1
|
Li Y, Wang W, Liu Z, Liu G, Li X. Blockade of ITGA2/3/5 Promotes Adipogenic Differentiation of Human Adipose-derived Mesenchymal Stem Cells. Cell Biochem Biophys 2024:10.1007/s12013-024-01545-w. [PMID: 39316261 DOI: 10.1007/s12013-024-01545-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 09/25/2024]
Abstract
The integrin α (ITGA) subfamily genes play a fundamental role in various cancers. However, the potential mechanism and application values of ITGA genes in adipogenic differentiation of human adipose-derived stem cells (hADSCs) remain elusive. This study confirmed that ITGA2/3/5 mRNA expressions were repressed during adipogenesis. Blockade of ITGA2/3/5 enhanced adipogenic differentiation of hADSCs. Oil red O staining found that more lipid droplets were apparent in the ITGA2/3/5 inhibition group following 14 d adipogenic induction than in the control group. In addition, inhibition of ITGA2/3/5 promoted the expression of adipogenesis-related genes (PPAR-γ, C/EBPα, FABP4). Mechanistically, ITGA2/3/5 functioned by regulating the Rac1 signaling pathway, which reasonably explains ITGA2/3/5's role in adipogenic differentiation of hADSCs. Our studies suggest that blockades of ITGA2/3/5 promote the adipogenic differentiation of hADSCs.
Collapse
Affiliation(s)
- Ying Li
- Department of Plastic and Burn Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Wendi Wang
- Department of Plastic and Burn Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Zijian Liu
- Department of Plastic and Burn Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Guangjing Liu
- Department of Plastic and Burn Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Xiaobing Li
- Department of Plastic and Burn Surgery, Tianjin First Central Hospital, Tianjin, China.
| |
Collapse
|
2
|
Foti R, Storti G, Palmesano M, Scioli MG, Fiorelli E, Terriaca S, Cervelli G, Kim BS, Orlandi A, Cervelli V. Senescence in Adipose-Derived Stem Cells: Biological Mechanisms and Therapeutic Challenges. Int J Mol Sci 2024; 25:8390. [PMID: 39125960 PMCID: PMC11312747 DOI: 10.3390/ijms25158390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Adipose tissue-derived stem cells (ADSCs) represent a subset of the mesenchymal stem cells in every adipose compartment throughout the body. ADSCs can differentiate into various cell types, including chondrocytes, osteocytes, myocytes, and adipocytes. Moreover, they exhibit a notable potential to differentiate in vitro into cells from other germinal lineages, including endothelial cells and neurons. ADSCs have a wide range of clinical applications, from breast surgery to chronic wounds. Furthermore, they are a promising cell population for future tissue-engineering uses. Accumulating evidence indicates a decreased proliferation and differentiation potential of ADSCs with an increasing age, increasing body mass index, diabetes mellitus, metabolic syndrome, or exposure to radiotherapy. Therefore, the recent literature thoroughly investigates this cell population's senescence mechanisms and how they can hinder its possible therapeutic applications. This review will discuss the biological mechanisms and the physio-pathological causes behind ADSC senescence and how they can impact cellular functionality. Moreover, we will examine the possible strategies to invert these processes, re-establishing the full regenerative potential of this progenitor population.
Collapse
Affiliation(s)
- Riccardo Foti
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.F.); (M.P.); (V.C.)
| | - Gabriele Storti
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.F.); (M.P.); (V.C.)
| | - Marco Palmesano
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.F.); (M.P.); (V.C.)
| | - Maria Giovanna Scioli
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Elena Fiorelli
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Sonia Terriaca
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Giulio Cervelli
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Bong Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8006 Zurich, Switzerland;
| | - Augusto Orlandi
- Anatomy Pathology Institute, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.G.S.); (E.F.); (S.T.); (A.O.)
| | - Valerio Cervelli
- Plastic Surgery, Department of Surgical Sciences, University of Rome “Tor Vergata”, 00133 Rome, Italy; (R.F.); (M.P.); (V.C.)
| |
Collapse
|
3
|
Li C, Gao S, Gao W. Research progress of orbital fat in histology and cytology: A review. Medicine (Baltimore) 2024; 103:e39040. [PMID: 39058805 PMCID: PMC11272235 DOI: 10.1097/md.0000000000039040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Orbital fat is an adipose tissue located behind orbital septum and originates from mesoderm and neural crest in ectoderm. It has been found that the histologic structure of orbital fat is different from subcutaneous and visceral fat. In addition, the regeneration and anti-inflammatory ability of stem cells derived from orbital fat have attracted much attention in recent years. This paper reviews the recent research progress on orbital fat, including its structure, origin, histological characteristics, and related stem cells.
Collapse
Affiliation(s)
- Changhao Li
- Department of Plastic and Cosmetic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shenzhen Gao
- Department of Plastic and Cosmetic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weicheng Gao
- Department of Plastic and Cosmetic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Sattar MA, Lingens LF, Guillaume VGJ, Goetzl R, Beier JP, Ruhl T. Association between Donor Age and Osteogenic Potential of Human Adipose Stem Cells in Bone Tissue Engineering. Curr Issues Mol Biol 2024; 46:1424-1436. [PMID: 38392210 PMCID: PMC10887920 DOI: 10.3390/cimb46020092] [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: 12/23/2023] [Revised: 01/26/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Adipose stem cells (ASCs) have multilineage differentiation capacity and hold great potential for regenerative medicine. Compared to bone marrow-derived mesenchymal stem cells (bmMSCs), ASCs are easier to isolate from abundant sources with significantly higher yields. It is generally accepted that bmMSCs show age-related changes in their proliferation and differentiation potentials, whereas this aspect is still controversial in the case of ASCs. In this review, we evaluated the existing data on the effect of donor age on the osteogenic potential of human ASCs. Overall, a poor agreement has been achieved because of inconsistent findings in the previous studies. Finally, we attempted to delineate the possible reasons behind the lack of agreements reported in the literature. ASCs represent a heterogeneous cell population, and the osteogenic potential of ASCs can be influenced by donor-related factors such as age, but also gender, lifestyle, and the underlying health and metabolic state of donors. Furthermore, future studies should consider experimental factors in in vitro conditions, including passaging, cryopreservation, culture conditions, variations in differentiation protocols, and readout methods.
Collapse
Affiliation(s)
- Md Abdus Sattar
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Lara F Lingens
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Vincent G J Guillaume
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Rebekka Goetzl
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| |
Collapse
|
5
|
Sagar S, Faizan MI, Chaudhary N, Singh V, Singh P, Gheware A, Sharma K, Azmi I, Singh VP, Kharya G, Mabalirajan U, Agrawal A, Ahmad T, Sinha Roy S. Obesity impairs cardiolipin-dependent mitophagy and therapeutic intercellular mitochondrial transfer ability of mesenchymal stem cells. Cell Death Dis 2023; 14:324. [PMID: 37173333 PMCID: PMC10181927 DOI: 10.1038/s41419-023-05810-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023]
Abstract
Mesenchymal stem cell (MSC) transplantation alleviates metabolic defects in diseased recipient cells by intercellular mitochondrial transport (IMT). However, the effect of host metabolic conditions on IMT and thereby on the therapeutic efficacy of MSCs has largely remained unexplored. Here we found impaired mitophagy, and reduced IMT in MSCs derived from high-fat diet (HFD)-induced obese mouse (MSC-Ob). MSC-Ob failed to sequester their damaged mitochondria into LC3-dependent autophagosomes due to decrease in mitochondrial cardiolipin content, which we propose as a putative mitophagy receptor for LC3 in MSCs. Functionally, MSC-Ob exhibited diminished potential to rescue mitochondrial dysfunction and cell death in stress-induced airway epithelial cells. Pharmacological modulation of MSCs enhanced cardiolipin-dependent mitophagy and restored their IMT ability to airway epithelial cells. Therapeutically, these modulated MSCs attenuated features of allergic airway inflammation (AAI) in two independent mouse models by restoring healthy IMT. However, unmodulated MSC-Ob failed to do so. Notably, in human (h)MSCs, induced metabolic stress associated impaired cardiolipin-dependent mitophagy was restored upon pharmacological modulation. In summary, we have provided the first comprehensive molecular understanding of impaired mitophagy in obese-derived MSCs and highlight the importance of pharmacological modulation of these cells for therapeutic intervention. A MSCs obtained from (HFD)-induced obese mice (MSC-Ob) show underlying mitochondrial dysfunction with a concomitant decrease in cardiolipin content. These changes prevent LC3-cardiolipin interaction, thereby reducing dysfunctional mitochondria sequestration into LC3-autophagosomes and thus impaired mitophagy. The impaired mitophagy is associated with reduced intercellular mitochondrial transport (IMT) via tunneling nanotubes (TNTs) between MSC-Ob and epithelial cells in co-culture or in vivo. B Pyrroloquinoline quinone (PQQ) modulation in MSC-Ob restores mitochondrial health, cardiolipin content, and thereby sequestration of depolarized mitochondria into the autophagosomes to alleviate impaired mitophagy. Concomitantly, MSC-Ob shows restoration of mitochondrial health upon PQQ treatment (MSC-ObPQQ). During co-culture with epithelial cells or transplantation in vivo into the mice lungs, MSC-ObPQQ restores IMT and prevents epithelial cell death. C Upon transplantation in two independent allergic airway inflammatory mouse models, MSC-Ob failed to rescue the airway inflammation, hyperactivity, metabolic changes in epithelial cells. D PQQ modulated MSCs restored these metabolic defects and restored lung physiology and airway remodeling parameters.
Collapse
Affiliation(s)
- Shakti Sagar
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Md Imam Faizan
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, 110025, India
| | - Nisha Chaudhary
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, 110025, India
| | - Vandana Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Praveen Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Atish Gheware
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Khushboo Sharma
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
| | - Iqbal Azmi
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, 110025, India
| | - Vijay Pal Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
| | - Gaurav Kharya
- Center for Bone Marrow Transplantation & Cellular Therapy Indraprastha Apollo Hospital, New Delhi, 110076, India
| | | | - Anurag Agrawal
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Tanveer Ahmad
- Multidisciplinary Center for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, 110025, India.
| | - Soumya Sinha Roy
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
6
|
Lim YJ, Jung GN, Park WT, Seo MS, Lee GW. Therapeutic potential of small extracellular vesicles derived from mesenchymal stem cells for spinal cord and nerve injury. Front Cell Dev Biol 2023; 11:1151357. [PMID: 37035240 PMCID: PMC10073723 DOI: 10.3389/fcell.2023.1151357] [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: 01/26/2023] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Neural diseases such as compressive, congenital, and traumatic injuries have diverse consequences, from benign mild sequelae to severe life-threatening conditions with associated losses of motor, sensory, and autonomic functions. Several approaches have been adopted to control neuroinflammatory cascades. Traditionally, mesenchymal stem cells (MSCs) have been regarded as therapeutic agents, as they possess growth factors and cytokines with potential anti-inflammatory and regenerative effects. However, several animal model studies have reported conflicting outcomes, and therefore, the role of MSCs as a regenerative source for the treatment of neural pathologies remains debatable. In addition, issues such as heterogeneity and ethical issues limited their use as therapeutic agents. To overcome the obstacles associated with the use of traditional agents, we explored the therapeutic potentials of extracellular vesicles (EVs), which contain nucleic acids, functional proteins, and bioactive lipids, and play crucial roles in immune response regulation, inflammation reduction, and cell-to-cell communication. EVs may surpass MSCs in size issue, immunogenicity, and response to the host environment. However, a comprehensive review is required on the therapeutic potential of EVs for the treatment of neural pathologies. In this review, we discuss the action mechanism of EVs, their potential for treating neural pathologies, and future perspectives regarding their clinical applications.
Collapse
Affiliation(s)
- Young-Ju Lim
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, Daegu, Republic of Korea
| | - Gyeong Na Jung
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, Daegu, Republic of Korea
| | - Wook-Tae Park
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, Daegu, Republic of Korea
| | - Min-Soo Seo
- Department of Veterinary Tissue Engineering, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Gun Woo Lee
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, Daegu, Republic of Korea
- *Correspondence: Gun Woo Lee,
| |
Collapse
|
7
|
Dave JR, Chandekar SS, Behera S, Desai KU, Salve PM, Sapkal NB, Mhaske ST, Dewle AM, Pokare PS, Page M, Jog A, Chivte PA, Srivastava RK, Tomar GB. Human gingival mesenchymal stem cells retain their growth and immunomodulatory characteristics independent of donor age. SCIENCE ADVANCES 2022; 8:eabm6504. [PMID: 35749495 PMCID: PMC9232118 DOI: 10.1126/sciadv.abm6504] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 05/06/2022] [Indexed: 05/28/2023]
Abstract
Aging has been reported to deteriorate the quantity and quality of mesenchymal stem cells (MSCs), which affect their therapeutic use in regenerative medicine. A dearth of age-related stem cell research further restricts their clinical applications. The present study explores the possibility of using MSCs derived from human gingival tissues (GMSCs) for studying their ex vivo growth characteristics and differentiation potential with respect to donor age. GMSCs displayed decreased in vitro adipogenesis and in vitro and in vivo osteogenesis with age, but in vitro neurogenesis remained unaffected. An increased expression of p53 and SIRT1 with donor age was correlated to their ability of eliminating tumorigenic events through apoptosis or autophagy, respectively. Irrespective of donor age, GMSCs displayed effective immunoregulation and regenerative potential in a mouse model of LPS-induced acute lung injury. Thus, we suggest the potential of GMSCs for designing cell-based immunomodulatory therapeutic approaches and their further extrapolation for acute inflammatory conditions such as acute respiratory distress syndrome and COVID-19.
Collapse
Affiliation(s)
- Jay R. Dave
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Sayali S. Chandekar
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Shubhanath Behera
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, 411007 Maharashtra, India
| | - Kaushik U. Desai
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Pradnya M. Salve
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Neha B. Sapkal
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Suhas T. Mhaske
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Ankush M. Dewle
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Parag S. Pokare
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| | - Megha Page
- Department of Dentistry, Deenanath Mangeshkar Hospital and Research Centre, Pune, 411004 Maharashtra, India
| | - Ajay Jog
- Department of Dentistry, Deenanath Mangeshkar Hospital and Research Centre, Pune, 411004 Maharashtra, India
| | - Pankaj A. Chivte
- Saraswati Danwantri Dental College and Hospital, Parbhani, 431401 Maharashtra, India
| | - Rupesh K. Srivastava
- Department of Biotechnology, All India Institute of Medical Science, New Delhi 110029, India
| | - Geetanjali B. Tomar
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, 411007 Maharashtra, India
| |
Collapse
|
8
|
Abstract
Frailty is a complex syndrome affecting a growing sector of the global population as medical developments have advanced human mortality rates across the world. Our current understanding of frailty is derived from studies conducted in the laboratory as well as the clinic, which have generated largely phenotypic information. Far fewer studies have uncovered biological underpinnings driving the onset and progression of frailty, but the stage is set to advance the field with preclinical and clinical assessment tools, multiomics approaches together with physiological and biochemical methodologies. In this article, we provide comprehensive coverage of topics regarding frailty assessment, preclinical models, interventions, and challenges as well as clinical frameworks and prevalence. We also identify central biological mechanisms that may be at play including mitochondrial dysfunction, epigenetic alterations, and oxidative stress that in turn, affect metabolism, stress responses, and endocrine and neuromuscular systems. We review the role of metabolic syndrome, insulin resistance and visceral obesity, focusing on glucose homeostasis, adenosine monophosphate-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR), and nicotinamide adenine dinucleotide (NAD+ ) as critical players influencing the age-related loss of health. We further focus on how immunometabolic dysfunction associates with oxidative stress in promoting sarcopenia, a key contributor to slowness, weakness, and fatigue. We explore the biological mechanisms involved in stem cell exhaustion that affect regeneration and may contribute to the frailty-associated decline in resilience and adaptation to stress. Together, an overview of the interplay of aging biology with genetic, lifestyle, and environmental factors that contribute to frailty, as well as potential therapeutic targets to lower risk and slow the progression of ongoing disease is covered. © 2022 American Physiological Society. Compr Physiol 12:1-46, 2022.
Collapse
Affiliation(s)
- Laís R. Perazza
- Department of Physical Therapy and Athletic Training, Boston University, Boston, Massachusetts, USA
| | - Holly M. Brown-Borg
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, USA
| | - LaDora V. Thompson
- Department of Physical Therapy and Athletic Training, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Wu SH, Yu JH, Liao YT, Liu KH, Chiang ER, Chang MC, Wang JP. Comparison of the Infant and Adult Adipose-Derived Mesenchymal Stem Cells in Proliferation, Senescence, Anti-oxidative Ability and Differentiation Potential. Tissue Eng Regen Med 2022; 19:589-601. [PMID: 35247199 PMCID: PMC9130449 DOI: 10.1007/s13770-022-00431-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/26/2021] [Accepted: 01/05/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Infant adipose-derived mesenchymal stem cells (ADSCs) collected from excised polydactyly fat tissue, which was surgical waste, could be cultured and expanded in vitro in this study. In addition, the collecting process would not cause pain in the host. In this study, the proliferation, reduction of senescence, anti-oxidative ability, and differentiation potential in the infant ADSCs were compared with those in the adult ADSCs harvested from thigh liposuction to determine the availability of infant ADSCs. METHODS Proliferation was determined by detecting the fold changes in cell numbers and doubling time periods. Senescence was analyzed by investigating the age-related gene expression levels and the replicative stress. The superoxide dismutase (SOD) gene expression, adipogenic, neurogenic, osteogenic, and tenogenic differentiation were compared by RT-qPCR. The chondrogenic differentiation efficiency was also determined using RT-qPCR and immunohistochemical staining. RESULTS The proliferation, SOD (SOD1, SOD2 and SOD3) gene expression, the stemness-related gene (c-MYC) and telomerase reverse transcriptase of the infant ADSCs at early passages were enhanced compared with those of the adults'. Cellular senescence related genes, including p16, p21 and p53, and replicative stress were reduced in the infant ADSCs. The adipogenic genes (PPARγ and LPL) and neurogenic genes (MAP2 and NEFH) of the infant ADSC differentiated cells were significantly higher than those of the adults' while the expression of the osteogenic genes (OCN and RUNX) and tenogenic genes (TNC and COL3A1) of both demonstrated opposite results. The chondrogenic markers (SOX9, COL2 and COL10) were enhanced in the infant ADSC differentiated chondrogenic pellets, and the expression levels of SODs were decreased during the differentiation process. CONCLUSION Cultured infant ADSCs demonstrate less cellular senescence and replicative stress, higher proliferation rates, better antioxidant defense activity, and higher potential of chondrogenic, adipogenic and neurogenic differentiation.
Collapse
Affiliation(s)
- Szu-Hsien Wu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, 112 Taiwan ,Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Jin-Huei Yu
- Department of Orthopedic Surgery, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, 33004 Taiwan
| | - Yu-Ting Liao
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, 112 Taiwan
| | - Kuo-Hao Liu
- Department of Orthopaedics, National Yang Ming Chiao Tung University Hospital, Yilan, 260 Taiwan
| | - En-Rung Chiang
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, 112 Taiwan
| | - Ming-Chau Chang
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, 112 Taiwan
| | - Jung-pan Wang
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, 112 Taiwan
| |
Collapse
|
10
|
Sadie-Van Gijsen H. Is Adipose Tissue the Fountain of Youth? The Impact of Adipose Stem Cell Aging on Metabolic Homeostasis, Longevity, and Cell-Based Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1286:225-250. [PMID: 33725357 DOI: 10.1007/978-3-030-55035-6_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Aging is driven by four interlinked processes: (1) low-grade sterile inflammation; (2) macromolecular and organelle dysfunction, including DNA damage, telomere erosion, and mitochondrial dysfunction; (3) stem cell dysfunction; and (4) an accumulation of senescent cells in tissues. Adipose tissue is not immune to the effects of time, and all four of these processes contribute to a decline of adipose tissue function with advanced age. This decline is associated with an increase in metabolic disorders. Conversely, optimally functioning adipose tissue generates signals that promote longevity. As tissue-resident progenitor cells that actively participate in adipose tissue homeostasis and dysregulation, adipose stem cells (ASCs) have emerged as a key feature in the relationship between age and adipose tissue function. This review will give a mechanistic overview of the myriad ways in which age affects ASC function and, conversely, how ASC function contribute to healthspan and lifespan. A central mediator in this relationship is the degree of resilience of ASCs to maintain stemness into advanced age and the consequent preservation of adipose tissue function, in particular subcutaneous fat. The last sections of this review will discuss therapeutic options that target senescent ASCs to extend healthspan and lifespan, as well as ASC-based therapies that can be used to treat age-related pathologies, and collectively, these therapeutic applications may transform the way we age.
Collapse
Affiliation(s)
- Hanél Sadie-Van Gijsen
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Campus, Parow, South Africa.
| |
Collapse
|
11
|
Horinouchi CDS, Barisón MJ, Robert AW, Kuligovski C, Aguiar AM, Dallagiovanna B. Influence of donor age on the differentiation and division capacity of human adipose-derived stem cells. World J Stem Cells 2020; 12:1640-1651. [PMID: 33505605 PMCID: PMC7789122 DOI: 10.4252/wjsc.v12.i12.1640] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/09/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human adipose-derived stromal/stem cells (hASCs) are one of the most useful types of mesenchymal stromal/stem cells, which are adult multipotent cells with great therapeutic potential for the treatment of several diseases. However, for successful clinical application, it is critical that high-quality cells can be obtained. Diverse factors seem to be able to influence cell quality and performance, especially factors related to donors’ intrinsic characteristics, such as age. Nevertheless, there is no consensus regarding this characteristic, and there is conflicting information in the literature.
AIM To investigate the growth kinetics and differentiation potential of adipose-derived stem cells isolated from the lipoaspirates of elderly and young donors.
METHODS hASCs were harvested from liposuctioned adipose tissue obtained from female donors (aged 20-70 years). Cells were distributed into two groups according to age range: old hASCs (oASCs, ≥ 55 years, n = 9) and young hASCs (yASCs, ≤ 35 years, n = 9). For each group, immunophenotypic characterization was performed by flow cytometry. Population doubling time was assessed over seven days. For adipogenic potential evaluation, lipid deposits were assessed after 7 d, 14 d and 21 d of adipogenic induction. Osteogenic potential was verified by analyzing cell mineralization after 14 d, 21 d and 28 d of osteogenic induction. mRNA expression of PPARγ2, CEBPA and Runx2 were detected by quantitative reverse transcription polymerase chain reaction.
RESULTS hASCs were successfully obtained, cultured, and grouped according to their age: yASCs (26.33 ± 4.66 years old) and oASCs (64.78 ± 4.58 years old). After maintenance of the cells in culture, there were no differences in morphology between cells from the young and old donors. Additionally, both groups showed classical immunophenotypic characteristics of mesenchymal stem/stromal cells. The average doubling time indicated that yASCs (4.09 ± 0.94 d) did not significantly differ from oASCs (4.19 ± 1.29 d). Concerning differentiation potential, after adipogenic and osteogenic induction, yASCs and oASCs were able to differentiate to greater levels than the noninduced control cells. However, no differences were found in the differentiation efficiency of yASCs and oASCs in adipogenesis or osteogenesis. Additionally, the mRNA expression of PPARγ2, CEBPA and Runx2 were similar in yASCs and oASCs.
CONCLUSION Our findings suggest that age does not seem to significantly affect the cell division or adipogenic or osteogenic differentiation ability of adipose-derived stem cells isolated from lipoaspirates.
Collapse
Affiliation(s)
- Cintia DS Horinouchi
- Stem Cells Basic Biology Laboratory, Instituto Carlos Chagas, Curitiba 81350010, Paraná, Brazil
| | - María Julia Barisón
- Stem Cells Basic Biology Laboratory, Instituto Carlos Chagas, Curitiba 81350010, Paraná, Brazil
| | - Anny W Robert
- Stem Cells Basic Biology Laboratory, Instituto Carlos Chagas, Curitiba 81350010, Paraná, Brazil
| | - Crisciele Kuligovski
- Stem Cells Basic Biology Laboratory, Instituto Carlos Chagas, Curitiba 81350010, Paraná, Brazil
| | - Alessandra M Aguiar
- Stem Cells Basic Biology Laboratory, Instituto Carlos Chagas, Curitiba 81350010, Paraná, Brazil
| | - Bruno Dallagiovanna
- Stem Cells Basic Biology Laboratory, Instituto Carlos Chagas, Curitiba 81350010, Paraná, Brazil
| |
Collapse
|
12
|
Robert AW, Marcon BH, Dallagiovanna B, Shigunov P. Adipogenesis, Osteogenesis, and Chondrogenesis of Human Mesenchymal Stem/Stromal Cells: A Comparative Transcriptome Approach. Front Cell Dev Biol 2020; 8:561. [PMID: 32733882 PMCID: PMC7362937 DOI: 10.3389/fcell.2020.00561] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/12/2020] [Indexed: 12/20/2022] Open
Abstract
Adipogenesis, osteogenesis and chondrogenesis of human mesenchymal stem/stromal cells (MSC) are complex and highly regulated processes. Over the years, several studies have focused on understanding the mechanisms involved in the MSC commitment to the osteogenic, adipogenic and/or chondrogenic phenotypes. High-throughput methodologies have been used to investigate the gene expression profile during differentiation. Association of data analysis of mRNAs, microRNAs, circular RNAs and long non-coding RNAs, obtained at different time points over these processes, are important to depict the complexity of differentiation. This review will discuss the results that were highlighted in transcriptome analyses of MSC undergoing adipogenic, osteogenic and chondrogenic differentiation. The focus is to shed light on key molecules, main signaling pathways and biological processes related to different time points of adipogenesis, osteogenesis and chondrogenesis.
Collapse
Affiliation(s)
- Anny W Robert
- Instituto Carlos Chagas - Fiocruz Paraná, Curitiba, Brazil
| | - Bruna H Marcon
- Instituto Carlos Chagas - Fiocruz Paraná, Curitiba, Brazil
| | | | | |
Collapse
|
13
|
Wang B, Liu Z, Chen VP, Wang L, Inman CL, Zhou Y, Guo C, Tchkonia T, Rowe DW, Kuchel GA, Robson P, Kirkland JL, Xu M. Transplanting cells from old but not young donors causes physical dysfunction in older recipients. Aging Cell 2020; 19:e13106. [PMID: 31971661 PMCID: PMC7059132 DOI: 10.1111/acel.13106] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/29/2019] [Accepted: 12/30/2019] [Indexed: 01/07/2023] Open
Abstract
Adipose-derived mesenchymal stem cell (ADSC)-based regenerative therapies have shown potential for use in many chronic diseases. Aging diminishes stem cell regenerative potential, yet it is unknown whether stem cells from aged donors cause adverse effects in recipients. ADSCs can be obtained using minimally invasive approaches and possess low immunogenicity. Nevertheless, we found that transplanting ADSCs from old donors, but not those from young donors, induces physical dysfunction in older recipient mice. Using single-cell transcriptomic analysis, we identified a naturally occurring senescent cell-like population in ADSCs primarily from old donors that resembles in vitro-generated senescent cells with regard to a number of key pathways. Our study reveals a previously unrecognized health concern due to ADSCs from old donors and lays the foundation for a new avenue of research to devise interventions to reduce harmful effects of ADSCs from old donors.
Collapse
Affiliation(s)
- Binsheng Wang
- UConn Center on Aging UConn Health Farmington Connecticut
- Department of Genetics and Genome Sciences UConn Health Farmington Connecticut
| | - Zukai Liu
- UConn Center on Aging UConn Health Farmington Connecticut
- Department of Genetics and Genome Sciences UConn Health Farmington Connecticut
- Biomedical Science Graduate Program UConn Health Farmington Connecticut
| | - Vicky P. Chen
- Department of Molecular Pharmacology and Experimental Therapeutics Mayo Clinic Rochester Minnesota
| | - Lichao Wang
- UConn Center on Aging UConn Health Farmington Connecticut
- Department of Genetics and Genome Sciences UConn Health Farmington Connecticut
| | | | - Yueying Zhou
- Xiangya Stomatological Hospital Central South University Changsha China
- Center for Regenerative Medicine and Skeletal Development UConn Health UConn Health Farmington Connecticut
| | - Chun Guo
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
- Center for Regenerative Medicine and Skeletal Development UConn Health UConn Health Farmington Connecticut
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
| | - David W. Rowe
- Center for Regenerative Medicine and Skeletal Development UConn Health UConn Health Farmington Connecticut
| | | | - Paul Robson
- Department of Genetics and Genome Sciences UConn Health Farmington Connecticut
- The Jackson Laboratory for Genomic Medicine Farmington Connecticut
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
| | - Ming Xu
- UConn Center on Aging UConn Health Farmington Connecticut
- Department of Genetics and Genome Sciences UConn Health Farmington Connecticut
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota
| |
Collapse
|
14
|
Abstract
Adipose stem cells (ASCs) are the basis of procedures intended for tissue regeneration. These cells are heterogeneous, owing to various factors, including the donor age, sex, body mass index, and clinical condition; the isolation procedure (liposuction or fat excision); the place from where the cells were sampled (body site and depth of each adipose depot); culture surface; type of medium (whether supplemented with fetal bovine serum or xeno-free), that affect the principal phenotypic features of ASCs. The features related to ASCs heterogeneity are relevant for the success of therapeutic procedures; these features include proliferation capacity, differentiation potential, immunophenotype, and the secretome. These are important characteristics for the success of regenerative tissue engineering, not only because of their effects upon the reconstruction and healing exerted by ASCs themselves, but also because of the paracrine signaling of ASCs and its impact on recipient tissues. Knowledge of sources of heterogeneity will be helpful in the standardization of ASCs-based procedures. New avenues of research could include evaluation of the effects of the use of more homo1geneous ASCs for specific purposes, the study of ASCs-recipient interactions in heterologous cell transplantation, and the characterization of epigenetic changes in ASCs, as well as investigations of the effect of the metabolome upon ASCs behavior in culture.
Collapse
|
15
|
Wagner DR, Karnik S, Gunderson ZJ, Nielsen JJ, Fennimore A, Promer HJ, Lowery JW, Loghmani MT, Low PS, McKinley TO, Kacena MA, Clauss M, Li J. Dysfunctional stem and progenitor cells impair fracture healing with age. World J Stem Cells 2019; 11:281-296. [PMID: 31293713 PMCID: PMC6600851 DOI: 10.4252/wjsc.v11.i6.281] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/26/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
Successful fracture healing requires the simultaneous regeneration of both the bone and vasculature; mesenchymal stem cells (MSCs) are directed to replace the bone tissue, while endothelial progenitor cells (EPCs) form the new vasculature that supplies blood to the fracture site. In the elderly, the healing process is slowed, partly due to decreased regenerative function of these stem and progenitor cells. MSCs from older individuals are impaired with regard to cell number, proliferative capacity, ability to migrate, and osteochondrogenic differentiation potential. The proliferation, migration and function of EPCs are also compromised with advanced age. Although the reasons for cellular dysfunction with age are complex and multidimensional, reduced expression of growth factors, accumulation of oxidative damage from reactive oxygen species, and altered signaling of the Sirtuin-1 pathway are contributing factors to aging at the cellular level of both MSCs and EPCs. Because of these geriatric-specific issues, effective treatment for fracture repair may require new therapeutic techniques to restore cellular function. Some suggested directions for potential treatments include cellular therapies, pharmacological agents, treatments targeting age-related molecular mechanisms, and physical therapeutics. Advanced age is the primary risk factor for a fracture, due to the low bone mass and inferior bone quality associated with aging; a better understanding of the dysfunctional behavior of the aging cell will provide a foundation for new treatments to decrease healing time and reduce the development of complications during the extended recovery from fracture healing in the elderly.
Collapse
Affiliation(s)
- Diane R Wagner
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Sonali Karnik
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Zachary J Gunderson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jeffery J Nielsen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, United States
| | - Alanna Fennimore
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Hunter J Promer
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - M Terry Loghmani
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 United States
| | - Todd O McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, United States
| | - Matthias Clauss
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jiliang Li
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| |
Collapse
|
16
|
Application of Periodontal Ligament-Derived Multipotent Mesenchymal Stromal Cell Sheets for Periodontal Regeneration. Int J Mol Sci 2019; 20:ijms20112796. [PMID: 31181666 PMCID: PMC6600219 DOI: 10.3390/ijms20112796] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023] Open
Abstract
Periodontitis is a chronic inflammatory disorder that causes destruction of the periodontal attachment apparatus including alveolar bone, the periodontal ligament, and cementum. Dental implants have been routinely installed after extraction of periodontitis-affected teeth; however, recent studies have indicated that many dental implants are affected by peri-implantitis, which progresses rapidly because of the failure of the immune system. Therefore, there is a renewed focus on periodontal regeneration aroundnatural teeth. To regenerate periodontal tissue, many researchers and clinicians have attempted to perform periodontal regenerative therapy using materials such as bioresorbable scaffolds, growth factors, and cells. The concept of guided tissue regeneration, by which endogenous periodontal ligament- and alveolar bone-derived cells are preferentially proliferated by barrier membranes, has proved effective, and various kinds of membranes are now commercially available. Clinical studies have shown the significance of barrier membranes for periodontal regeneration; however, the technique is indicated only for relatively small infrabony defects. Cytokine therapies have also been introduced to promote periodontal regeneration, but the indications are also for small size defects. To overcome this limitation, ex vivo expanded multipotent mesenchymal stromal cells (MSCs) have been studied. In particular, periodontal ligament-derived multipotent mesenchymal stromal cells are thought to be a responsible cell source, based on both translational and clinical studies. In this review, responsible cell sources for periodontal regeneration and their clinical applications are summarized. In addition, recent transplantation strategies and perspectives about the cytotherapeutic use of stem cells for periodontal regeneration are discussed.
Collapse
|
17
|
Alicka M, Major P, Wysocki M, Marycz K. Adipose-Derived Mesenchymal Stem Cells Isolated from Patients with Type 2 Diabetes Show Reduced "Stemness" through an Altered Secretome Profile, Impaired Anti-Oxidative Protection, and Mitochondrial Dynamics Deterioration. J Clin Med 2019; 8:E765. [PMID: 31151180 PMCID: PMC6617220 DOI: 10.3390/jcm8060765] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/16/2019] [Accepted: 05/21/2019] [Indexed: 02/06/2023] Open
Abstract
The widespread epidemic of obesity and type 2 diabetes (T2D), suggests that both disorders are closely linked. Several pre-clinical and clinical studies have showed that adipose-derived mesenchymal stem cells (ASC) transplantation is efficient and safe. Moreover, scientists have already highlighted the therapeutic capacity of their secretomes. In this study, we used quantitative PCR, a flow cytometry-based system, the ELISA method, spectrophotometry, and confocal and scanning electron microscopy, to compare the differences in proliferation activity, viability, morphology, mitochondrial dynamics, mRNA and miRNA expression, as well as the secretory activity of ASCs derived from two donor groups-non-diabetic and T2D patients. We demonstrated that ASCs from T2D patients showed a reduced viability and a proliferative potential. Moreover, they exhibited mitochondrial dysfunction and senescence phenotype, due to excessive oxidative stress. Significant differences were observed in the expressions of miRNA involved in cell proliferations (miR-16-5p, miR-146a-5p, and miR-145-5p), as well as miRNA and genes responsible for glucose homeostasis and insulin sensitivity (miR-24-3p, 140-3p, miR-17-5p, SIRT1, HIF-1α, LIN28, FOXO1, and TGFβ). We have observed a similar correlation of miR-16-5p, miR-146a-5p, miR-24-3p, 140-3p, miR-17-5p, and miR-145-5p expression in extracellular vesicles fraction. Furthermore, we have shown that ASCT2D exhibited a lower VEGF, adiponectin, and CXCL-12 secretion, but showed an overproduction of leptin. We have shown that type 2 diabetes attenuated crucial functions of ASC, like proliferation, viability, and secretory activity, which highly reduced their therapeutic efficiency.
Collapse
Affiliation(s)
- Michalina Alicka
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-365 Wrocław, Poland.
| | - Piotr Major
- 2'nd Department of General Surgery, Jagiellonian University Medical College, Kopernika 21, 31-501 Kraków, Poland.
| | - Michał Wysocki
- 2'nd Department of General Surgery, Jagiellonian University Medical College, Kopernika 21, 31-501 Kraków, Poland.
| | - Krzysztof Marycz
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-365 Wrocław, Poland.
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392 Gießen, Germany.
- International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114 Wisznia Mała, Poland.
| |
Collapse
|
18
|
Sun L, Chen X, Liu G, Zhang P. Subcutaneous Fat in the Upper Eyelids of Asians: Application to Blepharoplasty. Clin Anat 2019; 33:338-342. [PMID: 31056791 DOI: 10.1002/ca.23396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/11/2019] [Accepted: 04/26/2019] [Indexed: 11/10/2022]
Affiliation(s)
- Lei Sun
- Department of Plastic and Reconstructive SurgeryShanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai People's Republic of China
| | - Xi Chen
- Department of Plastic and Reconstructive SurgeryShanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai People's Republic of China
| | - Guangpeng Liu
- Department of Plastic and Reconstructive SurgeryShanghai Tenth People's Hospital, Tongji University School of Medicine Shanghai People's Republic of China
| | - Peng Zhang
- Department of OrthopaedicsShandong Provincial Hospital Affiliated to Shandong University Jinan People's Republic of China
| |
Collapse
|
19
|
Liu G, Liao C, Chen X, Xu Y, Tan J, Han F, Ye X. Identification and Characterization of Skeletal Muscle Stem Cells from Human Orbicularis Oculi Muscle. Tissue Eng Part C Methods 2019; 24:486-493. [PMID: 29993336 DOI: 10.1089/ten.tec.2018.0048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Skeletal muscle stem cell (SMSC) transplantation has shown great therapeutical potential in repairing muscle loss and dysfunction, but the muscle acquisition is usually a traumatic procedure causing pain and morbidity to the donor. In this study, we investigated the feasibility of isolating SMSCs from human orbicularis oculi muscle (OOM), which is routinely removed and discarded during ophthalmic cosmetic surgeries. OOM fragments were harvested from 18 female healthy donors undergoing upper eyelid plasties. Plastic-adherent cells were isolated from the muscles using a two-step plating method combined with collagenase digestion. A total of 15 cell cultures were successfully established from the muscle samples. These adherent cells were positive for the specific markers of SMSCs and could be directed toward the osteogenic, adipogenic, chondrogenic, and myogenic phenotypes in the presence of lineage-specific inductive media. Moreover, after cultured in the myogenic inductive medium for 3 weeks, the muscle cells were injected into the tibialis anterior muscles of nude mice and the cell fate was detected using a DiI-labeling technique. In vivo myogenesis was evidenced by the expression of DiI fluorescence after cell transplantation. The donor cells could be found in the satellite cell position and incorporated into the host myofibers. Our results demonstrated that human OOM represents a novel source of myogenic precursors with stem cell-like properties, which may provide a foundation for the SMSC-based therapeutics of skeletal muscle diseases.
Collapse
Affiliation(s)
- Guangpeng Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Caihe Liao
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Xi Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Yipin Xu
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Jian Tan
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Fang Han
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Xinhai Ye
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| |
Collapse
|
20
|
|
21
|
Multilineage-differentiating stress-enduring (Muse)-like cells exist in synovial tissue. Regen Ther 2018; 10:17-26. [PMID: 30525067 DOI: 10.1016/j.reth.2018.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/30/2018] [Accepted: 10/23/2018] [Indexed: 12/17/2022] Open
Abstract
Introduction Cartilage regeneration is a promising therapy for restoring joint function in patients with cartilage defects. The limited availability of autologous chondrocytes or chondrogenic progenitor cells is an obstacle to its clinical application. We investigated the existence and chondrogenic potential of synovial membrane-derived multilineage-differentiating stress-enduring (Muse)-like cells as an alternative cell source for cartilage regeneration. Methods Cells positive for stage-specific embryonic antigen-3 (SSEA-3), a marker of Muse cells, were isolated from the synovial membranes of 6 of 8 patients (median age, 53.5 years; range 36-72 years) by fluorescence-activated cell sorting. SSEA-3-positive cells were cultured in methylcellulose to examine their ability to form Muse clusters that are similar to the embryoid bodies formed by human embryonic stem cells. Muse clusters were expanded and chondrogenic potential of M-cluster-derived MSCs examined using a pellet culture system. Chondrogenic differentiation was evaluated by proteoglycan, safranin O, toluidine blue and type II collagen staining. To evaluate the practicality of the procedure for isolating Muse-like cells, we compared chondrogenic potential of M-cluster derived MSCs with expanded cells derived from the clusters formed by unsorted synovial cells. Results Synovial membranes contained SSEA-3-positive cells that after isolation exhibited Muse-like characteristics such as forming clusters that expressed NANOG, OCT3/4, and SOX2. In the pellet culture system, cell pellets created from the M-cluster-derived MSCs exhibited an increase in wet weight, which implied an increase in extracellular matrix production, displayed metachromasia with toluidine blue and safranin O staining and were aggrecan-positive and type II collagen-positive by immunostaining. Unsorted synovial cells also formed clusters in methylcellulose culture, and the expanded cell population derived from them exhibited chondrogenic potential. The histological and immunohistochemical appearance of chondrogenic pellet created from unsorted synovial cell-derived cells were comparable with that from M-cluster-derived MSCs. Conclusions Muse-like cells can be isolated from the human synovial membrane, even from older patients, and therefore may provide a source of multipotent cells for regenerative medicine. In addition, the cluster-forming cell population within synovial cells also has excellent chondrogenic potential. These cells may provide a more practical option for cartilage regeneration.
Collapse
Key Words
- APC, allophycocyanin
- BSA, bovine serum albumin
- Cartilage
- Chondrogenic potential
- DAB, 3,3′-diaminobenzidine
- DAPI, 4′,6-diamidino-2-phenylindole
- FBS, fetal bovine serum
- FITC, fluorescein isothiocyanate
- HRP, horseradish peroxidase
- Ig, immunoglobulin
- M-cluster, cluster cultured from synovial–Muse cells
- MC, methylcellulose
- MSC, mesenchymal stem cell
- Multilineage-differentiating stress-enduring cells
- Muse, multilineage-differentiating stress-enduring cells
- PBS, phosphate-buffered saline
- PE, phycoerythrin
- Regenerative medicine
- SSEA-3, stage-specific embryonic antigen-3
- SY-cluster, cluster cultured from unsorted synovial cells
- Stage-specific embryonic antigens-3
- αMEM, alpha-minimum essential medium
Collapse
|
22
|
Dubey NK, Mishra VK, Dubey R, Deng YH, Tsai FC, Deng WP. Revisiting the Advances in Isolation, Characterization and Secretome of Adipose-Derived Stromal/Stem Cells. Int J Mol Sci 2018; 19:ijms19082200. [PMID: 30060511 PMCID: PMC6121360 DOI: 10.3390/ijms19082200] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/08/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stromal/stem cells (ASCs) seems to be a promising regenerative therapeutic agent due to the minimally invasive approach of their harvest and multi-lineage differentiation potential. The harvested adipose tissues are further digested to extract stromal vascular fraction (SVF), which is cultured, and the anchorage-dependent cells are isolated in order to characterize their stemness, surface markers, and multi-differentiation potential. The differentiation potential of ASCs is directed through manipulating culture medium composition with an introduction of growth factors to obtain the desired cell type. ASCs have been widely studied for its regenerative therapeutic solution to neurologic, skin, wound, muscle, bone, and other disorders. These therapeutic outcomes of ASCs are achieved possibly via autocrine and paracrine effects of their secretome comprising of cytokines, extracellular proteins and RNAs. Therefore, secretome-derivatives might offer huge advantages over cells through their synthesis and storage for long-term use. When considering the therapeutic significance and future prospects of ASCs, this review summarizes the recent developments made in harvesting, isolation, and characterization. Furthermore, this article also provides a deeper insight into secretome of ASCs mediating regenerative efficacy.
Collapse
Affiliation(s)
- Navneet Kumar Dubey
- Ceramics and Biomaterials Research Group, Advanced Institute of Materials Science, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Viraj Krishna Mishra
- Applied Biotech Engineering Centre (ABEC), Department of Biotechnology, Ambala College of Engineering and Applied Research, Ambala 133101, India.
| | - Rajni Dubey
- Graduate Institute Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan.
| | - Yue-Hua Deng
- Stem Cell Research Center, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Life Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan.
| | - Feng-Chou Tsai
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Win-Ping Deng
- Stem Cell Research Center, Taipei Medical University, Taipei 11031, Taiwan.
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Basic medicine, Fu-Jen Catholic University, New Taipei City 24205, Taiwan.
| |
Collapse
|
23
|
Lai F, Kakudo N, Morimoto N, Taketani S, Hara T, Ogawa T, Kusumoto K. Platelet-rich plasma enhances the proliferation of human adipose stem cells through multiple signaling pathways. Stem Cell Res Ther 2018; 9:107. [PMID: 29661222 PMCID: PMC5902971 DOI: 10.1186/s13287-018-0851-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/18/2018] [Accepted: 03/20/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Platelet-rich plasma (PRP) is an autologous blood product that contains a high concentration of several growth factors. Platelet-derived growth factor (PDGF)-BB is a potential mitogen for human adipose-derived stem cells (hASCs). PRP stimulates proliferation of hASCs; however, the signaling pathways activated by PRP remain unclear. METHODS hASCs were cultured with or without PRP or PDGF-BB, and proliferation was assessed. hASCs were also treated with PRP or PDGF-BB with or without imatinib, which is a PDGF receptor tyrosine kinase inhibitor, or sorafenib, which is a multikinase inhibitor. Inhibition of cell proliferation was examined using anti-PDGF antibody (Abcam, Cambridge, UK), by cell counting. We assessed the effects of inhibitors of various protein kinases such as ERK1/2, JNK, p38, and Akt on the proliferation of hASCs. RESULTS The proliferation was remarkably promoted in cells treated with either 1% PRP or 10 ng/ml PDGF-BB, and both imatinib and sorafenib inhibited this proliferation. Anti-PDGF antibody (0.5 and 2 μg/ml) significantly decreased the proliferation of hASCs compared with control. PRP-mediated hASC proliferation was blocked by inhibitors of ERK1/2, Akt, and JNK, but not by an inhibitor of p38. CONCLUSIONS PRP promotes hASC proliferation, and PDGF-BB in PRP plays a major role in inducing the proliferation of hASCs. PRP promotes hASC proliferation via ERK1/2, PI3K/Akt, and JNK signaling pathways.
Collapse
Affiliation(s)
- Fangyuan Lai
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Natsuko Kakudo
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan.
| | - Naoki Morimoto
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Shigeru Taketani
- Department of Microbiology, Kansai Medical University, Osaka, 573-1010, Japan
| | - Tomoya Hara
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan.,Department of Oral Implantology, Osaka Dental University, Osaka, 573-1121, Japan
| | - Takeshi Ogawa
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Kenji Kusumoto
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| |
Collapse
|
24
|
The Effect of Age on the Regenerative Potential of Human Eyelid Adipose-Derived Stem Cells. Stem Cells Int 2018; 2018:5654917. [PMID: 29755530 PMCID: PMC5884196 DOI: 10.1155/2018/5654917] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 02/04/2018] [Accepted: 02/19/2018] [Indexed: 12/13/2022] Open
Abstract
Human eyelid adipose-derived stem cells (HEASCs) are a new source of autologous mesenchymal stem cells, which are derived from neuroectoderm and potentially applied in the tissue regeneration and cell therapies. Based on the prevalence of blepharoplasty in Asia and the availability of HEASCs, we investigated the effect of donor age on their characteristics and regenerative potential of HEASCs in vitro. The HEASCs were isolated from patients of three groups: (1) <20 years (n = 4), (2) >20 years, <45 years (n = 5), and (3) >55 years (n = 4). For each group, the proliferative capacity, colony-forming ability, surface markers, differentiation ability, wound healing function, and secreted protein were contrastively evaluated and quantified for statistical analysis. It was found that HEASCs were successfully isolated and cultured by an explant culture method. The proliferative rates, osteogenic and chondrogenic differentiation potentials, wound healing ability, and the expression of TGF-β1 and fibronectin protein of HEASCs significantly decreased as age increased. However, the expression of CD90 antigen and the adipogenic differentiation showed an age-related increase in HEASCs. As many degenerative diseases increase in prevalence with age, the age-related changes of the HEASCs proliferation potential, differentiation capacity, and wound healing ability should be taken into account whenever they are intended for use in research or cytotherapy.
Collapse
|
25
|
Characterization of Senescence of Human Adipose-Derived Stem Cells After Long-Term Expansion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1084:109-128. [PMID: 30242785 DOI: 10.1007/5584_2018_235] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Since the 1980s, adipose-derived stem cells (ASCs) have become a powerful and potential source for stem cell-based therapy, regenerative medicine, and even drug delivery in cancer treatment. The development of off-the-shelf mesenchymal stem cells (MSCs), including ASCs, has rapidly advanced in recent years with several clinical trials and approved products. In this technology, ASCs should be expanded long term in order to harvest higher cell number. In this study, senescence of ASCs after long-term expansion was evaluated. METHODS Human ASCs (hASCs) were isolated and cultured continuously at a density of 103 cells/cm2 up to passage 15. The cells were assessed for aging via changes in the following: characteristics of MSCs, mitochondrial activity, accumulation of beta-galactosidase, and expression of tumor suppressor genes. RESULTS The results showed that following in vitro expansion to the 15th passage, ASCs did not show changes in immunophenotype, except for decreased expression of CD105. However, the cells increased in size and in shape and complexity (toward the "fried egg" morphology). They also almost ceased to proliferate in passage 15. Nonetheless, they maintained in vitro differentiation potential toward osteoblasts, chondrocytes, and adipocytes. Expression of tumor suppressor genes p53 and p16 did not significantly change, while p27 was significantly downregulated. Mitochondrial activities also decreased slightly in culture from passage 5 to passage 10 and remained stable to passage 15. ASCs also showed increased accumulation of beta-galactosidase in culture, but it was negligible. CONCLUSION In conclusion, hASCs exhibited some particular characteristics of aged stem cells when the number of subculture cells increased. However, up to passage 10, ASCs also retained almost all of the characteristics of MSCs.
Collapse
|
26
|
Liu Q, Chen F, Wang L, Zhang Y. [Research progress of the donor factors and experimental factors affecting adipogenic differentiation of adipose derived stem cells]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2017; 31:1390-1395. [PMID: 29798597 PMCID: PMC8632588 DOI: 10.7507/1002-1892.201704057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 08/31/2017] [Indexed: 11/03/2022]
Abstract
Objective To summarize the donor factors and experimental factors that affect adipogenic differentiation of adipose derived stem cells, so as to provide reference for adipogenic differentiation of adipose derived stem cells. Methods The related research literature about donor factors and experimental factors affecting adipogenic differentiation of adipose derived stem cells in recent years was extensively reviewed and summarized. Results There are a lot of donor factors and experimental factors affecting adipogenic differentiation of adipose derived stem cells, but some of the factors are still controversial, such as donor age, health status, adipose tissue of different parts, and so on. These factors need to be further studied. Conclusion The donor factors and experimental factors that affect adipogenic differentiation of adipose derived stem cells should be deeply studied and the controversial issues should be clarified to lay a solid foundation for the application of adipose derived stem cells in adipose tissue engineering.
Collapse
Affiliation(s)
- Qin Liu
- Department of Medical Experiments, Wuhan General Hospital of Chinese PLA, Wuhan Hubei, 430070, P.R.China
| | - Fang Chen
- Department of Medical Experiments, Wuhan General Hospital of Chinese PLA, Wuhan Hubei, 430070, P.R.China
| | - Liping Wang
- Department of Medical Experiments, Wuhan General Hospital of Chinese PLA, Wuhan Hubei, 430070, P.R.China
| | - Yi Zhang
- Department of Medical Experiments, Wuhan General Hospital of Chinese PLA, Wuhan Hubei, 430070,
| |
Collapse
|
27
|
Barba M, Di Taranto G, Lattanzi W. Adipose-derived stem cell therapies for bone regeneration. Expert Opin Biol Ther 2017; 17:677-689. [PMID: 28374644 DOI: 10.1080/14712598.2017.1315403] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cell-based therapies exploit the heterogeneous and self-sufficient biological environment of stem cells to restore, maintain and improve tissue functions. Adipose-derived stem cells (ASCs) are, to this aim, promising cell types thanks to advantageous isolation procedures, growth kinetics, plasticity and trophic properties. Specifically, bone regeneration represents a suitable, though often challenging, target setting to test and apply ASC-based therapeutic strategies. Areas covered: ASCs are extremely plastic and secrete bioactive peptides that mediate paracrine functions, mediating their trophic actions in vivo. Numerous preclinical studies demonstrated that ASCs improve bone healing. Clinical trials are ongoing to validate the clinical feasibility of these approaches. This review is intended to define the state-of-the-art on ASCs, encompassing the biological features that make them suitable for bone regenerative strategies, and to provide an update on existing preclinical and clinical applications. Expert opinion: ASCs offer numerous advantages over other stem cells in terms of feasibility of clinical translation. Data obtained from in vivo experimentation are encouraging, and clinical trials are ongoing. More robust validations are thus expected to be achieved during the next few years, and will likely pave the way to optimized patient-tailored treatments for bone regeneration.
Collapse
Affiliation(s)
- Marta Barba
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Giuseppe Di Taranto
- b Department of Plastic, Reconstructive and Aesthetic Surgery , University of Rome "Sapienza" , Policlinico Umberto I, Rome , Italy
| | - Wanda Lattanzi
- a Institute of Anatomy and Cell Biology , Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|