem Cell ResearchHistology and Embryology DepartmentBiology InstituteState University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
12
|
Choi JY, Chun SY, Ha YS, Kim DH, Kim J, Song PH, Kim HT, Yoo ES, Kim BS, Kwon TG. Potency of Human Urine-Derived Stem Cells for Renal Lineage Differentiation. Tissue Eng Regen Med 2017; 14:775-785. [PMID: 30603527 PMCID: PMC6171660 DOI: 10.1007/s13770-017-0081-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/01/2017] [Accepted: 09/03/2017] [Indexed: 01/09/2023] Open
Abstract
Kidney is one of the most difficult organs for regeneration. Several attempts have been performed to regenerate renal tissue using stem cells, the results were not satisfactory. Urine is major product of kidney and contains cells from renal components. Moreover, urine-derived stem cells (USCs) can be easily obtained without any health risks throughout a patient's entire life. Here, we evaluated the utility of USCs for renal tissue regeneration. In this study, the ability of USCs to differentiate into renal lineage cells was compared with that of adipose tissue-derived stem cells (ADSCs) and amniotic fluid-derived stem cells (AFSCs), with respect to surface antigen expression, morphology, immunocytochemistry, renal lineage gene expression, secreted factors, immunomodulatory marker expression, in vivo safety, and renal differentiation potency. Undifferentiated USCs were positive for CD44 and CD73, negative for CD34 and CD45, and formed aggregates after 3 weeks of renal differentiation. Undifferentiated USCs showed high SSEA4 expression, while renal-differentiated cells expressed PAX2, WT1, and CADHERIN 6. In the stem/renal lineage-associated gene analysis, OCT4, SSEA4, and CD117 were significantly downregulated over time, while PAX2, LIM1, PDGFRA, E-CADHERIN, CD24, ACTB, AQP1, OCLN, and NPHS1 were gradually upregulated. In the in vivo safety evaluation, renal-differentiated USCs did not show abnormal histology. These findings demonstrated that USCs have a similar MSC potency, renal lineage-differentiation ability, immunomodulatory effects, and in vivo safety as ADSCs and AFSCs, and showed higher levels of growth factor secretion for paracrine effects. Therefore, urine and USCs can be one of good cell sources for kidney regeneration.
Collapse
Affiliation(s)
- Jae Young Choi
- Department of Urology, College of Medicine, Yeungnam University, 170 Hyunchung-ro, Nam-gu, Daegu, 42415 Korea
| | - So Young Chun
- Biomedical Research Institute, Kyungpook National University Hospital, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
| | - Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
| | - Dae Hwan Kim
- Department of Laboratory Animal Research Support Team, Yeungnam University Medical Center, 170 Hyunchung-ro, Nam-gu, Daegu, 42415 Korea
| | - Jeongshik Kim
- Department of Pathology, Central Hospital, 480 Munsu-ro, Nam-gu, Ulsan, 44667 Korea
| | - Phil Hyun Song
- Department of Urology, College of Medicine, Yeungnam University, 170 Hyunchung-ro, Nam-gu, Daegu, 42415 Korea
| | - Hyun Tae Kim
- Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
| | - Eun Sang Yoo
- Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
| | - Bum Soo Kim
- Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
| | - Tae Gyun Kwon
- Department of Urology, School of Medicine, Kyungpook National University, 130 Dongdeok-ro, Jung-gu, Daegu, 41944 Korea
- Department of Urology, Kyungpook National University Chilgok Hospital, 807 Hogukro, Buk-gu, Daegu, 41404 Korea
| |
Collapse
|
13
|
Nawaz M, Fatima F. Extracellular Vesicles, Tunneling Nanotubes, and Cellular Interplay: Synergies and Missing Links. Front Mol Biosci 2017; 4:50. [PMID: 28770210 PMCID: PMC5513920 DOI: 10.3389/fmolb.2017.00050] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 07/03/2017] [Indexed: 12/15/2022] Open
Abstract
The process of intercellular communication seems to have been a highly conserved evolutionary process. Higher eukaryotes use several means of intercellular communication to address both the changing physiological demands of the body and to fight against diseases. In recent years, there has been an increasing interest in understanding how cell-derived nanovesicles, known as extracellular vesicles (EVs), can function as normal paracrine mediators of intercellular communication, but can also elicit disease progression and may be used for innovative therapies. Over the last decade, a large body of evidence has accumulated to show that cells use cytoplasmic extensions comprising open-ended channels called tunneling nanotubes (TNTs) to connect cells at a long distance and facilitate the exchange of cytoplasmic material. TNTs are a different means of communication to classical gap junctions or cell fusions; since they are characterized by long distance bridging that transfers cytoplasmic organelles and intracellular vesicles between cells and represent the process of heteroplasmy. The role of EVs in cell communication is relatively well-understood, but how TNTs fit into this process is just emerging. The aim of this review is to describe the relationship between TNTs and EVs, and to discuss the synergies between these two crucial processes in the context of normal cellular cross-talk, physiological roles, modulation of immune responses, development of diseases, and their combinatory effects in tissue repair. At the present time this review appears to be the first summary of the implications of the overlapping roles of TNTs and EVs. We believe that a better appreciation of these parallel processes will improve our understanding on how these nanoscale conduits can be utilized as novel tools for targeted therapies.
Collapse
Affiliation(s)
- Muhammad Nawaz
- Department of Pathology and Forensic Medicine, Ribeirao Preto Medical School, University of São PauloSão Paulo, Brazil.,Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of GothenburgGothenburg, Sweden
| | - Farah Fatima
- Department of Pathology and Forensic Medicine, Ribeirao Preto Medical School, University of São PauloSão Paulo, Brazil
| |
Collapse
|
14
|
Burrello J, Monticone S, Gai C, Gomez Y, Kholia S, Camussi G. Stem Cell-Derived Extracellular Vesicles and Immune-Modulation. Front Cell Dev Biol 2016; 4:83. [PMID: 27597941 PMCID: PMC4992732 DOI: 10.3389/fcell.2016.00083] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/02/2016] [Indexed: 12/13/2022] Open
Abstract
Extra-cellular vesicles (EVs) are bilayer membrane structures enriched with proteins, nucleic acids, and other active molecules and have been implicated in many physiological and pathological processes over the past decade. Recently, evidence suggests EVs to play a more dichotomic role in the regulation of the immune system, whereby an immune response may be enhanced or supressed by EVs depending on their cell of origin and its functional state. EVs derived from antigen (Ag)-presenting cells for instance, have been involved in both innate and acquired (or adaptive) immune responses, as Ag carriers or presenters, or as vehicles for delivering active signaling molecules. On the other hand, tumor and stem cell derived EVs have been identified to exert an inhibitory effect on immune responses by carrying immuno-modulatory effectors, such as transcriptional factors, non-coding RNA (Species), and cytokines. In addition, stem cell-derived EVs have also been reported to impair dendritic cell maturation and to regulate the activation, differentiation, and proliferation of B cells. They have been shown to control natural killer cell activity and to suppress the innate immune response (IIR). Studies reporting the role of EVs on T lymphocyte modulation are controversial. Discrepancy in literature may be due to stem cell culture conditions, methods of EV purification, EV molecular content, and functional state of both parental and target cells. However, mesenchymal stem cell-derived EVs were shown to play a more suppressive role by shifting T cells from an activated to a T regulatory phenotype. In this review, we will discuss how stem cell-derived EVs may contribute toward the modulation of the immune response. Collectively, stem cell-derived EVs mainly exhibit an inhibitory effect on the immune system.
Collapse
Affiliation(s)
- Jacopo Burrello
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| | - Silvia Monticone
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| | - Chiara Gai
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| | - Yonathan Gomez
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| | - Sharad Kholia
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| | - Giovanni Camussi
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| |
Collapse
|
15
|
Funderburgh JL, Funderburgh ML, Du Y. Stem Cells in the Limbal Stroma. Ocul Surf 2016; 14:113-20. [PMID: 26804252 DOI: 10.1016/j.jtos.2015.12.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/16/2015] [Accepted: 12/24/2015] [Indexed: 12/13/2022]
Abstract
The corneal stroma contains a population of mesenchymal cells subjacent to the limbal basement membrane with characteristics of adult stem cells. These 'niche cells' support limbal epithelial stem cell viability. In culture by themselves, the niche cells display a phenotype typical of mesenchymal stem cells. These stromal stem cells exhibit a potential to differentiate to multiple cell types, including keratocytes, thus providing an abundant source of these rare cells for experimental and bioengineering applications. Stromal stem cells have also shown the ability to remodel pathological stromal tissue, suppressing inflammation and restoring transparency. Because stromal stem cells can be obtained by biopsy, they offer a potential for autologous stem cell treatment for stromal opacities. This review provides an overview of the status of work on this interesting cell population.
Collapse
Affiliation(s)
- James L Funderburgh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213.
| | - Martha L Funderburgh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Yiqin Du
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| |
Collapse
|
16
|
Wang J, Lin G, Alwaal A, Zhang X, Wang G, Jia X, Banie L, Villalta J, Lin CS, Lue TF. Kinetics of Label Retaining Cells in the Developing Rat Kidneys. PLoS One 2015; 10:e0144734. [PMID: 26650841 PMCID: PMC4674088 DOI: 10.1371/journal.pone.0144734] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 11/23/2015] [Indexed: 12/23/2022] Open
Abstract
Background The kidney is a specialized low-regenerative organ with several different types of cellular lineages. The BrdU label-retaining cell (LRCs) approach has been used as part of a strategy to identify tissue-specific stem cells in the kidney; however, because the complementary base pairing in double-stranded DNA blocks the access of the anti-BrdU antibody to BrdU subunits, the stem cell marker expression in BrdU-labeled cells are often difficult to detect. In this study, we introduced a new cell labeling and detection method in which BrdU was replaced with 5-ethynyl-2-deoxyuridine (EdU) and examined the time-dependent dynamic changes of EdU-labeled cells and potential stem/progenitor markers in the development of kidney. Methods Newborn rats were intraperitoneally injected with EdU, and their kidneys were harvested respectively at different time points at 1 day, 3 days, 1 week, 2 weeks, and 6 weeks post-injection. The kidney tissues were processed for EdU and cellular markers by immunofluorescence staining. Results At the early stage, LRCs labeled by EdU were 2176.0 ± 355.6 cells at day one in each renal tissue section, but dropped to 168 ± 48.4 cells by week 6. As time increased, the numbers of LRCs were differentially expressed in the renal cortex and papilla. At the postnatal day one, nearly twice as many cells in the cortex were EdU-labeled as compared to the papilla (28.6 ± 3.6% vs. 15.6 ± 3.4%, P<0.05), while there were more LRCs within the renal papilla since the postnatal week one, and at the postnatal week 6, one third as many cells in the cortex were EdU-labeled as compared to the papilla (2.5 ± 0.1% vs. 7.7 ± 2.7%, P<0.05). The long-term LRCs at 6-week time point were associated exclusively with the glomeruli in the cortex and the renal tubules in the papilla. At 6 weeks, the EdU-labeled LRCs combined with expression of CD34, RECA-1, Nestin, and Synaptopodin were discretely but widely distributed within the glomeruli; Stro-1 around the glomeruli; and α-smooth muscle actin (SMA) in arteries. Conversely, co-expression of CD34, RECA-1, and Nestin with the long term EdU-labeled LRCs was significantly lower in renal tubules (P<0.01), while Stro-1 and Synaptopodin were not detected. Conclusion Our data found that at 6-week time point, EdU-labeled LRCs existing in the glomeruli expressed undifferentiated podocyte and endothelial markers at high rates, while those in the renal tubules expressed Nestin and vascular markers at low rates. To understand the characterization and localization of these EdU-LRCs, further studies will be needed to test cell lineage tracing, clonogenicity and differentiation potency, and the contributions to the regeneration of the kidney in response to renal injury/repair.
Collapse
Affiliation(s)
- Jianwen Wang
- Department of Urology, Beijing ChaoYang Hospital, Capital Medical University, 8 Gongtinanlu, Beijing, 100020, China
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
- * E-mail:
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Amjad Alwaal
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Xiaoyu Zhang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Xingyuan Jia
- Department of Urology, Beijing ChaoYang Hospital, Capital Medical University, 8 Gongtinanlu, Beijing, 100020, China
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Jacqueline Villalta
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Ching-Shwun Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Tom F. Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| |
Collapse
|
17
|
Extracellular Vesicles: Evolving Factors in Stem Cell Biology. Stem Cells Int 2015; 2016:1073140. [PMID: 26649044 PMCID: PMC4663346 DOI: 10.1155/2016/1073140] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/09/2015] [Accepted: 07/16/2015] [Indexed: 12/18/2022] Open
Abstract
Stem cells are proposed to continuously secrete trophic factors that potentially serve as mediators of autocrine and paracrine activities, associated with reprogramming of the tumor microenvironment, tissue regeneration, and repair. Hitherto, significant efforts have been made to understand the level of underlying paracrine activities influenced by stem cell secreted trophic factors, as little is known about these interactions. Recent findings, however, elucidate this role by reporting the effects of stem cell derived extracellular vesicles (EVs) that mimic the phenotypes of the cells from which they originate. Exchange of genetic information utilizing persistent bidirectional communication mediated by stem cell-EVs could regulate stemness, self-renewal, and differentiation in stem cells and their subpopulations. This review therefore discusses stem cell-EVs as evolving communication factors in stem cell biology, focusing on how they regulate cell fates by inducing persistent and prolonged genetic reprogramming of resident cells in a paracrine fashion. In addition, we address the role of stem cell-secreted vesicles in shaping the tumor microenvironment and immunomodulation and in their ability to stimulate endogenous repair processes during tissue damage. Collectively, these functions ensure an enormous potential for future therapies.
Collapse
|