1
|
Jing D, Liu Q, Zhang H, Li Y, Jiang X, Cai Y, Wang X, Li L. miR-548az-5p induces amniotic epithelial cell senescence by regulating KATNAL1 expression in labor. Sci Rep 2024; 14:30380. [PMID: 39638877 PMCID: PMC11621115 DOI: 10.1038/s41598-024-82390-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/04/2024] [Indexed: 12/07/2024] Open
Abstract
Amniotic fluid exosomes (AF-Exos) from term labor (TL) cause amniotic membrane senescence and induce labor. However, the intrinsic mechanism through which this occurs remains unknown. Therefore, we performed microRNA (miRNA) microarray chip screening of AF-Exos obtained from TL and terms not in labor and discovered that the expression of miR-548az-5p was significantly upregulated in TL. This study aimed to explore the role of miR-548az-5p in AF-Exos-induced human amniotic epithelial cells (hAECs) senescence for labor initiation. Bioinformatics analysis revealed that Katanin catalytic subunit A1 like 1 (KATNAL1) is a potential miR-548az-5p target. In hAECs, the upregulation of miR-548az-5p suppressed KATNAL1 expression, disorganized microtubules, increased senescence-associated secretory phenotype-related biomarkers, and inhibited cell proliferation by cyclin D1 and cyclin-dependent kinase 6 (CDK6). This study identified that miR-548az-5p is involved in the senescence of amniotic epithelial cells by targeting KATNAL1 to induce labor. Notably, this study offers new perspectives on the mediation of cellular senescence using AF-Exos miRNAs, which results in labor.
Collapse
Affiliation(s)
- Die Jing
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, 250117, Shandong, China
| | - Qian Liu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
- Department of Obstetrics and Gynecology, Feixian County People's Hospital, Linyi, 273400, Shandong, China
| | - Hongyuan Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yuchen Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, 250117, Shandong, China
| | - Xiaotong Jiang
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, 250117, Shandong, China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Yanjun Cai
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, 250117, Shandong, China
| | - Xietong Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, 250117, Shandong, China.
- The Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, 250014, Shandong, China.
| | - Lei Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan, 250117, Shandong, China.
| |
Collapse
|
2
|
Zhao M, Liu S, Wang Y, Lv K, Lou P, Zhou P, Zhu J, Li L, Cheng J, Lu Y, Liu J. The mitochondria‒paraspeckle axis regulates the survival of transplanted stem cells under oxidative stress conditions. Theranostics 2024; 14:1517-1533. [PMID: 38389853 PMCID: PMC10879866 DOI: 10.7150/thno.88764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Rationale: Stem cell-based therapies have emerged as promising tools for tissue engineering and regenerative medicine, but their therapeutic efficacy is largely limited by the oxidative stress-induced loss of transplanted cells at injured tissue sites. To address this issue, we aimed to explore the underlying mechanism and protective strategy of ROS-induced MSC loss. Methods: Changes in TFAM (mitochondrial transcription factor A) signaling, mitochondrial function, DNA damage, apoptosis and senescence in MSCs under oxidative stress conditions were assessed using real-time PCR, western blotting and RNA sequencing, etc. The impact of TFAM or lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) knockdown or overexpression on mitochondrial function, DNA damage repair, apoptosis and senescence in MSCs was also analyzed. The effect of mitochondrion-targeted antioxidant (Mito-TEMPO) on the survival of transplanted MSCs was evaluated in a mouse model of renal ischemia/reperfusion (I/R) injury. Results: Mitochondrial ROS (mtROS) bursts caused defects in TFAM signaling and overall mitochondrial function, which further impaired NEAT1 expression and its mediated paraspeckle formation and DNA repair pathways in MSCs, thereby jointly promoting MSC senescence and death under oxidative stress. In contrast, targeted inhibition of the mtROS bursts is a sufficient strategy for attenuating early transplanted MSC loss at injured tissue sites, and coadministration of Mito-TEMPO improved the local retention of transplanted MSCs and reduced oxidative injury in ischemic kidneys. Conclusions: This study identified the critical role of the mitochondria‒paraspeckle axis in regulating cell survival and may provide insights into developing advanced stem cell therapies for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Meng Zhao
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Shuyun Liu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yizhuo Wang
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ke Lv
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peng Lou
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pingya Zhou
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaying Zhu
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Lan Li
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingqiu Cheng
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanrong Lu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingping Liu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Chou Y, Alfarafisa NM, Ikezawa M, Khairani AF. Progress in the Development of Stem Cell-Derived Cell-Free Therapies for Skin Aging. Clin Cosmet Investig Dermatol 2023; 16:3383-3406. [PMID: 38021432 PMCID: PMC10676866 DOI: 10.2147/ccid.s434439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023]
Abstract
Introduction The skin is a vital organ as the body's largest barrier, but its function declines with aging. Therefore, research into effective regeneration treatments must continue to advance. Stem cell transplantation, a cell-based therapy, has become a popular skin-aging treatment, although it comes with drawbacks like host immune reactions. Stem cell-derived cell-free therapies have emerged as an alternative, backed by promising preclinical findings. Stem cell secretomes and extracellular vesicles (EVs) are the key components in cell-free therapy from stem cells. However, comprehensive reviews on the mechanisms of such treatments for skin aging are still limited. Purpose This review discusses stem cell-derived cell-free therapy's potential mechanisms of action related to skin aging prevention by identifying specific molecular targets suitable for the interventions. Methods A search identified 27 relevant in vitro studies on stem cell-derived cell-free therapy interventions in skin aging model cells without restricting publication years using PubMed, Scopus, and Google Scholar. Results Stem cell-derived cell-free therapy can prevent skin aging through various mechanisms, such as (1) involvement of multiple regenerative pathways [NFkb, AP-1, MAPK, P-AKT, NRF2, SIRT-1]; (2) oxidative stress regulation [by reducing oxidants (HO-1, NQO1) and enhancing antioxidants (SOD1, CAT, GP, FRAP)]; (3) preventing ECM degradation [by increasing elastin, collagen, HA, TIMP, and reducing MMP]; (4) regulating cell activity [by reducing cell senescence (SA-β-gal), apoptosis, and cell cycle arrest (P53, P12, P16); and enhancing autophagy, cell migration, and cell proliferation (Ki67)] (5) Regulating the inflammatory pathway [by reducing IL-6, IL-1, TNF-⍺, and increasing TGF-β]. Several clinical trials have also revealed improvements in wrinkles, elasticity, hydration, pores, and pigmentation. Conclusion Stem cell-derived cell-free therapy is a potential novel treatment for skin aging by cell rejuvenation through various molecular mechanisms.
Collapse
Affiliation(s)
- Yoan Chou
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| | - Nayla Majeda Alfarafisa
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| | - Maiko Ikezawa
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Astrid Feinisa Khairani
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| |
Collapse
|
4
|
Anti-Aging Effect and Mechanism of Proanthocyanidins Extracted from Sea buckthorn on Hydrogen Peroxide-Induced Aging Human Skin Fibroblasts. Antioxidants (Basel) 2022; 11:antiox11101900. [PMID: 36290623 PMCID: PMC9598642 DOI: 10.3390/antiox11101900] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022] Open
Abstract
Oxidative stress is the leading cause of skin aging damage. Excessive accumulation of reactive oxygen species (ROS) in cells induced by hydrogen peroxide (H2O2) triggers a decrease in collagen synthesis and an increase in collagen degradation, which are biomarkers of skin aging. We evaluated the potential protective mechanism of Sea buckthorn proanthocyanidins (SBP) against the oxidative stress-induced skin aging process from multiple aspects. We treated human skin fibroblasts (HSFs) with 300 µmoL/L of H2O2 for 24 h, followed by 25, 50, and 100 µg/mL of SBP for 24 h. The results showed that SBP could enhance the activities of superoxide dismutase (SOD) and glutathione (GSH), effectively remove excess ROS, and significantly improve the changes in cell morphology and viability caused by excessive ROS in skin cells. In addition, SBP could promote the synthesis of Col I in aging HSFs through the TGF-β1/Smads pathway and inhibit the degradation of Col I by regulating the MMPs/TIMPs system, thereby maintaining the stability of the ECM structure to achieve anti-aging purposes. Finally, we studied the migration ability of SBP, and the results showed that 100 µg/mL of SBP was most conducive to the cell migration of senescent cells, laying a foundation for follow-up animal experiments. These results will increase the application value of SBP in the cosmetic and antioxidative functional food industries.
Collapse
|
5
|
He S, Zhou M, Zheng H, Wang Y, Wu S, Gao Y, Chen J. Resveratrol inhibits the progression of premature senescence partially by regulating v-rel avian reticuloendotheliosis viral oncogene homolog A (RELA) and sirtuin 1 (SIRT1). Ren Fail 2022; 44:171-183. [PMID: 35166167 PMCID: PMC8856048 DOI: 10.1080/0886022x.2022.2029488] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Objective To explore the effect of resveratrol in premature senescence and reveal its anti-premature senescence mechanisms through network pharmacology. Methods In this study, the H2O2-induced bone marrow mesenchymal stem cells (BMMSCs) premature senescence model is applied. Cell counting kit-8 assay, β-galactosidase staining and flow cytometry are conducted to detect the proliferation, senescence and apoptosis of BMMSCs. Bioinformatics analyses are used to screen and validate molecular targets of resveratrol acting on premature senescence. Dual-luciferase reporter assay is conducted to verify the interaction between v-rel avian reticuloendotheliosis viral oncogene homolog A (RELA) and sirtuin 1 (SIRT1). RT-qPCR and western blot are adopted to detect mRNA and protein levels of RELA, SIRT1, senescence-related genes and apoptosis-related genes. Results First, we proved that resveratrol alleviated the H2O2-induced senescence of BMMSCs. Then, bioinformatics analysis revealed that RELA was the downstream target of resveratrol and SIRT1 was the downstream target of RELA, respectively, involved in premature aging. RELA/SIRT1 may be the potential target of resveratrol for premature senescence. Notably, rescue experiments indicated that resveratrol inhibited premature senescence partially through targeting regulation RELA/SIRT1. Conclusion In our study, we confirm the functional role of the resveratrol-RELA- SIRT1 axis in the progression of premature senescence, which provides a latent target for premature senescence treatment.
Collapse
Affiliation(s)
- Shuangjun He
- Department of Orthopedic Surgery, Affiliated Danyang Hospital of Nantong University, The People's Hospital of Danyang, Danyang, China
| | - Meng Zhou
- Department of Orthopedic Surgery, Tengzhou Central People's Hospital, Tengzhou, China
| | - Hongming Zheng
- Department of Orthopedic Surgery, Affiliated Danyang Hospital of Nantong University, The People's Hospital of Danyang, Danyang, China
| | - Yaowei Wang
- Department of Orthopedic Surgery, Affiliated Danyang Hospital of Nantong University, The People's Hospital of Danyang, Danyang, China
| | - Shuhua Wu
- Department of Orthopedic Surgery, Affiliated Danyang Hospital of Nantong University, The People's Hospital of Danyang, Danyang, China
| | - Yuan Gao
- Department of Forensic Science, Medical School of Soochow University, Suzhou, China
| | - Jianhong Chen
- Department of Orthopedic Surgery, Affiliated Danyang Hospital of Nantong University, The People's Hospital of Danyang, Danyang, China
| |
Collapse
|
6
|
Fathi I, Miki T. Human Amniotic Epithelial Cells Secretome: Components, Bioactivity, and Challenges. Front Med (Lausanne) 2022; 8:763141. [PMID: 35083233 PMCID: PMC8784524 DOI: 10.3389/fmed.2021.763141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Human amniotic epithelial cells (hAECs) derived from placental tissue have received significant attention as a promising tool in regenerative medicine. Several studies demonstrated their anti-inflammatory, anti-fibrotic, and tissue repair potentials. These effects were further shown to be retained in the conditioned medium of hAECs, suggesting their paracrine nature. The concept of utilizing the hAEC-secretome has thus evolved as a therapeutic cell-free option. In this article, we review the different components and constituents of hAEC-secretome and their influence as demonstrated through experimental studies in the current literature. Studies examining the effects of conditioned medium, exosomes, and micro-RNA (miRNA) derived from hAECs are included in this review. The challenges facing the application of this cell-free approach will also be discussed based on the current evidence.
Collapse
Affiliation(s)
- Ibrahim Fathi
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| | - Toshio Miki
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Preparation of Drug Sustained-Release Scaffold with De-Epithelized Human Amniotic Epithelial Cells and Thiolated Chitosan Nanocarriers and Its Repair Effect on Spinal Cord Injury. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:6294148. [PMID: 35070240 PMCID: PMC8767368 DOI: 10.1155/2022/6294148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 11/22/2022]
Abstract
The disability rate of spinal cord injury (SCI) is extremely high, and stem cell inhibition is one of the most effective schemes in treating the spinal cord, but the survival rate is extremely low after stem cell transplantation, so it cannot be widely used in clinic. Studies have revealed that loading stem cells with biological scaffolds can effectively improve the survival rate and effect after stem cell transplantation. Therefore, this research was devised to analyze the repair effect of thiolated chitosan nanocarriers scaffold carrying de-epithelized human amniotic epithelial cells (HAECs) on SCI. And we used thiolated chitosan as nanocarriers, aiming to provide a reliable theoretical basis for future clinical practice. Through experiments, we concluded that the Tarlov and BBB scores of rats with SCI were raised under the intervention of thiolated chitosan carrying HAECs, while the inflammatory factors in serum, oxidative stress reaction in spinal cord tissue, apoptosis rate of nerve cells, and autophagy protein expression were all suppressed. Thus, the thiolated chitosan carrying HAECs may be applied to treat SCI by suppressing autophagy protein expression, oxidative stress response, and release of inflammatory factors in spinal cord tissue, which may be a new clinical therapy for SCI in the future. Even though we cannot understand exactly the therapeutic mechanism of thiolated chitosan carrying HAECs for SCI, the real clinical application of thiolated chitosan carrying HAECs needs to be confirmed by human experiments.
Collapse
|
8
|
Damayanti RH, Rusdiana T, Wathoni N. Mesenchymal Stem Cell Secretome for Dermatology Application: A Review. Clin Cosmet Investig Dermatol 2021; 14:1401-1412. [PMID: 34675575 PMCID: PMC8502696 DOI: 10.2147/ccid.s331044] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/21/2021] [Indexed: 12/25/2022]
Abstract
Secretome, also known as conditioned medium, is a secreted molecule from mesenchymal stem cells (MSCs) that has a variety of biological activities that can be used in various therapies, especially on the skin applications. A lack of conventional therapies makes secretome as a promising alternative therapy. The presence of growth factors, cytokines, and extracellular vesicles including microvesicles and exosomes in secretome has been widely reported, which serves in improving the proliferation and migration of cells to help in skin regeneration. Therefore, we were able to optimize the use of this secretome in a well-needed special review related to its work in addressing various skin problems. So, in this article, we discussed the benefits and biological activity of secretome on the skin application. This review was compiled based on the approval of several sites, such as Scopus, PubMed, Science Direct, and Google Scholar with the terms "MSC secretome for skin," "secretome for skin," "secretome dermatology," "secretome conditioned medium for skin," "secretome conditioned medium for skin wound," "secretome conditioned medium for aging," "secretome conditioned medium for hair growth," and "secretome conditioned medium for psoriasis." A total of 215 articles were collected for selection, of which 90 articles were used. Based on the results, it was concluded that secretome has a variety of useful activities to regenerate and repair tissue damage that have not been used on the skin, such as for wound healing, photoprotection, promotion of hair growth, psoriasis treatment, and other application as antimicrobial.
Collapse
Affiliation(s)
- Restu Harisma Damayanti
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45353, Indonesia
| | - Taofik Rusdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45353, Indonesia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45353, Indonesia
| |
Collapse
|
9
|
Liu QW, Huang QM, Wu HY, Zuo GSL, Gu HC, Deng KY, Xin HB. Characteristics and Therapeutic Potential of Human Amnion-Derived Stem Cells. Int J Mol Sci 2021; 22:ijms22020970. [PMID: 33478081 PMCID: PMC7835733 DOI: 10.3390/ijms22020970] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/06/2021] [Accepted: 01/14/2021] [Indexed: 02/08/2023] Open
Abstract
Stem cells including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and adult stem cells (ASCs) are able to repair/replace damaged or degenerative tissues and improve functional recovery in experimental model and clinical trials. However, there are still many limitations and unresolved problems regarding stem cell therapy in terms of ethical barriers, immune rejection, tumorigenicity, and cell sources. By reviewing recent literatures and our related works, human amnion-derived stem cells (hADSCs) including human amniotic mesenchymal stem cells (hAMSCs) and human amniotic epithelial stem cells (hAESCs) have shown considerable advantages over other stem cells. In this review, we first described the biological characteristics and advantages of hADSCs, especially for their high pluripotency and immunomodulatory effects. Then, we summarized the therapeutic applications and recent progresses of hADSCs in treating various diseases for preclinical research and clinical trials. In addition, the possible mechanisms and the challenges of hADSCs applications have been also discussed. Finally, we highlighted the properties of hADSCs as a promising source of stem cells for cell therapy and regenerative medicine and pointed out the perspectives for the directions of hADSCs applications clinically.
Collapse
Affiliation(s)
- Quan-Wen Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.-W.L.); (Q.-M.H.); (H.-Y.W.); (G.-S.-L.Z.); (H.-C.G.); (K.-Y.D.)
| | - Qi-Ming Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.-W.L.); (Q.-M.H.); (H.-Y.W.); (G.-S.-L.Z.); (H.-C.G.); (K.-Y.D.)
- School of Life and Science, Nanchang University, Nanchang 330031, China
| | - Han-You Wu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.-W.L.); (Q.-M.H.); (H.-Y.W.); (G.-S.-L.Z.); (H.-C.G.); (K.-Y.D.)
| | - Guo-Si-Lang Zuo
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.-W.L.); (Q.-M.H.); (H.-Y.W.); (G.-S.-L.Z.); (H.-C.G.); (K.-Y.D.)
| | - Hao-Cheng Gu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.-W.L.); (Q.-M.H.); (H.-Y.W.); (G.-S.-L.Z.); (H.-C.G.); (K.-Y.D.)
- School of Life and Science, Nanchang University, Nanchang 330031, China
| | - Ke-Yu Deng
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.-W.L.); (Q.-M.H.); (H.-Y.W.); (G.-S.-L.Z.); (H.-C.G.); (K.-Y.D.)
- School of Life and Science, Nanchang University, Nanchang 330031, China
| | - Hong-Bo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (Q.-W.L.); (Q.-M.H.); (H.-Y.W.); (G.-S.-L.Z.); (H.-C.G.); (K.-Y.D.)
- School of Life and Science, Nanchang University, Nanchang 330031, China
- Correspondence: ; Tel.: +86-791-8396-9015
| |
Collapse
|
10
|
Singh BK, Tripathi M, Sandireddy R, Tikno K, Zhou J, Yen PM. Decreased autophagy and fuel switching occur in a senescent hepatic cell model system. Aging (Albany NY) 2020; 12:13958-13978. [PMID: 32712601 PMCID: PMC7425478 DOI: 10.18632/aging.103740] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/18/2020] [Indexed: 02/05/2023]
Abstract
Although aging in the liver contributes to the development of chronic liver diseases such as NAFLD and insulin resistance, little is known about the molecular and metabolic details of aging in hepatic cells. To examine these issues, we used sequential oxidative stress with hydrogen peroxide to induce premature senescence in AML12 hepatic cells. The senescent cells exhibited molecular and metabolic signatures, increased SA-βGal and γH2A.X staining, and elevated senescence and pro-inflammatory gene expression that resembled livers from aged mice. Metabolic phenotyping showed fuel switching towards glycolysis and mitochondrial glutamine oxidation as well as impaired energy production. The senescent AML12 cells also had increased mTOR signaling and decreased autophagy which likely contributed to the fuel switching from β-oxidation that occurred in normal AML12 cells. Additionally, senescence-associated secretory phenotype (SASP) proteins from conditioned media of senescent cells sensitized normal AML12 cells to palmitate-induced toxicity, a known pathological effect of hepatic aging. In summary, we have generated senescent AML12 cells which displayed the molecular hallmarks of aging and also exhibited the aberrant metabolic phenotype, mitochondrial function, and cell signaling that occur in the aged liver.
Collapse
Affiliation(s)
- Brijesh Kumar Singh
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Madhulika Tripathi
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Reddemma Sandireddy
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Keziah Tikno
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jin Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Paul Michael Yen
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore 169857, Singapore.,Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|