1
|
Begh MZA, Zehravi M, Bhuiyan MAK, Molla MR, Raman K, Emran TB, Ullah MH, Ahmad I, Osman H, Khandaker MU. Recent advances in stem cell approaches to neurodegeneration: A comprehensive review with mechanistic insight. Pathol Res Pract 2025; 271:156013. [PMID: 40381433 DOI: 10.1016/j.prp.2025.156013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Revised: 05/10/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
The progressive nature of neurodegenerative diseases (NDs), such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis, presents substantial problems because current treatments are still obscure. Stem cell-based treatments are emerging as a viable solution to address the significant gaps in treating these severe diseases. This study provides a comprehensive analysis of the latest advancements in stem cell research, focusing on the treatment of NDs. Various types of stem cells, such as adult, induced pluripotent, and embryonic stem cells, and their potential applications in immunomodulation, neurotrophic factor release, and neuronal development are also discussed. Recent clinical studies reveal outcomes, challenges, and solutions, with advancements in disease-specific neural cell production, gene editing, and improved stem cell transplantation transport strategies. The review discussed future perspectives on developing more effective stem cell-based interventions. Biomaterials are being used for cell distribution and personalized medicine techniques to improve treatment outcomes, while exploring stem cell treatments for NDs and identifying areas for further research.
Collapse
Affiliation(s)
- Md Zamshed Alam Begh
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1216, Bangladesh.
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia.
| | | | - M Raju Molla
- Department of Pharmacy, Atish Dipankar University of Science and Technology, Dhaka 1230, Bangladesh
| | - Kannan Raman
- Department of Pharmacology, St. John's College of Pharmaceutical Sciences & Research, Kattappana, Idukki, Kerala, India
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1216, Bangladesh
| | - Md Habib Ullah
- Department of Physics, American International University-Bangladesh (AIUB), 408/1, Kuratoli, Khilkhet, Dhaka 1229, Bangladesh
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Hamid Osman
- Department of Radiological Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Mayeen Uddin Khandaker
- Applied Physics and Radiation Technologies Group, CCDCU, Faculty of Engineering and Technology, Sunway University, Bandar Sunway, 47500 Selangor, Malaysia; Department of Physics, College of Science, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| |
Collapse
|
2
|
Hyun J, Lee SY, An J, Lee YB, Bhang SH. Strengthening the cellular function of dermal fibroblasts and dermal papilla cells using nanovesicles extracted from stem cells using blue light-based photobiomodulation technology. Biomater Sci 2025; 13:1209-1221. [PMID: 39902823 DOI: 10.1039/d4bm01591f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Human dermal fibroblasts (hDFs) play a critical role in skin health by producing extracellular matrix (ECM) components essential for structural stability, while hair follicle dermal papilla cells (HFDPCs) are key to hair follicle growth and regeneration. However, factors such as UV radiation, oxidative stress, and aging impair the functions of hDFs and HFDPCs, leading to decrement in ECM production and skin maintenance and hair loss conditions like alopecia. Recent advances in nanovesicles (NVs) derived from human adipose-derived stem cells (hADSCs) have shown an innovative way in the regenerative medicine field, particularly with promise for enhancing the functionality of diverse cell types. NVs, filled with diverse bioactive molecules, are non-immunogenic, biologically stable, and capable of promoting cellular activities. To further enhance the therapeutic potential of NVs, photobiomodulation (PBM) using blue light has emerged as a promising application. Optimized blue light irradiation can induce moderate levels of reactive oxygen species production in hADSCs, activating signaling pathways that upregulate angiogenic and regenerative markers in hADSCs. In this study, blue light-irradiated NVs demonstrated superior efficacy in promoting hDF proliferation, ECM synthesis, and the functionality of HFDPCs, resulting in enhanced skin maintenance and hair follicle regeneration. This approach presents a safer and more efficient way for treating skin and hair disorders, highlighting the potential use of blue light-irradiated NVs as an innovative therapeutic strategy.
Collapse
Affiliation(s)
- Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Sang Yoon Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Jiseon An
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - You Bin Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
3
|
Wang J, Jian K, Yang Q, Gu C, Sheng J, Zhou Y, Yin H, Zhang Z, Hua K, Zhang C. Retarding human adipose-derived MSCs senescence and promoting tendon repair using cell sheet engineering with a histone methyltransferase inhibitor. Sci Rep 2025; 15:6198. [PMID: 39979391 PMCID: PMC11842574 DOI: 10.1038/s41598-025-89234-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
Mesenchymal stem cell (MSC) holds immense potential as candidates for cell therapy in the treatment of tendon injuries due to their remarkable ability for multiple cell differentiation. However, the proliferative and differentiation capacity of MSCs has been limited by cellular senescence during the process of expanding culture. Therefore, in this study, our aim was to maintain the beneficial properties of MSCs. We found that SETD7, a histone methyltransferase, was upregulated during ex vivo expansion of human adipose-derived mesenchymal stem cells (hAD-MSCs). Pharmacological inhibition of SETD7 with PFI-2 in hAD-MSCs cultures delayed their senescence, as evident by the diminished expression of senescent-associated genes and the maintenance of their proliferation and differentiation capacity. Upon transplantation, cell sheets derived from hAD-MSCs expanded with PFI-2 were better able to accelerate tendon repair. Therefore, the present findings reveal that SETD7 is an important target to improve the expansion of hAD-MSCs by delaying senescence, which is importance for the development of efficient stem cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Junjuan Wang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Ke Jian
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Qing Yang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Chunyi Gu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Jiajun Sheng
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Yan Zhou
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Hantian Yin
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Zhihan Zhang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Kouzhen Hua
- Department of Anatomy, School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311300, China.
| | - Can Zhang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, 410082, China.
| |
Collapse
|
4
|
de Freitas ALP, Han SW, Martin PKM, Ferreira LM. Effect of adipose-derived mesenchymal stem cells on the viability of the transverse rectus abdominis myocutaneous flap in rats. Clinics (Sao Paulo) 2025; 80:100590. [PMID: 39908748 PMCID: PMC11847128 DOI: 10.1016/j.clinsp.2025.100590] [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: 03/11/2024] [Revised: 09/14/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
INTRODUCTION The Transverse Rectus Abdominis Myocutaneous (TRAM) flap is used for breast reconstruction, but involves the risk of necrosis. Adipose tissue-derived mesenchymal Stem Cells (ADSCs) can be used to stimulate neovascularization and reduce the risk of TRAM flap necrosis. AIM Determine the effect of ADSCs on TRAM flap viability in rats. METHODS Twenty-four Wistar-EPM rats were distributed into three groups (n = 8). A right caudal pedicled TRAM flap was performed in all the animals and was the only procedure performed in Group TRAM. The additional procedures of intradermal injection of α-MEM culture medium and intradermal injection of α-MEM containing ADSCs labeled with a fluorescent marker were performed in Groups α-MEM and α-MEM-SC, respectively. The percentage of flap necrosis was determined, and the level of neovascularization and distribution of stem cells in the TRAM flap was assessed using immunohistochemical analysis and fluorescence microscopy, respectively. RESULTS The percentage of necrosis observed in Group α-MEM-SC was lower than that observed in Groups TRAM and α-MEM, namely 23.36 % vs. 50.42 % and 53.57 %, respectively (p < 0.05). In Zone IV of the flap, the number of vessels was greater in Group α-MEM-SC than in the other groups (p < 0.05). Multiple stem cells were observed in the four zones of the flap in Group α-MEM-SC. No stem cells were observed in Groups TRAM or α-MEM. CONCLUSION ADSCs increased TRAM flap viability and the number of vessels in Zone IV of the flap in rats.
Collapse
Affiliation(s)
| | - Sang Won Han
- MSc Interdisciplinary Center for Gene Therapy (CINTERGEN), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | | - Lydia Masako Ferreira
- Division of Plastic Surgery, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| |
Collapse
|
5
|
Yoshizumi K, Saito N, Wu Y, Shirado T, Asahi R, Mori M, Yamamoto Y, Sowa Y, Yoshimura K. Adipose-derived Stem Cells and Wound Healing Are Progressively Impaired Long-term After Radiotherapy in Mice. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2025; 13:e6419. [PMID: 39872086 PMCID: PMC11771654 DOI: 10.1097/gox.0000000000006419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/29/2024] [Indexed: 01/29/2025]
Abstract
Background The pathogenesis of deterministic radiation damage is not clearly understood, but it has been reported that fibroinflammatory pathways are up-regulated. We hypothesized that the number of adipose-derived stem/stromal cells (ASCs) decline after radiotherapies, preventing normalization of fibrosis and angiogenesis, resulting in chronic radiation damages that progress over time. Methods Dorsal skin of 8-week-old male BALB/cfC3H mice was irradiated with 10 Gy weekly for 4 weeks. At 1, 3, 6, 9, and 12 months after radiotherapy (n = 5, 5, 5, 5, and 4), tissue hemoglobin oxygen saturation, and time until epithelialization were evaluated. Skin biopsies were measured for thickness and CD34+/isolectin- stem/stromal cell count. Nonirradiated (NRT) controls were evaluated at each time point as well (n = 5 each). Results Compared with NRT controls, time until epithelialization was significantly longer at 1 month (28 ± 3, P < 0.01); not statistically different at 3 months (16 ± 2, P = 0.32); and lengthened over time at 6 months (20 ± 2, P = 0.21), 9 months (28 ± 2, P < 0.01), and 12 months (26 ± 3, P < 0.01), as did tissue oxygen saturation. The number of CD34+/isolectin- ASCs decreased over time, at 1 month (5.3 ± 1.3, P = 0.01), 3 months (6.0 ± 1.4, P = 0.03), 6 months (4.0 ± 0.8, P < 0.01), 9 months (1.7 ± 0.5, P < 0.01), and 12 months (0.3 ± 0.5, P < 0.01). The subcutaneous fatty layer was significantly thinner at 3 months (116 ± 33, P < 0.01), 6 months (147 ± 22, P = 0.02), 9 months (52 ± 12, P = 0.04), and 12 months (89 ± 19, P = 0.04), but not at 1 month (141 ± 18, P = 0.43). Conclusions After 6 months postirradiation, the number of ASCs continued to decline over time, accompanied by irreversible progression of fibrosis, atrophy, and ischemia, which resulted in impaired wound healing.
Collapse
Affiliation(s)
- Kayo Yoshizumi
- From the Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| | - Natsumi Saito
- From the Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| | - Yunyan Wu
- From the Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| | - Takako Shirado
- From the Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| | - Rintaro Asahi
- From the Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| | - Masanori Mori
- From the Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| | - Yoshihiro Yamamoto
- From the Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| | - Yoshihiro Sowa
- From the Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| | - Kotaro Yoshimura
- From the Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
6
|
Sajjad MW, Muzamil F, Sabir M, Ashfaq UA. Regenerative Medicine and Nanotechnology Approaches against Cardiovascular Diseases: Recent Advances and Future Prospective. Curr Stem Cell Res Ther 2025; 20:50-71. [PMID: 38343052 DOI: 10.2174/011574888x263530230921074827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/07/2023] [Accepted: 07/14/2023] [Indexed: 01/31/2025]
Abstract
Regenerative medicine refers to medical research focusing on repairing, replacing, or regenerating damaged or diseased tissues or organs. Cardiovascular disease (CVDs) is a significant health issue globally and is the leading cause of death in many countries. According to the Centers for Disease Control and Prevention (CDC), one person dies every 34 seconds in the United States from cardiovascular diseases, and according to a World Health Organization (WHO) report, cardiovascular diseases are the leading cause of death globally, taking an estimated 17.9 million lives each year. Many conventional treatments are available using different drugs for cardiovascular diseases, but these treatments are inadequate. Stem cells and nanotechnology are promising research areas for regenerative medicine treating CVDs. Regenerative medicines are a revolutionary strategy for advancing and successfully treating various diseases, intending to control cardiovascular disorders. This review is a comprehensive study of different treatment methods for cardiovascular diseases using different types of biomaterials as regenerative medicines, the importance of different stem cells in therapeutics, the expanded role of nanotechnology in treatment, the administration of several types of stem cells, their tracking, imaging, and the final observation of clinical trials on many different levels as well as it aims to keep readers up to pace on emerging therapeutic applications of some specific organs and disorders that may improve from regenerative medicine shortly.
Collapse
Affiliation(s)
- Muhammad Waseem Sajjad
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Fatima Muzamil
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Maida Sabir
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| |
Collapse
|
7
|
Ventura C, Bondioli E, de Vita R, Rigotti G, Morigi F, Scarpellini F, Di Fede F, Nanni-Costa A, Melandri D. Autologous Cryopreserved Adipose Tissue Using an Innovative Technique: An In Vitro Biological Characterization. Aesthet Surg J 2024; 45:NP16-NP24. [PMID: 39302643 DOI: 10.1093/asj/sjae192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/23/2024] [Accepted: 09/18/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Utilization of autologous adipose tissue transplantation in plastic and orthopedic surgery such as breast reconstruction and intra-articular injection has become an attractive surgical treatment with satisfactory clinical outcomes. Nevertheless, repeated liposuctions necessary to harvest fatty tissue, normally performed with sedation or general anesthesia, may represent a noteworthy concern. OBJECTIVES The aim of this study was to demonstrate through an in vitro characterization the validity of the surgical option of cryopreserved autologous adipose tissue harvested in a single shot for repeated graft transfer in breast reconstruction without impairment of cell viability and sterility. METHODS Adipose tissue was collected by standard liposuction from patients who needed numerous fat grafting procedures for breast reconstruction. According to an innovative and patented cryopreservation method, autologous adipose tissue was subsequently fractioned in a sterile bag system and frozen at the RER Tissue Bank of the Emilia Romagna Region. Each graft was evaluated for sterility and cell viability immediately after harvesting, and 1, 3, 6, 12, and preliminarily 18 months after cryopreservation and thawing. RESULTS In vitro results showed that after processing, middle-term and long-term cryopreservation, and subsequent thawing, autologous cryopreserved adipose tissue retained absence of bacterial contamination, high cellular viability, and unmodified histomorphological properties, thereby ensuring maintenance of the stromal vascular niche and the filling properties in different multistep surgical procedures. CONCLUSIONS In vitro study and sterility assessment showed that autologous cryopreserved adipose tissue grafting is a safe procedure, making it possible to avoid multiple liposuction surgery. No impairment of sterility, cell viability, or morphology was observed over time.
Collapse
|
8
|
Thi K, Del Toro K, Licon-Munoz Y, Sayaman RW, Hines WC. Comprehensive identification, isolation, and culture of human breast cell types. J Biol Chem 2024; 300:107637. [PMID: 39122004 PMCID: PMC11459906 DOI: 10.1016/j.jbc.2024.107637] [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: 02/08/2024] [Revised: 07/03/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
Tissues are formed and shaped by cells of many different types and are orchestrated through countless interactions. Deciphering a tissue's biological complexity thus requires studying it at cell-level resolution, where molecular and biochemical features of different cell types can be explored and thoroughly dissected. Unfortunately, the lack of comprehensive methods to identify, isolate, and culture each cell type from many tissues has impeded progress. Here, we present a method for the breadth of cell types composing the human breast. Our goal has long been to understand the essence of each of these different breast cell types, to reveal the underlying biology explaining their intrinsic features, the consequences of interactions, and their contributions to the tissue. This biological exploration has required cell purification, deep-RNA sequencing, and a thorough dissection of the genes and pathways defining each cell type. While the molecular analysis is presented in an adjoining article, we present here an exhaustive cellular dissection of the human breast and explore its cellular composition and histological organization. Moreover, we introduce a novel FACS antibody panel and rigorous gating strategy capable of isolating each of the 12 major breast cell types to purity. Finally, we describe the creation of primary cell models from nearly every breast cell type-some the first of their kind-and submit these as critical tools for studying the dynamic cellular interactions within breast tissues and tumors. Together, this body of work delivers a unique perspective of the breast, revealing insights into its cellular, molecular, and biochemical composition.
Collapse
Affiliation(s)
- Kate Thi
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Katelyn Del Toro
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Yamhilette Licon-Munoz
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Rosalyn W Sayaman
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - William C Hines
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA.
| |
Collapse
|
9
|
Pozo-Pérez L, Tornero-Esteban P, López-Bran E. Clinical and preclinical approach in AGA treatment: a review of current and new therapies in the regenerative field. Stem Cell Res Ther 2024; 15:260. [PMID: 39148125 PMCID: PMC11328498 DOI: 10.1186/s13287-024-03801-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/11/2024] [Indexed: 08/17/2024] Open
Abstract
Androgenetic alopecia (AGA) is the most prevalent type of hair loss. Its morbility is mainly psychological although an increased incidence in melanoma has also been observed in affected subjects. Current drug based therapies and physical treatments are either unsuccessful in the long term or have relevant side effects that limit their application. Therefore, a new therapeutic approach is needed to promote regenerative enhancement alternatives. These treatment options, focused on the cellular niche restoration, could be the solution to the impact of dihydrotestosterone in the hair follicle microenvironment. In this context emerging regenerative therapies such as Platelet-rich plasma or Platelet-rich fibrine as well as hair follicle stem cells and mesenchymal stem cell based therapies and their derivatives (conditioned medium CM or exoxomes) are highlighting in the evolving landscape of hair restoration. Nanotechnology is also leading the way in AGA treatment through the design of bioinks and nanobiomaterials whose structures are being configuring in a huge range of cases by means of 3D bioprinting. Due to the increasing number and the rapid creation of new advanced therapies alternatives in the AGA field, an extended review of the current state of art is needed. In addition this review provides a general insight in current and emerging AGA therapies which is intented to be a guidance for researchers highlighting the cutting edge treatments which are recently gaining ground.
Collapse
Affiliation(s)
- Lorena Pozo-Pérez
- Dermatology Department, Clínico San Carlos Hospital, Madrid, Spain.
- Institute for Health Research of Clinico San Carlos Hospital (IdISSC), Madrid, Spain.
| | - Pilar Tornero-Esteban
- Cellular GMP Manufacturing Facility, Institute for Health Research of Clinico San Carlos Hospital (IdISSC), Madrid, Spain
| | | |
Collapse
|
10
|
Gokaltun A, Asik E, Byrne D, Yarmush ML, Usta OB. Supercooled preservation of cultured primary rat hepatocyte monolayers. Front Bioeng Biotechnol 2024; 12:1429412. [PMID: 39076209 PMCID: PMC11284110 DOI: 10.3389/fbioe.2024.1429412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
Supercooled preservation (SCP) is a technology that involves cooling a substance below its freezing point without initiating ice crystal formation. It is a promising alternative to prolong the preservation time of cells, tissues, engineered tissue products, and organs compared to the current practices of hypothermic storage. Two-dimensional (2D) engineered tissues are extensively used in in vitro research for drug screening and development and investigation of disease progression. Despite their widespread application, there is a lack of research on the SCP of 2D-engineered tissues. In this study, we presented the effects of SCP at -2 and -6°C on primary rat hepatocyte (PRH) monolayers for the first time and compared cell viability and functionality with cold storage (CS, + 4°C). We preserved PRH monolayers in two different commercially available solutions: Hypothermosol-FRS (HTS-FRS) and the University of Wisconsin (UW) with and without supplements (i.e., polyethylene glycol (PEG) and 3-O-Methyl-Α-D-Glucopyranose (3-OMG)). Our findings revealed that UW with and without supplements were inadequate for the short-term preservation of PRH monolayers for both SCP and CS with high viability, functionality, and monolayer integrity. The combination of supplements (PEG and 3-OMG) in the HTS-FRS solution outperformed the other groups and yielded the highest viability and functional capacity. Notably, PRH monolayers exhibited superior viability and functionality when stored at -2°C through SCP for up to 3 days compared to CS. Overall, our results demonstrated that SCP is a feasible approach to improving the short-term preservation of PRH monolayers and enables readily available 2D-engineered tissues to advance in vitro research. Furthermore, our findings provide insights into preservation outcomes across various biological levels, from cells to tissues and organs, contributing to the advancement of bioengineering and biotechnology.
Collapse
Affiliation(s)
- Aslihan Gokaltun
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Shriners Hospitals for Children, Boston, MA, United States
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, United States
- Department of Chemical Engineering, Hacettepe University, Ankara, Türkiye
| | - Eda Asik
- Shriners Hospitals for Children, Boston, MA, United States
- Department of Bioengineering, Hacettepe University, Ankara, Türkiye
| | - Delaney Byrne
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Shriners Hospitals for Children, Boston, MA, United States
| | - Martin L. Yarmush
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Shriners Hospitals for Children, Boston, MA, United States
- Department of Biomedical Engineering, Rutgers University, Newark, NJ, United States
| | - O. Berk Usta
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Shriners Hospitals for Children, Boston, MA, United States
| |
Collapse
|
11
|
Lau CS, Park SY, Ethiraj LP, Singh P, Raj G, Quek J, Prasadh S, Choo Y, Goh BT. Role of Adipose-Derived Mesenchymal Stem Cells in Bone Regeneration. Int J Mol Sci 2024; 25:6805. [PMID: 38928517 PMCID: PMC11204188 DOI: 10.3390/ijms25126805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Bone regeneration involves multiple factors such as tissue interactions, an inflammatory response, and vessel formation. In the event of diseases, old age, lifestyle, or trauma, bone regeneration can be impaired which could result in a prolonged healing duration or requiring an external intervention for repair. Currently, bone grafts hold the golden standard for bone regeneration. However, several limitations hinder its clinical applications, e.g., donor site morbidity, an insufficient tissue volume, and uncertain post-operative outcomes. Bone tissue engineering, involving stem cells seeded onto scaffolds, has thus been a promising treatment alternative for bone regeneration. Adipose-derived mesenchymal stem cells (AD-MSCs) are known to hold therapeutic value for the treatment of various clinical conditions and have displayed feasibility and significant effectiveness due to their ease of isolation, non-invasive, abundance in quantity, and osteogenic capacity. Notably, in vitro studies showed AD-MSCs holding a high proliferation capacity, multi-differentiation potential through the release of a variety of factors, and extracellular vesicles, allowing them to repair damaged tissues. In vivo and clinical studies showed AD-MSCs favoring better vascularization and the integration of the scaffolds, while the presence of scaffolds has enhanced the osteogenesis potential of AD-MSCs, thus yielding optimal bone formation outcomes. Effective bone regeneration requires the interplay of both AD-MSCs and scaffolds (material, pore size) to improve the osteogenic and vasculogenic capacity. This review presents the advances and applications of AD-MSCs for bone regeneration and bone tissue engineering, focusing on the in vitro, in vivo, and clinical studies involving AD-MSCs for bone tissue engineering.
Collapse
Affiliation(s)
- Chau Sang Lau
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - So Yeon Park
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Lalith Prabha Ethiraj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Priti Singh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Grace Raj
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
| | - Jolene Quek
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT 06269, USA;
| | - Yen Choo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (Y.C.)
| | - Bee Tin Goh
- National Dental Centre Singapore, National Dental Research Institute Singapore, Singapore 168938, Singapore; (C.S.L.); (S.Y.P.); (L.P.E.); (G.R.)
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
12
|
Aleynik DY, Charykova IN, Rubtsova YP, Linkova DD, Farafontova EA, Egorikhina MN. Specific Features of the Functional Activity of Human Adipose Stromal Cells in the Structure of a Partial Skin-Equivalent. Int J Mol Sci 2024; 25:6290. [PMID: 38927998 PMCID: PMC11203524 DOI: 10.3390/ijms25126290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Mesenchymal adipose stromal cells (ASCs) are considered the most promising and accessible material for translational medicine. ASCs can be used independently or within the structure of scaffold-based constructs, as these not only ensure mechanical support, but can also optimize conditions for cell activity, as specific features of the scaffold structure have an impact on the vital activity of the cells. This manuscript presents a study of the secretion and accumulation that occur in a conditioned medium during the cultivation of human ASCs within the structure of such a partial skin-equivalent that is in contact with it. It is demonstrated that the ASCs retain their functional activity during cultivation both within this partial skin-equivalent structure and, separately, on plastic substrates: they proliferate and secrete various proteins that can then accumulate in the conditioned media. Our comparative study of changes in the conditioned media during cultivation of ASCs on plastic and within the partial skin-equivalent structure reveals the different dynamics of the release and accumulation of such secretory factors in the media under a variety of conditions of cell functioning. It is also demonstrated that the optimal markers for assessment of the ASCs' secretory functions in the studied partial skin-equivalent structure are the trophic factors VEGF-A, HGF, MCP, SDF-1α, IL-6 and IL-8. The results will help with the development of an algorithm for preclinical studies of this skin-equivalent in vitro and may be useful in studying various other complex constructs that include ASCs.
Collapse
Affiliation(s)
| | | | | | | | | | - Marfa N. Egorikhina
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia; (D.Y.A.); (I.N.C.); (Y.P.R.); (D.D.L.); (E.A.F.)
| |
Collapse
|
13
|
de Jong MME, Fokkema C, Papazian N, Czeti Á, Appelman MK, Vermeulen M, van Heusden T, Hoogenboezem RM, van Beek G, Tahri S, Sanders MA, van de Woestijne PC, Gay F, Moreau P, Büttner-Herold M, Bruns H, van Duin M, Broijl A, Sonneveld P, Cupedo T. An IL-1β-driven neutrophil-stromal cell axis fosters a BAFF-rich protumor microenvironment in individuals with multiple myeloma. Nat Immunol 2024; 25:820-833. [PMID: 38600356 DOI: 10.1038/s41590-024-01808-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
Human bone marrow permanently harbors high numbers of neutrophils, and a tumor-supportive bias of these cells could significantly impact bone marrow-confined malignancies. In individuals with multiple myeloma, the bone marrow is characterized by inflammatory stromal cells with the potential to influence neutrophils. We investigated myeloma-associated alterations in human marrow neutrophils and the impact of stromal inflammation on neutrophil function. Mature neutrophils in myeloma marrow are activated and tumor supportive and transcribe increased levels of IL1B and myeloma cell survival factor TNFSF13B (BAFF). Interactions with inflammatory stromal cells induce neutrophil activation, including BAFF secretion, in a STAT3-dependent manner, and once activated, neutrophils gain the ability to reciprocally induce stromal activation. After first-line myeloid-depleting antimyeloma treatment, human bone marrow retains residual stromal inflammation, and newly formed neutrophils are reactivated. Combined, we identify a neutrophil-stromal cell feed-forward loop driving tumor-supportive inflammation that persists after treatment and warrants novel strategies to target both stromal and immune microenvironments in multiple myeloma.
Collapse
Affiliation(s)
- Madelon M E de Jong
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Cathelijne Fokkema
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Natalie Papazian
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Ágnes Czeti
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Marjolein K Appelman
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Michael Vermeulen
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Teddie van Heusden
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Remco M Hoogenboezem
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Gregory van Beek
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Sabrin Tahri
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Mathijs A Sanders
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | | | - Francesca Gay
- Clinical Trial Unit, Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Philippe Moreau
- Department of Hematology, Nantes University Hospital Hotel-Dieu, Nantes, France
| | - Maike Büttner-Herold
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Heiko Bruns
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mark van Duin
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Annemiek Broijl
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Pieter Sonneveld
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands.
| | - Tom Cupedo
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands.
| |
Collapse
|
14
|
Mizui T, Inagaki A, Nakamura Y, Imura T, Uematsu SS, Miyagi S, Kamei T, Unno M, Watanabe K, Goto M. A Recombinant Peptide Device Combined with Adipose Tissue-Derived Stem Cells Enhances Subcutaneous Islet Engraftment. Cells 2024; 13:499. [PMID: 38534342 PMCID: PMC10968997 DOI: 10.3390/cells13060499] [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: 01/05/2024] [Revised: 02/28/2024] [Accepted: 03/10/2024] [Indexed: 03/28/2024] Open
Abstract
Subcutaneous space has been considered an attractive site for islet graft transplantation; however, the oxygen tension and vascularization are insufficient for islet graft survival. We investigated whether subcutaneous pre-implantation of a recombinant peptide (RCP) device with adipose tissue-derived stem cells (ADSCs) enhanced subcutaneous islet engraftment. RCP devices with/without syngeneic ADSCs were pre-implanted into the subcutaneous space of C57BL/6 mice. Syngeneic islets (300 or 120 islet equivalents (IEQs)) were transplanted into the pre-treated space after diabetes induction using streptozotocin. The cure rates of groups in which RCP devices were implanted four weeks before transplantation were significantly better than the intraportal transplantation group when 300 IEQs of islets were transplanted (p < 0.01). The blood glucose changes in the RCP+ADSCs-4w group was significantly ameliorated in comparison to the RCP-4w group when 120 IEQs of islets were transplanted (p < 0.01). Immunohistochemical analyses showed the collagen III expression in the islet capsule of the RCP+ADSCs-4w group was significantly enhanced in comparison to the RCP-4w and RCP+ADSCs-d10 groups (p < 0.01, p < 0.01). In addition, the number of von Willebrand factor-positive vessels within islets in the RCP+ADSCs-4w group was significantly higher than the RCP-4w group. These results suggest that using ADSCs in combination with an RCP device could enhance the restoration of the extracellular matrices, induce more efficient prevascularization within islets, and improve the graft function.
Collapse
Affiliation(s)
- Takahiro Mizui
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Akiko Inagaki
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.I.); (T.I.); (K.W.)
| | - Yasuhiro Nakamura
- Division of Pathology, Graduate School of Medicine, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan;
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.I.); (T.I.); (K.W.)
| | - Satomi Suzuki Uematsu
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
| | - Shigehito Miyagi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
| | - Takashi Kamei
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
| | - Kimiko Watanabe
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.I.); (T.I.); (K.W.)
| | - Masafumi Goto
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai 980-0872, Japan; (T.M.); (S.S.U.); (S.M.); (T.K.); (M.U.)
- Division of Transplantation and Regenerative Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (A.I.); (T.I.); (K.W.)
| |
Collapse
|
15
|
Kang QM, Wang J, Chen SM, Song SR, Yu SC. Glioma-associated mesenchymal stem cells. Brain 2024; 147:755-765. [PMID: 37850820 DOI: 10.1093/brain/awad360] [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: 01/03/2023] [Revised: 09/06/2023] [Accepted: 10/05/2023] [Indexed: 10/19/2023] Open
Abstract
Recent studies have revealed that glioma-associated mesenchymal stem cells play instrumental roles in tumorigenesis and tumour progression and cannot be ignored as a cellular component of the glioma microenvironment. Nevertheless, the origin of these cells and their roles are poorly understood. The only relevant studies have shown that glioma-associated mesenchymal stem cells play a large role in promoting tumour proliferation, invasion and angiogenesis. This review provides a comprehensive summary of their discovery and definition, origin, differences from other tissue-derived mesenchymal stem cells, spatial distribution, functions and prognostic and therapeutic opportunities to deepen the understanding of these cells and provide new insight into the treatment of glioma.
Collapse
Affiliation(s)
- Qing-Mei Kang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Shi-Man Chen
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Si-Rong Song
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing, 400038, China
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing, 400038, China
- Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing, 400038, China
- Jin-feng Laboratory, Chongqing, 401329, China
| |
Collapse
|
16
|
Tantuway V, Jeyaraman M, Nallakumarasamy A, Prikh MB, Sharma AK, Sharma R. Functional Outcome Analysis of Autologous Stromal Vascular Fraction (SVF) (Sahaj Therapy ®) Using Direct Sonication in Osteonecrosis of the Femoral Head (ONFH): A 6-Year Follow-Up Study. Indian J Orthop 2024; 58:68-78. [PMID: 38161400 PMCID: PMC10754810 DOI: 10.1007/s43465-023-01041-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/28/2023] [Indexed: 01/03/2024]
Abstract
Introduction We investigated the safety, efficacy, functional, and clinical outcomes of intra-osseous implantation of mechanically isolated, autologous stromal vascular fraction (SVF), an Australian patented direct ultrasonication technology (Sahaj Therapy®) in osteonecrosis of the femoral head (ONFH). Materials and Methods A total of 32 cases of ONFH were enrolled in the study after confirming with an MRI of the affected hip. All cases were treated with an intra-osseous autologous SVF implantation [4-5 cc with the cellular dosage of 8.0 × 107 cells with a viability of > 85% SVF cells] on the same surgical sitting. All the cases were followed up clinically, functionally, and radiologically at regular intervals. A comparison of mean HOOS scores at different follow-ups was done using Paired 't'-test. A P value of < 0.05 was considered significant. Results In our study, male preponderance was seen (53.1%). According to the modified Ficat and Arlet classification, the most common grade of ONFH was grade 2 [right: 25 hips and left: 25 hips]. There was a statistically significant improvement in the mean HOOS score of the right hip (n = 10) and left hip (n = 9) from preoperative time till 72 months (P < 0.05). The follow-up MRI of the affected hips shows improved osteogenesis without any further worsening of the contour of the femoral head. No adverse effects were seen in any of the study participants. Conclusion For individuals with ONFH, treated with intra-osseous autologous SVF implantation in the same surgical procedure is an innovative and promising treatment modality. Even after 6 years of follow-up, the study participants with ONFH have shown good clinical and functional outcomes with autologous SVF.
Collapse
Affiliation(s)
- Vinay Tantuway
- Department of Orthopaedics and Traumatology, Index Medical College Hospital and Research Centre, Indore, Madhya Pradesh India
- Sahaj Regenerative Cell Therapeutics, Indore, Madhya Pradesh India
| | - Madhan Jeyaraman
- Sahaj Regenerative Cell Therapeutics, Indore, Madhya Pradesh India
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh India
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX USA
| | - Arulkumar Nallakumarasamy
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu India
| | - Mittal B. Prikh
- Department of Orthopaedics, Navjivan Hospital, Ahmedabad, Gujarat India
| | - Aashish K. Sharma
- Department of Orthopaedics and Joint Replacement, CK Birla Hospitals, Jaipur, Rajasthan India
| | - Raj Sharma
- Sahaj Regenerative Cell Therapeutics, Indore, Madhya Pradesh India
| |
Collapse
|
17
|
Verling SD, Mashoudy K, Gompels M, Goldenberg G. Regenerative Medicine in Clinical and Aesthetic Dermatology. A COMPREHENSIVE GUIDE TO MALE AESTHETIC AND RECONSTRUCTIVE PLASTIC SURGERY 2024:65-79. [DOI: 10.1007/978-3-031-48503-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
18
|
Zhou Z, Zhang X, Wang S, Wang X, Mao J. A Powerful Tool in the Treatment of Myocardial Ischemia-Reperfusion Injury: Natural and Nanoscale Modified Small Extracellular Vesicles Derived from Mesenchymal Stem Cells. Int J Nanomedicine 2023; 18:8099-8112. [PMID: 38164265 PMCID: PMC10758182 DOI: 10.2147/ijn.s443716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MI/RI) constitutes a pivotal determinant impacting the long-term prognosis of individuals afflicted by ischemic cardiomyopathy subsequent to reperfusion therapy. Stem cells have garnered extensive application within the realm of MI/RI investigation, yielding tangible outcomes. Stem cell therapy encounters certain challenges in its application owing to the complexities associated with stem cell acquisition, a diminished homing rate, and a brief in vivo lifespan. Small extracellular vesicles (sEV) originating from mesenchymal stem cells (MSCs) have been demonstrated to possess the benefits of abundant availability, reduced immunogenicity, and a diminished tumorigenic incidence. They can exert their effects on damaged organs, improving injuries by transporting a lot of constituents, including proteins, RNA, lipid droplets, and more. This phenomenon has garnered substantial attention in the context of MI/RI treatment. Simultaneously, MSC-derived sEV (MSC-sEV) can exhibit enhanced therapeutic advantages through bioengineering modifications, biomaterial incorporation, and natural drug interventions. Within this discourse, we shall appraise the utilization of MSC-sEV and their derivatives in the context of MI/RI treatment, aiming to offer valuable insights for future research endeavors related to MI/RI.
Collapse
Affiliation(s)
- Zhou Zhou
- Cardiovascular Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People’s Republic of China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Xuan Zhang
- Cardiovascular Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People’s Republic of China
| | - Shuai Wang
- Cardiovascular Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People’s Republic of China
| | - Xianliang Wang
- Cardiovascular Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People’s Republic of China
| | - Jingyuan Mao
- Cardiovascular Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine/National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People’s Republic of China
| |
Collapse
|
19
|
Choi JW, Lim S, Jung SE, Jeong S, Moon H, Song BW, Kim IK, Lee S, Hwang KC, Kim SW. Enhanced Osteocyte Differentiation: Cathepsin D and L Secretion by Human Adipose-Derived Mesenchymal Stem Cells. Cells 2023; 12:2852. [PMID: 38132172 PMCID: PMC10742070 DOI: 10.3390/cells12242852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) have the potential to differentiate into bone, cartilage, fat, and neural cells and promote tissue regeneration and healing. It is known that they can have variable responses to hypoxic conditions. In the present study, we aimed to explore diverse changes in the cells and secretome of ASCs under a hypoxic environment over time and to present the possibility of ASCs as therapeutic agents from a different perspective. The expression differences of proteins between normoxic and hypoxic conditions (6, 12, or 24 h) were specifically investigated in human ASCs using 2-DE combined with MALDI-TOF MS analysis, and secreted proteins in ASC-derived conditioned media (ASC-derived CM) were examined by an adipokine array. In addition, genetic and/or proteomic interactions were assessed using a DAVID and miRNet functional annotation bioinformatics analysis. We found that 64 and 5 proteins were differentially expressed in hypoxic ASCs and in hypoxic ASC-derived CM, respectively. Moreover, 7 proteins among the 64 markedly changed spots in hypoxic ASCs were associated with bone-related diseases. We found that two proteins, cathepsin D (CTSD) and cathepsin L (CTSL), identified through an adipokine array independently exhibited significant efficacy in promoting osteocyte differentiation in bone-marrow-derived mesenchymal stem cells (BM-MSCs). This finding introduces a promising avenue for utilizing hypoxia-preconditioned ASC-derived CM as a potential therapeutic approach for bone-related diseases.
Collapse
Affiliation(s)
- Jung-Won Choi
- Medical Science Research Institute, College of Medicine, Catholic Kwandong University, Incheon Metropolitan City 22711, Republic of Korea; (J.-W.C.); (S.E.J.)
| | - Soyeon Lim
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Incheon Metropolitan City 22711, Republic of Korea; (S.L.); (B.-W.S.); (I.-K.K.); (S.L.); (K.-C.H.)
| | - Seung Eun Jung
- Medical Science Research Institute, College of Medicine, Catholic Kwandong University, Incheon Metropolitan City 22711, Republic of Korea; (J.-W.C.); (S.E.J.)
| | - Seongtae Jeong
- The Interdisciplinary Graduate Program in Integrative Biotechnology, Yonsei University, Seoul 03722, Republic of Korea;
| | - Hanbyeol Moon
- Department of Integrated Omics for Biomedical Sciences, Graduate School, Yonsei University, Seoul 03722, Republic of Korea;
| | - Byeong-Wook Song
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Incheon Metropolitan City 22711, Republic of Korea; (S.L.); (B.-W.S.); (I.-K.K.); (S.L.); (K.-C.H.)
| | - Il-Kwon Kim
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Incheon Metropolitan City 22711, Republic of Korea; (S.L.); (B.-W.S.); (I.-K.K.); (S.L.); (K.-C.H.)
| | - Seahyoung Lee
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Incheon Metropolitan City 22711, Republic of Korea; (S.L.); (B.-W.S.); (I.-K.K.); (S.L.); (K.-C.H.)
| | - Ki-Chul Hwang
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Incheon Metropolitan City 22711, Republic of Korea; (S.L.); (B.-W.S.); (I.-K.K.); (S.L.); (K.-C.H.)
| | - Sang Woo Kim
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Incheon Metropolitan City 22711, Republic of Korea; (S.L.); (B.-W.S.); (I.-K.K.); (S.L.); (K.-C.H.)
| |
Collapse
|
20
|
Guillaume VGJ, Lanckohr LS, Lippold EF, Beier JP, Ruhl T. Effects of epinephrine, lidocaine, and prilocaine on viability and differentiation capacity of human adipose stem cells. J Plast Reconstr Aesthet Surg 2023; 87:408-415. [PMID: 37939646 DOI: 10.1016/j.bjps.2023.10.104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 11/10/2023]
Abstract
INTRODUCTION Local anesthetics (LAs) are routinely administered in plastic and reconstructive surgery, e.g., as tumescent anesthesia adjunct in liposuction. Historically, these substances were assumed to act cytotoxically. Thus, the application of LA was avoided when handling adipose stem cells (ASCs). We recently determined that most LAs are not cytotoxic when ASCs are exposed to concentrations used for tumescent liposuction. However, there is limited information when combining LA with epinephrine and about the effects of prilocaine on ASCs. METHODS We analyzed the effects of prilocaine or lidocaine in co-exposure with epinephrine on the viability of primary human ASCs, i.e., proliferation, metabolic activity, and cytotoxicity, using crystal violet-staining, PrestoBlue®-, and WST-1 assay. We quantified the impact of short-term incubation of lidocaine and epinephrine on the differentiation of ASCs into the adipogenic, chondrogenic, and osteogenic lineage. RESULTS After 2 h, prilocaine (10 mM) significantly reduced metabolic activity and cell numbers, whereas lidocaine only inhibited metabolic activity. After 6 h, prilocaine (10 mM) and lidocaine significantly decreased metabolic activity as well as cell numbers. The application of high concentrations of epinephrine did not affect cell numbers but diminished metabolic activity. Combining lidocaine with epinephrine had no additional cytotoxic effect. Differentiation into the chondrogenic lineage was significantly inhibited by epinephrine. CONCLUSIONS Deducing from our data, neither lidocaine combined with epinephrine nor prilocaine has a cytotoxic impact on ASCs in vitro at concentrations equivalent to those in tumescent anesthesia and has no long-lasting effect on the differentiation capacity of ASCs into the osteogenic and adipogenic lineage.
Collapse
Affiliation(s)
- Vincent G J Guillaume
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, NRW, Germany.
| | - Laura S Lanckohr
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, NRW, Germany
| | - Ella F Lippold
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, NRW, Germany
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, NRW, Germany
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, NRW, Germany
| |
Collapse
|
21
|
Guimarães LB, Machado DPD, Carvalho Versiani Caldeira BF, Vieira LTM, Santos GA, Araújo FR, Machado LT, Gomes DA, Ocarino NDM, Serakides R, Reis AMS. Kisspeptin (Kp-10) inhibits in vitro osteogenic differentiation of multipotent mesenchymal stromal cells extracted from the bone marrow of adult rats. Acta Histochem 2023; 125:152112. [PMID: 37948785 DOI: 10.1016/j.acthis.2023.152112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
Kisspeptin (Kp-10) is a neuropeptide that binds to GPR54 receptors, exerting several functions mainly in the nervous and reproductive systems of the body. However, its effects and mechanisms of action on the skeletal system remain poorly understood. This study evaluated the effects of different concentrations of Kp-10 on in vitro osteogenic differentiation of multipotent mesenchymal stromal cells (MSCs) extracted from the bone marrow (BM) of adult Wistar rats. Two-month-old female rats were euthanized to extract BM from long bones to obtain MSCs. Four experimental groups were established in vitro: a control and Kp-10 at concentrations of 0.01, 0.05 and, 0.1 µg/mL. After induction of osteogenic differentiation, cell viability was evaluated using the 3-(4,5-dimethylthiazol-2-yl)- 2,5-diphenyl tetrazolium bromide (MTT) assay, alkaline phosphatase activity, collagen synthesis, percentage of area covered by MSCs/field and mineralized nodules/field, and immunocytochemistry of the GPR54 receptor tests. Furthermore, evaluation of gene transcripts for type I collagen, Runx-2, Bmp-2, bone sialoprotein, osteocalcin and osteopontin was performed using real-time RT-qPCR. It was observed that MSCs expressed GPR54 receptor to which Kp-10 binds during osteogenic differentiation, promoting a negative effect on osteogenic differentiation. This effect was observed at all the Kp-10 concentrations in a concentration-dependent manner, characterized by a decrease in the activity of alkaline phosphatase, collagen synthesis, mineralized nodules, and decreased expression of gene transcripts for type I collagen, osteocalcin, osteopontin, and Runx-2. Thus, Kp-10 inhibits in vitro osteogenic differentiation of MSCs extracted from the BM of adult Wistar rats.
Collapse
Affiliation(s)
- Laís Bitencourt Guimarães
- Departamento de Patologia Geral do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, CEP: 30.161-970 Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Portela Dias Machado
- Departamento de Farmacologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, CEP: 30.161-970 Belo Horizonte, Minas Gerais, Brazil
| | - Beatriz Ferreira Carvalho Versiani Caldeira
- Departamento de Patologia Geral do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, CEP: 30.161-970 Belo Horizonte, Minas Gerais, Brazil
| | - Larissa Tiemi Matuzake Vieira
- Departamento de Patologia Geral do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, CEP: 30.161-970 Belo Horizonte, Minas Gerais, Brazil
| | - Gabriela Alves Santos
- Departamento de Patologia Geral do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, CEP: 30.161-970 Belo Horizonte, Minas Gerais, Brazil
| | - Fabiana Rocha Araújo
- Núcleo de Células Tronco e Terapia Celular Animal (NCT-TCA) do Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, campus Pampulha da UFMG, Av. Antônio Carlos 6627, Caixa Postal 567, CEP 30.123-970 Belo Horizonte, Minas Gerais, Brazil
| | - Leonardo Teotônio Machado
- Departamento de Patologia Geral do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, CEP: 30.161-970 Belo Horizonte, Minas Gerais, Brazil
| | - Dawidson Assis Gomes
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, CEP: 30.161-970 Belo Horizonte, Minas Gerais, Brazil
| | - Natália de Melo Ocarino
- Núcleo de Células Tronco e Terapia Celular Animal (NCT-TCA) do Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, campus Pampulha da UFMG, Av. Antônio Carlos 6627, Caixa Postal 567, CEP 30.123-970 Belo Horizonte, Minas Gerais, Brazil
| | - Rogéria Serakides
- Núcleo de Células Tronco e Terapia Celular Animal (NCT-TCA) do Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, campus Pampulha da UFMG, Av. Antônio Carlos 6627, Caixa Postal 567, CEP 30.123-970 Belo Horizonte, Minas Gerais, Brazil
| | - Amanda Maria Sena Reis
- Departamento de Patologia Geral do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627, CEP: 30.161-970 Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
22
|
Caliani Carrera AL, Minto BW, Malard P, Brunel HDSS. The Role of Mesenchymal Stem Cell Secretome (Extracellular Microvesicles and Exosomes) in Animals' Musculoskeletal and Neurologic-Related Disorders. Vet Med Int 2023; 2023:8819506. [PMID: 38023428 PMCID: PMC10645499 DOI: 10.1155/2023/8819506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
The advances in regenerative medicine are very important for the development of medicine and the discovery of stem cells has shown a greater capacity to raise the level of therapeutic quality while their use becomes more accessible, especially in their mesenchymal form. In veterinary medicine, it is not different. The use of those cells, as well as recent advances related to the use of their extracellular vesicles, demonstrates a great opportunity to enhance therapeutic methods and ensure more life quality for patients, which can be in clinical or surgical treatments. Knowing the advances in these modalities and the growing clinical and surgery research and demands for innovations in orthopedic and neurology medicines, this paper aimed to review the literature about the methodologies of use and applications such as the pathways of action and the advances that were postulated for microvesicles and exosomes derived from mesenchymal stem cells in veterinary medicine, especially for musculoskeletal disorders and related injuries.
Collapse
Affiliation(s)
- Alefe Luiz Caliani Carrera
- Department of Clinical and Veterinary Surgery, São Paulo State University (UNESP), Av Paulo Donato Castelane s/n, Jaboticabal, São Paulo, Brazil
| | - Bruno Watanabe Minto
- Department of Clinical and Veterinary Surgery, São Paulo State University (UNESP), Av Paulo Donato Castelane s/n, Jaboticabal, São Paulo, Brazil
| | - Patrícia Malard
- Catholic University of Brasilia, Brasília, Federal District, Brazil
| | | |
Collapse
|
23
|
Cero C, Shu W, Reese AL, Douglas D, Maddox M, Singh AP, Ali SL, Zhu AR, Katz JM, Pierce AE, Long KT, Nilubol N, Cypess RH, Jacobs JL, Tian F, Cypess AM. Standardized In Vitro Models of Human Adipose Tissue Reveal Metabolic Flexibility in Brown Adipocyte Thermogenesis. Endocrinology 2023; 164:bqad161. [PMID: 37944134 PMCID: PMC11032247 DOI: 10.1210/endocr/bqad161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/10/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Functional human brown and white adipose tissue (BAT and WAT) are vital for thermoregulation and nutritional homeostasis, while obesity and other stressors lead, respectively, to cold intolerance and metabolic disease. Understanding BAT and WAT physiology and dysfunction necessitates clinical trials complemented by mechanistic experiments at the cellular level. These require standardized in vitro models, currently lacking, that establish references for gene expression and function. We generated and characterized a pair of immortalized, clonal human brown (hBA) and white (hWA) preadipocytes derived from the perirenal and subcutaneous depots, respectively, of a 40-year-old male individual. Cells were immortalized with hTERT and confirmed to be of a mesenchymal, nonhematopoietic lineage based on fluorescence-activated cell sorting and DNA barcoding. Functional assessments showed that the hWA and hBA phenocopied primary adipocytes in terms of adrenergic signaling, lipolysis, and thermogenesis. Compared to hWA, hBA were metabolically distinct, with higher rates of glucose uptake and lactate metabolism, and greater basal, maximal, and nonmitochondrial respiration, providing a mechanistic explanation for the association between obesity and BAT dysfunction. The hBA also responded to the stress of maximal respiration by using both endogenous and exogenous fatty acids. In contrast to certain mouse models, hBA adrenergic thermogenesis was mediated by several mechanisms, not principally via uncoupling protein 1 (UCP1). Transcriptomics via RNA-seq were consistent with the functional studies and established a molecular signature for each cell type before and after differentiation. These standardized cells are anticipated to become a common resource for future physiological, pharmacological, and genetic studies of human adipocytes.
Collapse
Affiliation(s)
- Cheryl Cero
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weiguo Shu
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Amy L Reese
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Diana Douglas
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Michael Maddox
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
- Current Affiliation: Vita Therapeutics, 801 W. Baltimore Street, Suite 301, Baltimore, MD 21201, USA
| | - Ajeet P Singh
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Sahara L Ali
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander R Zhu
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jacqueline M Katz
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anne E Pierce
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kelly T Long
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Naris Nilubol
- Surgical Oncology Program, Center for Cancer Research, NCI, NIH, 10 Center Drive, Room 4-5952, Bethesda, MD 20892, USA
| | - Raymond H Cypess
- American Type Culture Collection, 10801 University Blvd, Manassas, VA 20110, USA
| | - Jonathan L Jacobs
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Fang Tian
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
24
|
Vinikoor T, Dzidotor GK, Le TT, Liu Y, Kan HM, Barui S, Chorsi MT, Curry EJ, Reinhardt E, Wang H, Singh P, Merriman MA, D'Orio E, Park J, Xiao S, Chapman JH, Lin F, Truong CS, Prasadh S, Chuba L, Killoh S, Lee SW, Wu Q, Chidambaram RM, Lo KWH, Laurencin CT, Nguyen TD. Injectable and biodegradable piezoelectric hydrogel for osteoarthritis treatment. Nat Commun 2023; 14:6257. [PMID: 37802985 PMCID: PMC10558537 DOI: 10.1038/s41467-023-41594-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/11/2023] [Indexed: 10/08/2023] Open
Abstract
Osteoarthritis affects millions of people worldwide but current treatments using analgesics or anti-inflammatory drugs only alleviate symptoms of this disease. Here, we present an injectable, biodegradable piezoelectric hydrogel, made of short electrospun poly-L-lactic acid nanofibers embedded inside a collagen matrix, which can be injected into the joints and self-produce localized electrical cues under ultrasound activation to drive cartilage healing. In vitro, data shows that the piezoelectric hydrogel with ultrasound can enhance cell migration and induce stem cells to secrete TGF-β1, which promotes chondrogenesis. In vivo, the rabbits with osteochondral critical-size defects receiving the ultrasound-activated piezoelectric hydrogel show increased subchondral bone formation, improved hyaline-cartilage structure, and good mechanical properties, close to healthy native cartilage. This piezoelectric hydrogel is not only useful for cartilage healing but also potentially applicable to other tissue regeneration, offering a significant impact on the field of regenerative tissue engineering.
Collapse
Affiliation(s)
- Tra Vinikoor
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Godwin K Dzidotor
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Thinh T Le
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Yang Liu
- Center of Digital Dentistry/Department of Prosthodontics/Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Research Center of Engineering and Technology for Computerized Dentistry & NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Ho-Man Kan
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Srimanta Barui
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Meysam T Chorsi
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Eli J Curry
- Eli Lilly and Company, 450 Kendall Street, Cambridge, MA, 02142, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Emily Reinhardt
- Department of Pathobiology and Veterinary Science, University of Connecticut, 61 North Eagleville Road, Unit 3089, Storrs, CT, 06269, USA
| | - Hanzhang Wang
- Pathology and Laboratory Medicine, University of Connecticut Health Center, 63 Farmington Avenue, Farmington, CT, 06030, USA
| | - Parbeen Singh
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Marc A Merriman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Ethan D'Orio
- Department of Advanced Manufacturing for Energy Systems Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Jinyoung Park
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Shuyang Xiao
- Department of Materials Science and Engineering & Institute of Materials Science, University of Connecticut, 25 King Hill Road, Unit 3136, Storrs, CT, 06269-3136, USA
| | - James H Chapman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Feng Lin
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Cao-Sang Truong
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Lisa Chuba
- Center for Comparative Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Shaelyn Killoh
- Center for Comparative Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Seok-Woo Lee
- Department of Materials Science and Engineering & Institute of Materials Science, University of Connecticut, 25 King Hill Road, Unit 3136, Storrs, CT, 06269-3136, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Qian Wu
- Pathology and Laboratory Medicine, University of Connecticut Health Center, 63 Farmington Avenue, Farmington, CT, 06030, USA
| | - Ramaswamy M Chidambaram
- Center for Comparative Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Kevin W H Lo
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Cato T Laurencin
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science and Engineering & Institute of Materials Science, University of Connecticut, 25 King Hill Road, Unit 3136, Storrs, CT, 06269-3136, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery University of Connecticut Health, Farmington, CT, 06030, USA
| | - Thanh D Nguyen
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|
25
|
Cremona M, Rusconi G, Ferrario A, Mariotta L, Gola M, Soldati G. Processing Adipose Tissue Samples in a GMP Environment Standardizes the Use of SVF in Cell Therapy Treatments: Data on 302 Patients. Biomedicines 2023; 11:2533. [PMID: 37760974 PMCID: PMC10525825 DOI: 10.3390/biomedicines11092533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/09/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Stromal vascular fraction (SVF) cells, together with adipose-derived mesenchymal stem cells, are becoming the tool of choice for many clinical applications. Currently, nearly 200 clinical trials are running worldwide to prove the efficacy of this cell type in treating many diseases and pathological conditions. To reach the goals of cell therapies and produce ATMPs as drugs for regenerative medicine, it is necessary to properly standardize GMP processes and, thus, collection methods, transportation strategies, extraction protocols, and characterization procedures, without forgetting that all the tissues of the human body are characterized by a wide inter-individual variability which is genetically determined and acquired during life. Here, we compare 302 samples processed under GMP rules to exclude the influence of the operator and of the anatomical site of collection. The influence of variability in the ages and genders of patients, along with laboratory parameters such as total cell number, cell viability, stem cell number, and other stromal vascular fraction cell subpopulations, has been compared. The results show that when the laboratory protocol is standardized, the variability of quantifiable cell parameters is widely statistically non-significant, meaning that we can take a further step toward standardized advanced cell therapy products.
Collapse
Affiliation(s)
- Martina Cremona
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| | - Giulio Rusconi
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| | | | - Luca Mariotta
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
- Swiss Stem Cells Biotech AG, 8008 Zürich, Switzerland
| | - Mauro Gola
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| | - Gianni Soldati
- Swiss Stem Cell Foundation, 6900 Lugano, Switzerland; (M.C.)
| |
Collapse
|
26
|
Sonmez Kaplan S, Sazak Ovecoglu H, Genc D, Akkoc T. TNF-α, IL-1B and IL-6 affect the differentiation ability of dental pulp stem cells. BMC Oral Health 2023; 23:555. [PMID: 37568110 PMCID: PMC10422753 DOI: 10.1186/s12903-023-03288-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND This in vitro study examined the effect of the inflammatory cytokines (tumour necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6) on osteogenic, chondrogenic, and adipogenic differentiation of dental pulp stem cells (DPSCs) which have significant relevance in future regenerative therapies. METHODS DPSCs were isolated from the impacted third molar dental pulp and determined with flow cytometry analysis. DPSCs were divided into into 5 main groups with 3 subdivisions for each group making a total of 15 groups. Experimental groups were stimulated with TNF-α, IL-1β, IL-6, and a combination of all three to undergo osteogenic, chondrogenic, and adipogenic differentiation protocols. Next, the differentiation of each group was examined with different staining procedures under a light microscope. Histological analysis of osteogenic, chondrogenic, and adipogenic differentiated pellets was assessed using a modified Bern score. Statistical significance determined using one-way analysis of variance, and correlations were assessed using Pearson's test (two-tailed). RESULTS Stimulation with inflammatory cytokines significantly inhibited the osteogenic, chondrogenic and adipogenic differentiation of DPSCs in terms of matrix and cell formation resulting in weak staining than the unstimulated groups with inflammatory cytokines. On contrary, the unstimulated groups of MSCs have shown to be highly proliferative ability in terms of osteogenic, chondrogenic, and adipogenic differentiation. CONCLUSIONS DPSCs have high osteogenic, chondrogenic, and adipogenic differentiation capabilities. Pretreatment with inflammatory cytokines decreases the differentiation ability in vitro, thus inhibiting tissue formation.
Collapse
Affiliation(s)
- Sema Sonmez Kaplan
- Department of Endodontics, Faculty of Dentistry, Biruni University, 10. Yıl Caddesi Protokol Yolu No: 45, 34010, Topkapı, Istanbul, Turkey.
| | - Hesna Sazak Ovecoglu
- Faculty of Dentistry Department of Endodontics, Marmara University, Istanbul, Turkey
| | - Deniz Genc
- Department of Pediatric Health & Diseases Faculty of Health Sciences, Muğla Sıtkı Koçman University, Mugla, Turkey
- Research Laboratories Center, Immunology and Stem Cell Laboratory, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Tunc Akkoc
- Immunology Department, Marmara University Medical Faculty, Istanbul, Turkey
| |
Collapse
|
27
|
Hikichi M, Shimizu T, Sato K. Development of supercooling preservation method of adherent cultured human cells. J Biochem 2023; 174:273-278. [PMID: 37141918 DOI: 10.1093/jb/mvad040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/17/2023] [Accepted: 05/01/2023] [Indexed: 05/06/2023] Open
Abstract
Cryopreservation of mammalian cells is an important technology; however, freezing damage due to osmotic pressure differences and ice crystal formation is inevitable. In addition, cryopreserved cells cannot be used immediately after thawing in many cases. Therefore, in this study, we developed a method for supercooling and preserving adherent cells using a precision temperature-controlled CO2 incubator. The effects of the cooling rate from 37 to -4°C, the warming rate from -4 to 37°C and a preservation solution on cell viability after storage were examined. Human hepatocarcinoma-derived cell line HepG2 cells, preserved with HypoThermosol FRS at -4°C with a cooling rate of -0.028°C/min (24 h from 37°C to -4°C) and warming to 37°C at a rate of +1.0°C/min (40 min from -4 to 37°C), displayed high cell viability after 14 days of preservation. The superiority of supercooling preservation at -4°C was demonstrated by comparing the obtained results with that of refrigerated preservation at +4°C. Cells preserved for 14 days under optimal conditions showed no cell shape abnormalities and may be used for experiments immediately after thawing. The optimized supercooling preservation method determined in this study is suitable for the temporary preservation of adherent cultured cells.
Collapse
Affiliation(s)
- Maaya Hikichi
- School of Science and Technology, Gunma University, Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| | - Takuya Shimizu
- R&D Division, Sanden Retail Systems Corporation, ARCA West 8F, 1-2-4, Kinshi, Sumida-ku, Tokyo 130-8563, Japan
| | - Kiichi Sato
- School of Science and Technology, Gunma University, Tenjin-cho, Kiryu, Gunma 376-8515, Japan
| |
Collapse
|
28
|
Tel A, Miotti G, Ius T, Marco LD, Robiony M, Parodi PC, Panciani PP, Zeppieri M. Stem Cells in Facial Regenerative Surgery: Current Clinical Applications. A Multidisciplinary, Systematic Review. FRONT BIOSCI-LANDMRK 2023; 28:123. [PMID: 37395035 DOI: 10.31083/j.fbl2806123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/11/2023] [Accepted: 05/29/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Facial reconstruction represents one of the main challenges for surgeons. Stem cells (SC) represent the most studied solution for tissue regeneration. This approach appears particularly promising in combination with bioengineered scaffolds and 3D bioprinting. The purpose of this systematic review is to define the main domains of current application of SC therapy within contemporary clinical workflows, evaluate indications and limitations, report current knowledge in this innovative field of research, and define the landscape of evidence for such approaches. METHODS A systematic review was performed on the pertinent literature regarding stem cell-based cell therapies currently available in the reconstruction of the face. The review used the main databases for scientific literature based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. RESULTS A total of 15 papers were selected after an independent search was performed. The two major domains for current application of stem cells in clinical practice were bone and skin categories. CONCLUSIONS Cell therapy in the field of facial reconstruction represents a promising approach. The evidence regarding the current clinical use, however, seems to show this option to be limited. Bioengineering advances and the parallel development of 3D bioprinting technology can potentially enhance the role of stem cells in the future.
Collapse
Affiliation(s)
- Alessandro Tel
- Clinic of Maxillofacial Surgery, Head-Neck and NeuroScience Department University Hospital of Udine, 33100 Udine, Italy
| | - Giovanni Miotti
- Department of Plastic Surgery, University Hospital of Udine, 33100 Udine, Italy
| | - Tamara Ius
- Neurosurgery Unit, Head-Neck and NeuroScience Department University Hospital of Udine, 33100 Udine, Italy
| | - Luca de Marco
- Department of Plastic Surgery, University Hospital of Udine, 33100 Udine, Italy
| | - Massimo Robiony
- Clinic of Maxillofacial Surgery, Head-Neck and NeuroScience Department University Hospital of Udine, 33100 Udine, Italy
| | - Pier Camillo Parodi
- Department of Plastic Surgery, University Hospital of Udine, 33100 Udine, Italy
| | - Pier Paolo Panciani
- Division of Neurosurgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25123 Brescia, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| |
Collapse
|
29
|
Jing Y, Zhou J, Guo F, Yu L, Ren X, Yin X. Betaine regulates adipogenic and osteogenic differentiation of hAD-MSCs. Mol Biol Rep 2023; 50:5081-5089. [PMID: 37101008 DOI: 10.1007/s11033-023-08404-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/23/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND With an ageing population, the incidence of bone loss and obesity are increasing. Numerous studies emphasized the multidirectional differentiation ability of mesenchymal stem cells (MSCs), and reported betaine modulated the osteogenic differentiation and adipogenic differentiation of MSCs in vitro. We wondered how betaine affected the differentiation of hAD-MSCs and hUC-MSCs. METHODS AND RESULTS ALP staining and alizarin red S (ARS) staining were proved 10 mM betaine significantly increased the number of ALP-positive cells and plaque calcified extracellular matrices, accompanying by the up-regulation of OPN, Runx-2 and OCN. Oil red O staining demonstrated the number and size of lipid droplets were reduced, the expression of adipogenic master genes such as PPARγ, CEBPα and FASN were down-regulated simultaneously. For further investigating the mechanism of betaine on hAD-MSCs, RNA-seq was performed in none-differentiation medium. The Gene Ontology (GO) analysis showed fat cell differentiation and bone mineralization function terms were enriched, and KEGG showed PI3K-Akt signaling pathway, cytokine-cytokine receptor interaction and ECM-receptor interaction pathways were enriched in betaine treated hAD-MSCs, demonstrated betaine had a positive inducing effect on osteogenic of hAD-MSCs in the non-differentiation medium in vitro, which is opposite to the effect on adipogenic differentiation. CONCLUSIONS Our study demonstrated that betaine promoted osteogenic and compromised adipogenic differentiation of hUC-MSCs and hAD-MSCs upon low concentration administration. PI3K-Akt signaling pathway, cytokine-cytokine receptor interaction and ECM-receptor interaction were significantly enriched under betaine-treated. We showed hAD-MSCs were more sensitive to betaine stimulation and have a better differentiation ability than hUC-MSCs. Our results contributed to the exploration of betaine as an aiding agent for MSCs therapy.
Collapse
Affiliation(s)
- Yue Jing
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, Liaoning Province, China
| | - Jian Zhou
- College of Pharmaceutical Sciences, Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Fenghua Guo
- Jiangsu Pulu Rui Medical Technology Co., Ltd, Xuzhou, Jiangsu Province, China
| | - Lin Yu
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, Liaoning Province, China
| | - Xiaomeng Ren
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, Liaoning Province, China
| | - Xiushan Yin
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, Liaoning Province, China.
| |
Collapse
|
30
|
Yaylacı S, Kaçaroğlu D, Hürkal Ö, Ulaşlı AM. An enzyme-free technique enables the isolation of a large number of adipose-derived stem cells at the bedside. Sci Rep 2023; 13:8005. [PMID: 37198228 DOI: 10.1038/s41598-023-34915-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 05/09/2023] [Indexed: 05/19/2023] Open
Abstract
Adipose tissue derived stromal cells (ADSCs) play a crucial role in research and applications of regenerative medicine because they can be rapidly isolated in high quantities. Nonetheless, their purity, pluripotency, differentiation capacity, and stem cell marker expression might vary greatly depending on technique and tools used for extraction and harvesting. There are two methods described in the literature for isolating regenerative cells from adipose tissue. The first technique is enzymatic digestion, which utilizes many enzymes to remove stem cells from the tissue they reside in. The second method involves separating the concentrated adipose tissue using non-enzymatic, mechanical separation methods. ADSCs are isolated from the stromal-vascular fraction (SVF) of processed lipoaspirate, which is the lipoaspirate's aqueous portion. The purpose of this work was to evaluate a unique device 'microlyzer' for generating SVF from adipose tissue using a mechanical technique that required minimal intervention. The Microlyzer was examined using tissue samples from ten different patients. The cells that were retrieved were characterized in terms of their cell survival, phenotype, proliferation capacity, and differentiation potential. The number of progenitor cells extracted only from the microlyzed tissue was in comparable amount to the number of progenitor cells acquired by the gold standard enzymatic approach. The cells that were collected from each group exhibit similar levels of viability as well as proliferation rates. In addition, the differentiation potentials of the cells derived from the microlyzed tissue were investigated, and it was discovered that cells isolated through microlyzer entered the differentiation pathways more quickly and displayed a greater level of marker gene expression than cells isolated by enzymatic methods. These findings suggest that microlyzer, particularly in regeneration investigations, will allow quick and high rate cell separation at the bedside.
Collapse
Affiliation(s)
- Seher Yaylacı
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, 06800, Turkey.
| | - Demet Kaçaroğlu
- Department of Medical Biology, Faculty of Medicine, Lokman Hekim University, Ankara, 06800, Turkey
| | - Özgür Hürkal
- Plastic Reconstructive and Aesthetic Surgery, Lokman Hekim Hospital, Ankara, 06800, Turkey
| | - Alper Murat Ulaşlı
- Physical Therapy and Rehabilitation, Faculty of Health Sciences, Lokman Hekim University, Ankara, 06800, Turkey
- Romatem Ankara Physical Therapy and Rehabilitation Center, Ankara, 06700, Turkey
| |
Collapse
|
31
|
Karimi T, Pan Z, Potaman VN, Alt EU. Conversion of Unmodified Stem Cells to Pacemaker Cells by Overexpression of Key Developmental Genes. Cells 2023; 12:1381. [PMID: 37408215 PMCID: PMC10216671 DOI: 10.3390/cells12101381] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 07/07/2023] Open
Abstract
Arrhythmias of the heart are currently treated by implanting electronic pacemakers and defibrillators. Unmodified adipose tissue-derived stem cells (ASCs) have the potential to differentiate into all three germ layers but have not yet been tested for the generation of pacemaker and Purkinje cells. We investigated if-based on overexpression of dominant conduction cell-specific genes in ASCs-biological pacemaker cells could be induced. Here we show that by overexpression of certain genes that are active during the natural development of the conduction system, the differentiation of ASCs to pacemaker and Purkinje-like cells is feasible. Our study revealed that the most effective procedure consisted of short-term upregulation of gene combinations SHOX2-TBX5-HCN2, and to a lesser extent SHOX2-TBX3-HCN2. Single-gene expression protocols were ineffective. Future clinical implantation of such pacemaker and Purkinje cells, derived from unmodified ASCs of the same patient, could open up new horizons for the treatment of arrythmias.
Collapse
Affiliation(s)
- Tahereh Karimi
- Heart and Vascular Institute, Department of Medicine, Tulane University Health Science Center, 1430 Tulane Ave, New Orleans, LA 70112, USA;
- Alliance of Cardiovascular Researchers, 2211 Augusta Dr #10, Houston, TX 77057, USA
| | - Zhizhong Pan
- University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vladimir N. Potaman
- Alliance of Cardiovascular Researchers, 2211 Augusta Dr #10, Houston, TX 77057, USA
- InGeneron Inc., 8205 El Rio Street, Houston, TX 77054, USA
| | - Eckhard U. Alt
- Heart and Vascular Institute, Department of Medicine, Tulane University Health Science Center, 1430 Tulane Ave, New Orleans, LA 70112, USA;
- Alliance of Cardiovascular Researchers, 2211 Augusta Dr #10, Houston, TX 77057, USA
- InGeneron Inc., 8205 El Rio Street, Houston, TX 77054, USA
- Sanford Health, University of South Dakota, Sioux Falls, SD 57104, USA
- Isar Klinikum Munich, Sonnenstr 24-26, 80331 Munich, Germany
| |
Collapse
|
32
|
Chang CJ, Hsiao YC, Hang NLT, Yang TS. Biophotonic Effects of Low-Level Laser Therapy on Adipose-Derived Stem Cells for Soft Tissue Deficiency. Ann Plast Surg 2023; 90:S158-S164. [PMID: 36752397 DOI: 10.1097/sap.0000000000003376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
PURPOSES The objectives of this study are to use diode lasers for low-level laser therapy (LLLT) and to assess its applicability and effects in adipose-derived stem cell (ADSC) growth processes. METHODS Studies were conducted on the diode laser with wavelengths of 622.7, 527.1, and 467.3 nm. The mechanism of action of LLL illumination was studied on ADSCs, isolated from human tissue, and then cultured by examining different wavelengths to determine the relevant light parameters for optimal responses. We used enzyme-linked immunosorbent assay and real-time polymerase chain to determine the percentages of fibroblast-mediated procollagen type 1 and matrix metallopeptidase 1 (MMP-1), MMP-2, and MMP-9 production at different wavelengths. The levels of lactate dehydrogenase produced by ADSCs after LLL illumination were assessed as well. Clinical results from 20 patients treated for soft tissue deficiency were collected for assessment of ADSC-assisted lipotransfer. RESULTS Low-level laser (622.7 nm) illumination on cell cultures in vitro increased ADSCs proliferation, type 1 procollagen expression, collagen production, as well as MMP-1, MMP-2, and MMP-9 relative expression. Statistical analysis demonstrated a significant difference in red light (622.7 nm) versus green light (527.1 nm) and blue light (467.3 nm, P < 0.05). No significant differences were noted between the effects of green and blue lights. In clinical application, all patients attained significant improvement with treatment in the final outcome assessment after 6 months. CONCLUSIONS Low-level laser illumination may affect ADSCs growth processes and ADSC-assisted lipotransfer for soft tissue deformity, scar treatment, wound healing, and other reconstructive surgery.
Collapse
Affiliation(s)
| | - Yen-Chang Hsiao
- From the Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung University
| | | | - Tzu-Sen Yang
- Graduate Institute of Biomedical Optomechatronics, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
33
|
El-Husseiny HM, Kaneda M, Mady EA, Yoshida T, Doghish AS, Tanaka R. Impact of Adipose Tissue Depot Harvesting Site on the Multilineage Induction Capacity of Male Rat Adipose-Derived Mesenchymal Stem Cells: An In Vitro Study. Int J Mol Sci 2023; 24:7513. [PMID: 37108673 PMCID: PMC10138771 DOI: 10.3390/ijms24087513] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Recently, substantial attention has been paid toward adipose-derived mesenchymal stem cells (AdMSCs) as a potential therapy in tissue engineering and regenerative medicine applications. Rat AdMSCs (r-AdMSCs) are frequently utilized. However, the influence of the adipose depot site on the multilineage differentiation potential of the r-AdMSCs is still ambiguous. Hence, the main objective of this study was to explore the influence of the adipose tissue harvesting location on the ability of r-AdMSCs to express the stem-cell-related markers and pluripotency genes, as well as their differentiation capacity, for the first time. Herein, we have isolated r-AdMSCs from the inguinal, epididymal, peri-renal, and back subcutaneous fats. Cells were compared in terms of their phenotype, immunophenotype, and expression of pluripotency genes using RT-PCR. Additionally, we investigated their potential for multilineage (adipogenic, osteogenic, and chondrogenic) induction using special stains confirmed by the expression of the related genes using RT-qPCR. All cells could positively express stem cell marker CD 90 and CD 105 with no significant in-between differences. However, they did not express the hematopoietic markers as CD 34 and CD 45. All cells could be induced successfully. However, epididymal and inguinal cells presented the highest capacity for adipogenic and osteogenic differentiation (21.36-fold and 11.63-fold for OPN, 29.69-fold and 26.68-fold for BMP2, and 37.67-fold and 22.35-fold for BSP, respectively, in epididymal and inguinal cells (p < 0.0001)). On the contrary, the subcutaneous cells exhibited a superior potential for chondrogenesis over the other sites (8.9-fold for CHM1 and 5.93-fold for ACAN, (p < 0.0001)). In conclusion, the adipose tissue harvesting site could influence the differentiation capacity of the isolated AdMSCs. To enhance the results of their employment in various regenerative cell-based therapies, it is thus vital to take the collection site selection into consideration.
Collapse
Affiliation(s)
- Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Division of Animal Life Sciences, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan
| | - Eman A. Mady
- Department of Animal Hygiene, Behavior, and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh 13736, Elqaliobiya, Egypt
| | - Tadashi Yoshida
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Ahmed S. Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City 11829, Cairo, Egypt
- Department of Biochemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11651, Cairo, Egypt
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi 183-8509, Tokyo, Japan
| |
Collapse
|
34
|
Ambrosio MR, Migliaccio T, Napolitano F, Di Somma S, Maneli G, Amato J, Pagano B, Randazzo A, Portella G, Formisano P, Malfitano AM. Targeting G-quadruplex motifs interferes with differentiation of adipose-derived mesenchymal stem cells. Stem Cell Res Ther 2023; 14:98. [PMID: 37076894 PMCID: PMC10116735 DOI: 10.1186/s13287-023-03320-9] [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: 11/24/2022] [Accepted: 03/29/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND G-quadruplex (G4) motifs are nucleic acid secondary structures observed in mammalian genomes and transcriptomes able to regulate various cellular processes. Several small molecules have been developed so far to modulate G4 stability, frequently associated with anticancer activity. However, how G4 structures are regulated over homeostatic conditions is mostly unexplored. Here, we used human adipose-derived mesenchymal stem cells (ASCs) to address the role of G4 motifs during adipogenic differentiation. METHODS Adipocyte differentiation of ASCs was investigated in the presence or absence of a well-known G4 ligand, Braco-19. Cell viability was determined by sulforhodamine B assay. Cell dimension and granularity, DNA G4 motifs and cell cycle were detected by flow cytometry. Lipid droplet accumulation was assessed by Oil Red O staining. Cell senescence was evaluated by β-galactosidase staining. Gene expression was measured by qPCR. Protein release in the extracellular medium was quantified by ELISA. RESULTS Braco-19 used at non-cytotoxic concentrations induced morphological changes in mature adipocytes partially restoring an undifferentiated-like status. Braco-19 reduced lipid vacuolization and PPARG, AP2, LEP and TNFA mRNA levels in terminally differentiated cells. No effect was observed in cell senescence, fibrotic markers, IL-6 and IL-8 production, while the secretion of VEGF was dose-dependently reduced. Interestingly, G4 structures were increased in differentiated adipocytes compared to their precursors. Braco-19 treatment reduced G4 content in mature adipocytes. CONCLUSIONS Our data highlight a new role of G4 motifs as genomic structural elements related to human ASC differentiation into mature adipocytes, with potential implications in physio-pathological processes.
Collapse
Affiliation(s)
- Maria Rosaria Ambrosio
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - Teresa Migliaccio
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Fabiana Napolitano
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Sarah Di Somma
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Giovanni Maneli
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
| | - Jussara Amato
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Bruno Pagano
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Giuseppe Portella
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy.
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy.
| | - Anna Maria Malfitano
- Department of Translational Medical Sciences, University "Federico II", 80131, Naples, Italy.
| |
Collapse
|
35
|
Ai G, Meng M, Guo J, Li C, Zhu J, Liu L, Liu B, Yang W, Shao X, Cheng Z, Wang L. Adipose-derived stem cells promote the repair of chemotherapy-induced premature ovarian failure by inhibiting granulosa cells apoptosis and senescence. Stem Cell Res Ther 2023; 14:75. [PMID: 37038203 PMCID: PMC10088140 DOI: 10.1186/s13287-023-03297-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 03/23/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Chemotherapeutic drugs, particularly alkylating cytotoxics such as cyclophosphamide (CTX), play an important role to induce premature ovarian failure (POF). Hormone replacement therapy (HRT) is a widely used treatment to improve hormone secretion. However, the long-term HRT increases the risk of breast cancer and cardiovascular disease are attracting concerns. Therefore, there is an urgent need to develop a safe and effective treatment for POF. METHOD Adipose-derived stem cells (ADSCs) were isolated and identified from human adipose tissue. For POF modeling, CTX were intraperitoneal injected into CTX-acute group, CTX-chronic group, CTX-acute + ADSCs group and CTX-chronic + ADSCs group rats; For transplantation, ADSCs were transplanted into POF rats through tail-vein. The control group rats were injected with PBS. The effects of POF modeling and transplantation were determined by estrous cycle analysis, histopathological analysis, immunohistochemical staining and apoptosis-related marker. To evaluate the effects of ADSC on granulosa cells in vitro, CTX-induced senescent KGN cells were co-cultured with ADSCs, and senescent-related marker expression was investigated by immunofluorescent staining. RESULTS In vivo studies revealed that ADSCs transplantation reduced the apoptosis of ovarian granulosa cells and secretion of follicle-stimulating hormone. The number of total follicles, primordial follicles, primary follicles, and mature follicles and secretion of anti-Müllerian hormone and estradiol (E2) were also increased by ADSCs. The estrous cycle was also improved by ADSC transplantation. Histopathological analysis showed that CTX-damaged ovarian microenvironment was improved by ADSCs. Furthermore, TUNEL staining indicated that apoptosis of granulosa cells was decreased by ADSCs. In vitro assay also demonstrated that ADSC markedly attenuated CTX-induced senescence and apoptosis of granulosa cell. Mechanistically, both in vivo and in vitro experiments proved that ADSC transplantation suppressed activation of the PI3K/Akt/mTOR axis. CONCLUSION Our experiment demonstrated that a single injection of high-dose CTX was a less damaging chemotherapeutic strategy than continuous injection of low-dose CTX, and tail-vein injection of ADSCs was a potential approach to promote the restoration of CTX-induced POF.
Collapse
Affiliation(s)
- Guihai Ai
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Meng Meng
- Department of Gynecology and Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Jing Guo
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Caixia Li
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jihui Zhu
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Li Liu
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Biting Liu
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Wenhan Yang
- Department of Gynecology and Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Xiaowen Shao
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhongping Cheng
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
- Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
- Tongji University School of Medicine, Shanghai, 200092, China.
| | - Lian Wang
- Department of Gynecology and Obstetrics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
- Gynecologic Minimally Invasive Surgery Research Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
36
|
Lara ML, Carvalho MG, de Souza FF, Schmith RA, Codognoto VM, De Vita B, Freitas Dell'Aqua CDP, Landim FDC, Alvarenga MLE. Influence of culture conditions on the secretome of mesenchymal stem cells derived from feline adipose tissue: Proteomics approach. Biochimie 2023; 211:78-86. [PMID: 36931338 DOI: 10.1016/j.biochi.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
This study aimed to describe the secretome of mesenchymal stem cells derived from feline adipose tissue (AD-MSCs) and compare the effects of different culture conditions on AD-MSC proteomics using a shotgun approach. Adipose tissue was collected from 5 female cats and prepared to culture. Conditioned media was collected at third passage, in which the cells were cultured under 4 conditions, normoxia with fetal bovine serum (N + FBS), hypoxia with FBS (H + FBS), normoxia without FBS (N - FBS), and hypoxia without FBS (H - FBS). Then, the secretome was concentrated and prepared for proteomic approaches. Secretomes cultured with FBS-free medium had more than twice identified proteins in comparison with the secretomes cultured with FBS. In contrast, hypoxic conditions did not increase protein amount and affected only a small proteome fraction. Relevant proteins were related to the extracellular matrix promoting environmental modulation, influencing cell signaling pathways, and providing a suitable environment for cell proliferation and maintenance. Moreover, other proteins were also related to cell adhesion, migration and morphogenesis. Culture conditions can influence protein abundance in AD-MSC secretome, and can give also more specificity to cell and cell-free treatments for different diseases.
Collapse
Affiliation(s)
- Maria Laura Lara
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Marcos Gomides Carvalho
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Fabiana Ferreira de Souza
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil.
| | - Rubia Alves Schmith
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Viviane Maria Codognoto
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Bruna De Vita
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil; International Product Marketing Manager - PROCARE HEALTH, Universitat de Barcelona, Barcelona, Catalunha, Spain
| | - Camila de Paula Freitas Dell'Aqua
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Fernada da Cruz Landim
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil
| | - Marina Landim E Alvarenga
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil; Omics Animal Biotechnology, Botucatu, São Paulo, Brazil
| |
Collapse
|
37
|
Abstract
SUMMARY Over the past 30 years, there has been a dramatic increase in the use of autologous fat grafting for soft-tissue augmentation and to improve facial skin quality. Several studies have highlighted the impact of aging on adipose tissue, leading to a decrease of adipose tissue volume and preadipocyte proliferation and increase of fibrosis. Recently, there has been a rising interest in adipose tissue components, including adipose-derived stem/stromal cells (ASCs) because of their regenerative potential, including inflammation, fibrosis, and vascularization modulation. Because of their differentiation potential and paracrine function, ASCs have been largely used for fat grafting procedures, as they are described to be a key component in fat graft survival. However, many parameters as surgical procedures or adipose tissue biology could change clinical outcomes. Variation on fat grafting methods have led to numerous inconsistent clinical outcomes. Donor-to-donor variation could also be imputed to ASCs, tissue inflammatory state, or tissue origin. In this review, the authors aim to analyze (1) the parameters involved in graft survival, and (2) the effect of aging on adipose tissue components, especially ASCs, that could lead to a decrease of skin regeneration and fat graft retention. CLINICAL RELEVANCE STATEMENT This review aims to enlighten surgeons about known parameters that could play a role in fat graft survival. ASCs and their potential mechanism of action in regenerative medicine are more specifically described.
Collapse
|
38
|
Mollashahi B, Latifi-Navid H, Owliaee I, Shamdani S, Uzan G, Jamehdor S, Naserian S. Research and Therapeutic Approaches in Stem Cell Genome Editing by CRISPR Toolkit. Molecules 2023; 28:1982. [PMID: 36838970 PMCID: PMC9961668 DOI: 10.3390/molecules28041982] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
The most widely used genome editing toolkit is CRISPR (clustered regularly interspaced short palindromic repeats). It provides the possibility of replacing and modifying DNA and RNA nucleotides. Furthermore, with advancements in biological technology, inhibition and activation of the transcription of specific gene(s) has become possible. Bioinformatics tools that target the evolution of CRISPR-associated protein 9 (Cas9) turn this protein into a vehicle that is specific for a DNA or RNA region with single guide RNA (sgRNA). This toolkit could be used by researchers to investigate the function of stem cell gene(s). Here, in this review article, we cover recent developments and applications of this technique in stem cells for research and clinical purposes and discuss different CRISPR/Cas technologies for knock-out, knock-in, activation, or inhibition of gene expression. Additionally, a comparison of several deliveries and off-target detecting strategies is discussed.
Collapse
Affiliation(s)
- Behrouz Mollashahi
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 14965/161, Iran
| | - Iman Owliaee
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamedan 6517838636, Iran
| | - Sara Shamdani
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Paris-Saclay University, 94807 Villejuif, France
- CellMedEx, 94100 Saint Maur Des Fossés, France
| | - Georges Uzan
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Paris-Saclay University, 94807 Villejuif, France
| | - Saleh Jamehdor
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamedan 6517838636, Iran
| | - Sina Naserian
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Paris-Saclay University, 94807 Villejuif, France
- CellMedEx, 94100 Saint Maur Des Fossés, France
| |
Collapse
|
39
|
Cruciani S, Delitala AP, Cossu ML, Ventura C, Maioli M. Management of Obesity and Obesity-Related Disorders: From Stem Cells and Epigenetics to Its Treatment. Int J Mol Sci 2023; 24:2310. [PMID: 36768633 PMCID: PMC9916844 DOI: 10.3390/ijms24032310] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Obesity is a complex worldwide disease, characterized by an abnormal or excessive fat accumulation. The onset of this pathology is generally linked to a complex network of interactions among genetic and environmental factors, aging, lifestyle, and diets. During adipogenesis, several regulatory mechanisms and transcription factors are involved. As fat cells grow, adipose tissue becomes increasingly large and dysfunctional, losing its endocrine function, secreting pro-inflammatory cytokines, and recruiting infiltrating macrophages. This long-term low-grade systemic inflammation results in insulin resistance in peripheral tissues. In this review we describe the main mechanisms involved in adipogenesis, from a physiological condition to obesity. Current therapeutic strategies for the management of obesity and the related metabolic syndrome are also reported.
Collapse
Affiliation(s)
- Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Consorzio Interuniversitario “Istituto Nazionale Biostrutture e Biosistemi” (INBB), Viale delle Medaglie d’Oro 305, 00136 Roma, Italy
| | | | - Maria Laura Cossu
- General Surgery Unit 2 “Clinica Chirurgica” Medical, Surgical and Experimental Sciences Department, University of Sassari, 07100 Sassari, Italy
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Engineering, Eldor Lab, Istituto Nazionale di Biostrutture e Biosistemi (INBB), Via di Corticella 183, 40128 Bologna, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Consorzio Interuniversitario “Istituto Nazionale Biostrutture e Biosistemi” (INBB), Viale delle Medaglie d’Oro 305, 00136 Roma, Italy
- Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| |
Collapse
|
40
|
Diri D, Alasaad H, Muhammed H, Ibrahim J. Case report: adipose-derived mesenchymal stem cells combined with core decompression in the treatment of early-stage avascular necrosis of the femoral head. Int J Surg Case Rep 2022; 102:107861. [PMID: 36603496 PMCID: PMC9826853 DOI: 10.1016/j.ijscr.2022.107861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/25/2022] [Accepted: 12/29/2022] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Core decompression is a well-known modality for treating the early stages of avascular necrosis of the femoral head (AVN), however, several methods have been suggested to augment this procedure and improve the outcomes. CASE REPORT A 52 male was diagnosed with a stage I AVN of the femoral head and treated with core decompression (CD) and injection of adipose-derived mesenchymal stem cells (AD-MSCs). The MRI showed full healing of the lesion after 3 months with significant clinical and functional improvement. DISCUSSION AD-MSCs could have the same capabilities as bone marrow-derived stem cells with many advantages, implantation of AD-MSCs in orthopedics and as an augmentation of core decompression has been tried before, but no clear guidelines nor methods of application are well established in the literature. CONCLUSION Implantation of AD-MSCs with Core decompression could be an effective modality to treat osteonecrosis of the femoral head in pre-collapse stages, however, we need bigger clinical studies to determine the actual effectiveness of this method.
Collapse
|
41
|
ÖZGENÇ Ö, ÖZEN A. Osteogenic Differentiation of Canine Adipose Derived Mesenchymal Stem Cells on B-TCP and B-TCP/Collagen Biomaterials. ANKARA ÜNIVERSITESI VETERINER FAKÜLTESI DERGISI 2022. [DOI: 10.33988/auvfd.1130705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Mesenchymal stem cells are adult stem cells that have the ability to differentiate into osteogenic, chondrogenic, adipogenic and myogenic lineages. In the field of orthopedics and traumatology, mesenchymal stem cells in combination with biomaterials are used especially for the treatment of bone fractures and diseases in both humans and animals. The purpose of this study is to promote growth, proliferation and osteogenic differentiation of mesenchymal stem cells that were isolated from the adipose tissue of canines on B-TCP (Beta-tricalcium phosphate) and B-TCP/Collagen biomaterials. MTT analysis was performed to test the cell adhesion and proliferation on B-TCP and B-TCP/Collagen biomaterials that were used to mimic the extracellular matrix of three-dimensional bone tissue. Scanning electron microscope analysis was performed to show general surface characters of B-TCP and B-TCP /Collagen biomaterials. The osteoinductive capacities of the B-TCP and B-TCP/Collagen biomaterials were determined by alkaline phosphatase and Von Kossa stainings, and RT-PCR analysis. The ALP activity of the B-TCP/Col containing material was significantly higher than the B-TCP on the first days. In terms of gene expression, there were no significant differences except 14th-day SPARC gene expression. The results of Von Kossa staining indicate that B-TCP/Col has above the desired level degradation capacity. As a result of this research, although it is advantageous in terms of alkaline phosphatase activity and osteogenic gene expression compared to B-TCP material, it is thought that B-TCP/Collagen biomaterial should be developed for use in bone tissue engineering due to its high degradation property.
Collapse
|
42
|
Zheng Z, Li P, Shen F, Shi Y, Shao C. Mesenchymal Stem/Stromal Cells in Cancer: from Initiation to Metastasis. Arch Med Res 2022; 53:785-793. [PMID: 36462949 DOI: 10.1016/j.arcmed.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/02/2022] [Indexed: 12/05/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) exist in many tissues and have pleiotropic potential to self-renew and differentiate into multiple cell types. Recent research in tumor biology has focused on their low immunogenicity and tumorhoming properties. MSCs promote cancer initiation, progression, and metastasis through several different mechanisms, including epithelial-mesenchymal transition (EMT), angiogenesis, and through their interaction with immune cells. In this review, we discuss the recent advances in our understanding of the pathogenic role of MSCs in regulating tumor initiation, progression, and metastasis, thus providing a strong rationale for targeting MSCs in cancer therapy.
Collapse
Affiliation(s)
- Zhiyuan Zheng
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Cancer Center, Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Peng Li
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Cancer Center, Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fangrong Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China.
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
43
|
Optimizing aseptic and serum milieu for the isolation of human whole umbilical cord tissue-derived mesenchymal stem cells. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2022. [DOI: 10.1186/s43088-022-00308-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Mesenchymal stem cells (MSCs) have become an attractive tool for tissue engineering and targets in clinical transplantation due to their regeneration potential and immune-suppressive capacity. The human umbilical cord, which is discarded at birth, can provide an inexhaustible source of stem cells for therapy. They are reported to contain immune privilege cells which may be suitable for allogenic-based therapies. However, the use of MSCs for therapeutic application is based on their subsequent large-scale in vitro expansion. A fast and efficient protocol for the generation of large quantities of MSCs is required to meet the clinical demand and biomedical research needs.
Results
MSCs were isolated from the umbilical cord by explants and enzymatic digestion and cultured in the appropriate growth medium resulted in the propagation of more than 1X 108 cells within 15 days from the single umbilical cord.
Conclusion
The isolation efficiency, cell yield, colony-forming unit fibroblast (CFU-F), growth kinetics, phenotypic characteristics of UCMSCs were determined.
Collapse
|
44
|
Lenna S, Brozovich A, Hirase T, Paradiso F, Weiner BK, Taraballi F. Comparison between Cancellous Trabecular and Cortical Specimens from Human Lumbar Spine Samples as an Alternative Source of Mesenchymal Stromal Cells. Stem Cells Dev 2022; 31:672-683. [PMID: 36039931 DOI: 10.1089/scd.2022.0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Due to their immunosuppressive potential and ability to differentiate into multiple musculoskeletal cell lineages, mesenchymal stromal cells (MSCs) became popular in clinical trials for the treatment of musculoskeletal disorders. The aim of this study was to isolate and characterize native populations of MSCs from human cortical and cancellous bone from the posterior elements of the lumbar spine and determine what source of MSCs yield better quality and quantity of cells to be potentially use for spinal fusion repair. We were able to show that MSCs from trabecular and cortical spine had the typical MSC morphology and expression markers; the ability to differentiate in adipocyte, chondrocyte, or osteoblast but they did not have a consistent pattern in the expression of the specific differentiation lineage genes. Moreover, MSCs from both sites demonstrated an immune suppression profile suggesting that these cells may have a more promising success in applications related to immunomodulation more than exploring their ability to drive osteogenesis to prevent nonunion in spine fusion procedures.
Collapse
Affiliation(s)
- Stefania Lenna
- Houston Methodist Research Institute, Houston, Texas, United States;
| | - Ava Brozovich
- Houston Methodist Academic Institute, Houston, Texas, United States;
| | - Takashi Hirase
- Houston Methodist Orthopedics & Sports Medicine Texas Medical Center, Houston, Texas, United States;
| | | | - Bradley K Weiner
- The Houston Methodist Research Institute, Department of Nanomedicine, Houston, Texas, United States.,Houston Methodist Hospital, Department of Orthopedic Surgery, Houston, Texas, United States;
| | | |
Collapse
|
45
|
Ibrahim AS, El-Shinawi M, Sabet S, Ibrahim SA, Mohamed MM. Role of adipose tissue-derived cytokines in the progression of inflammatory breast cancer in patients with obesity. Lipids Health Dis 2022; 21:67. [PMID: 35927653 PMCID: PMC9351154 DOI: 10.1186/s12944-022-01678-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Inflammatory breast cancer (IBC) represents a deadly aggressive phenotype of breast cancer (BC) with a unique clinicopathological presentation and low survival rate. In fact, obesity represents an important risk factor for BC. Although several studies have identified different cellular-derived and molecular factors involved in IBC progression, the role of adipocytes remains unclear. Cancer-associated adipose tissue (CAAT) expresses a variety of adipokines, which contribute to tumorigenesis and the regulation of cancer stem cell (CSC). This research investigated the potential effect of the secretome of CAAT explants from patients with BC on the progression and metastasis of the disease. METHODS This study established an ex-vivo culture of CAAT excised from IBC (n = 13) vs. non-IBC (n = 31) patients with obesity and profiled their secretome using a cytokine antibody array. Furthermore, the quantitative PCR (qPCR) methodology was used to validate the levels of predominant cytokines at the transcript level after culture in a medium conditioned by CAAT. Moreover, the impact of the CAAT secretome on the expression of epithelial-mesenchymal transition (EMT) and cells with stem cell (CSC) markers was studied in the non-IBC MDA-MB-231 and the IBC SUM-149 cell lines. The statistical differences between variables were evaluated using the chi-squared test and unpaired a Student's t-test. RESULTS The results of cytokine array profiling revealed an overall significantly higher level of a panel of 28 cytokines secreted by the CAAT ex-vivo culture from IBC patients with obesity compared to those with non-IBC. Of note, interleukin-6 (IL-6), interleukin-8 (IL-8), and monocyte chemo-attractant protein 1 (MCP-1) were the major adipokines secreted by the CAAT IBC patients with obesity. Moreover, the qPCR results indicated a significant upregulation of the IL-6, IL-8, and MCP-1 mRNAs in CAAT ex-vivo culture of patients with IBC vs. those with non-IBC. Intriguingly, a qPCR data analysis showed that the CAAT secretome secretions from patients with non-IBC downregulated the mRNA levels of the CD24 CSC marker and of the epithelial marker E-cadherin in the non-IBC cell line. By contrast, E-cadherin was upregulated in the SUM-149 cell. CONCLUSIONS This study identified the overexpression of IL-6, IL-8, and MCP-1 as prognostic markers of CAAT from patients with IBC but not from those with non-IBC ; moreover, their upregulation might be associated with IBC aggressiveness via the regulation of CSC and EMT markers. This study proposed that targeting IL-6, IL-8, and MCP-1 may represent a therapeutic option that should be considered in the treatment of patients with IBC.
Collapse
Affiliation(s)
- Aya Saber Ibrahim
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| | - Mohamed El-Shinawi
- Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt
- International Affairs, Galala University, Suez, Egypt
| | - Salwa Sabet
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | | | - Mona Mostafa Mohamed
- Zoology Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
- Molecular Biotechnology Program, Faculty of Science, Galala University, Suez, Egypt
| |
Collapse
|
46
|
Flehr A, Källgård J, Alvén J, Lagerstrand K, Papalini E, Wheeler M, Vandenput L, Kahl F, Axelsson KF, Sundh D, Mysore RS, Lorentzon M. Development of a novel method to measure bone marrow fat fraction in older women using high-resolution peripheral quantitative computed tomography. Osteoporos Int 2022; 33:1545-1556. [PMID: 35113175 PMCID: PMC9187531 DOI: 10.1007/s00198-021-06224-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/26/2021] [Indexed: 11/24/2022]
Abstract
UNLABELLED Bone marrow adipose tissue (BMAT) has been implicated in a number of conditions associated with bone deterioration and osteoporosis. Several studies have found an inverse relationship between BMAT and bone mineral density (BMD), and higher levels of BMAT in those with prevalent fracture. Magnetic resonance imaging (MRI) is the gold standard for measuring BMAT, but its use is limited by high costs and low availability. We hypothesized that BMAT could also be accurately quantified using high-resolution peripheral quantitative computed tomography (HR-pQCT). METHODS In the present study, a novel method to quantify the tibia bone marrow fat fraction, defined by MRI, using HR-pQCT was developed. In total, 38 postmenopausal women (mean [standard deviation] age 75.9 [3.1] years) were included and measured at the same site at the distal (n = 38) and ultradistal (n = 18) tibia using both MRI and HR-pQCT. To adjust for partial volume effects, the HR-pQCT images underwent 0 to 10 layers of voxel peeling to remove voxels adjacent to the bone. Linear regression equations were then tested for different degrees of voxel peeling, using the MRI-derived fat fractions as the dependent variable and the HR-pQCT-derived radiodensity as the independent variables. RESULTS The most optimal HR-pQCT derived model, which applied a minimum of 4 layers of peeled voxel and with more than 1% remaining marrow volume, was able to explain 76% of the variation in the ultradistal tibia bone marrow fat fraction, measured with MRI (p < 0.001). CONCLUSION The novel HR-pQCT method, developed to estimate BMAT, was able to explain a substantial part of the variation in the bone marrow fat fraction and can be used in future studies investigating the role of BMAT in osteoporosis and fracture prediction.
Collapse
Affiliation(s)
- Alison Flehr
- Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Julius Källgård
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jennifer Alvén
- Dept. of Molecular and Clinical Medicine, Inst. of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Computer Vision and Medical Image Analysis, Dept. of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Kerstin Lagerstrand
- Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Radiation Physics, Institution of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Evin Papalini
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Michael Wheeler
- Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Liesbeth Vandenput
- Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Kahl
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Kristian F Axelsson
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Primary Care, Skövde, Sweden
| | - Daniel Sundh
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Raghunath Shirish Mysore
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mattias Lorentzon
- Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia.
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Region Västra Götaland, Geriatric Medicine, Sahlgrenska University Hospital, Mölndal, Sweden.
| |
Collapse
|
47
|
Krawczenko A, Klimczak A. Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells and Their Contribution to Angiogenic Processes in Tissue Regeneration. Int J Mol Sci 2022; 23:ijms23052425. [PMID: 35269568 PMCID: PMC8910401 DOI: 10.3390/ijms23052425] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are widely described in the context of their regenerative and immunomodulatory activity. MSCs are isolated from various tissues and organs. The most frequently described sources are bone marrow and adipose tissue. As stem cells, MSCs are able to differentiate into other cell lineages, but they are usually reported with respect to their paracrine potential. In this review, we focus on MSCs derived from adipose tissue (AT-MSCs) and their secretome in regeneration processes. Special attention is given to the contribution of AT-MSCs and their derivatives to angiogenic processes described mainly in the context of angiogenic dysfunction. Finally, we present clinical trials registered to date that concern the application of AT-MSCs and their secretome in various medical conditions.
Collapse
|
48
|
Adipose-Derived Stem Cell-Incubated HA-Rich Sponge Matrix Implant Modulates Oxidative Stress to Enhance VEGF and TGF-β Secretions for Extracellular Matrix Reconstruction In Vivo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9355692. [PMID: 35082971 PMCID: PMC8786469 DOI: 10.1155/2022/9355692] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/20/2021] [Indexed: 12/11/2022]
Abstract
This study demonstrated both adipose-derived stem cells (ASCs) in vitro and in vivo combined with three-dimensional (3D) porous sponge matrices on implant wound healing. Sponge matrices were created from hyaluronic acid (HA), collagen (Col), and gelatin (Gel), constructing two types: HA-L (low content) and HA-H (high content), to be cross-linked with 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC). Fourier transform infrared spectroscopy method verified carboxyl groups of HA and amino groups of Col and Gel reacting between the raw materials and scaffolds to identify the successive cross-linking. The swelling ratios of two types of sponge matrices were analyzed by water absorption capabilities, and the results displayed both over 30-fold dry scaffold weight enhancements. In biodegradation tests, matrices were hydrolyzed over time by three cutaneous enzymes, hyaluronidase, lysozyme, and collagenase I. ASCs from rats were cultured within the HA-H scaffold, demonstrating higher antioxidative abilities and secretions on related genes and proteins compared to the other two groups. The ASC HA-H matrix promoted cell proliferation to stimulate capillary angiogenesis inducer secretions, including vascular endothelial growth factor (VEGF) and transforming growth factor-β (TGF-β). In vivo histological examinations showed ASCs from implanted HA-H implant transported into the subcutis, and rat skin cells also infiltrated into the original matrix zone to increase the extracellular matrix (ECM) reconstructions. Our experimental data revealed that the ASC HA-H sponge implant was effective in improving wound repair.
Collapse
|
49
|
Liu Y, Dzidotor G, Le TT, Vinikoor T, Morgan K, Curry EJ, Das R, McClinton A, Eisenberg E, Apuzzo LN, Tran KTM, Prasad P, Flanagan TJ, Lee SW, Kan HM, Chorsi MT, Lo KWH, Laurencin CT, Nguyen TD. Exercise-induced piezoelectric stimulation for cartilage regeneration in rabbits. Sci Transl Med 2022; 14:eabi7282. [PMID: 35020409 DOI: 10.1126/scitranslmed.abi7282] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
More than 32.5 million American adults suffer from osteoarthritis, and current treatments including pain medicines and anti-inflammatory drugs only alleviate symptoms but do not cure the disease. Here, we have demonstrated that a biodegradable piezoelectric poly(L-lactic acid) (PLLA) nanofiber scaffold under applied force or joint load could act as a battery-less electrical stimulator to promote chondrogenesis and cartilage regeneration. The PLLA scaffold under applied force or joint load generated a controllable piezoelectric charge, which promoted extracellular protein adsorption, facilitated cell migration or recruitment, induced endogenous TGF-β via calcium signaling pathway, and improved chondrogenesis and cartilage regeneration both in vitro and in vivo. Rabbits with critical-sized osteochondral defects receiving the piezoelectric scaffold and exercise treatment experienced hyaline-cartilage regeneration and completely healed cartilage with abundant chondrocytes and type II collagen after 1 to 2 months of exercise (2 to 3 months after surgery including 1 month of recovery before exercise), whereas rabbits treated with nonpiezoelectric scaffold and exercise treatment had unfilled defect and limited healing. The approach of combining biodegradable piezoelectric tissue scaffolds with controlled mechanical activation (via physical exercise) may therefore be useful for the treatment of osteoarthritis and is potentially applicable to regenerating other injured tissues.
Collapse
Affiliation(s)
- Yang Liu
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Godwin Dzidotor
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Thinh T Le
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Tra Vinikoor
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Kristin Morgan
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Eli J Curry
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ritopa Das
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Aneesah McClinton
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Ellen Eisenberg
- Division of Oral and Maxillofacial Diagnostic Sciences, School of Dental Medicine, University of Connecticut, Farmington, CT 06030, USA
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Lorraine N Apuzzo
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
| | - Khanh T M Tran
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Pooja Prasad
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
| | - Tyler J Flanagan
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Seok-Woo Lee
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Ho-Man Kan
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Meysam T Chorsi
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Kevin W H Lo
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Thanh D Nguyen
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
50
|
Ragni E, Viganò M, De Luca P, Pedrini E, de Girolamo L. Adipose-Derived Stem/Stromal Cells, Stromal Vascular Fraction, and Microfragmented Adipose Tissue. ORTHOBIOLOGICS 2022:47-61. [DOI: 10.1007/978-3-030-84744-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|