1
|
Cui S, Wang L, Zhao H, Lu F, Chen Y, Gu Y. Skin wound repairing effects of adipose mesenchymal stem cells is promoted by the combined application of insulin-like growth factor 1: The key role of miR-21-5p-mediated signaling transduction. Cytokine 2024; 184:156760. [PMID: 39317084 DOI: 10.1016/j.cyto.2024.156760] [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: 06/17/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Mesenchymal stem cells (ADMSCs) have been applied to the treatment of skin injuries and the co-administration of cytokines can enhance the effects. In the current study, the promoting effects of insulin-like growth factor 1 (IGF-1) on the skin wound healing effects of adipose-derived MSCs (ADMSCs) were assessed and the associated mechanism was explored by focusing on miR-21-5p mediated pathways. ADMSCs were isolated from epididymis rats, and skin wounded rats were employed as the in vivo model for evaluating the effect of ADMCs on skin healing and secretion of cytokines. Then a microarray assay was employed to select potential miR target of IGF-1 on ADMSCs. The level of the selected miR was modulated in ADMSCs, and the effects on skin injuries were also assessed. Administration of ADMSCs promoted skin wound healing and induced the production of bFGF, IL-1β, PDGF, SDF-1, IGF-1, and TNF-α. The co-administration of IGF-1 and ADMSCs strengthened the effect of ADMSCs on skin wound by suppressing activity of matrix metalloproteinase-1 (MMP-1). At molecular level, the treatment of IGF-1 up-regulated miR-21-5p level in ADMSCs, which then suppressed the expression of KLF6 in injured skin tissues and promoted wound healing. The inhibition of miR-21-5p counteracted the promoting effects of IGF-1 on the skin healing effects of ADMSCs. Findings outlined in the current study indicated that IGF-1 could promote the wound healing effects of ADMSCs by up-regulating miR-21-5p level.
Collapse
Affiliation(s)
- Shuo Cui
- Department of Microsurgery, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | | | - Huafei Zhao
- Department of Microsurgery, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Fei Lu
- Department of Microsurgery, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Yuhua Chen
- Department of Microsurgery, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Yadong Gu
- Department of Microsurgery, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| |
Collapse
|
2
|
Teratani T, Fujimoto Y, Sakuma Y, Kasahara N, Maeda M, Miki A, Lefor AK, Sata N, Kitayama J. Improved Preservation of Rat Small Intestine Transplantation Graft by Introduction of Mesenchymal Stem Cell-Secreted Fractions. Transpl Int 2024; 37:11336. [PMID: 38962471 PMCID: PMC11219629 DOI: 10.3389/ti.2024.11336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
Segmental grafts from living donors have advantages over grafts from deceased donors when used for small intestine transplantation. However, storage time for small intestine grafts can be extremely short and optimal graft preservation conditions for short-term storage remain undetermined. Secreted factors from mesenchymal stem cells (MSCs) that allow direct activation of preserved small intestine grafts. Freshly excised Luc-Tg LEW rat tissues were incubated in preservation solutions containing MSC-conditioned medium (MSC-CM). Preserved Luc-Tg rat-derived grafts were then transplanted to wild-type recipients, after which survival, injury score, and tight junction protein expression were examined. Luminance for each graft was determined using in vivo imaging. The findings indicated that 30-100 and 3-10 kDa fractions of MSC-CM have superior activating effects for small intestine preservation. Expression of the tight-junction proteins claudin-3, and zonula occludens-1 preserved for 24 h in University of Wisconsin (UW) solution containing MSC-CM with 50-100 kDa, as shown by immunostaining, also indicated effectiveness. Reflecting the improved graft preservation, MSC-CM preloading of grafts increased survival rate from 0% to 87%. This is the first report of successful transplantation of small intestine grafts preserved for more than 24 h using a rodent model to evaluate graft preservation conditions that mimic clinical conditions.
Collapse
Affiliation(s)
- Takumi Teratani
- Division of Translational Research, Jichi Medical University, Tochigi, Japan
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Yasuhiro Fujimoto
- Transplantation Surgery, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Yasunaru Sakuma
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Naoya Kasahara
- Division of Translational Research, Jichi Medical University, Tochigi, Japan
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Masashi Maeda
- Division of Translational Research, Jichi Medical University, Tochigi, Japan
| | - Atsushi Miki
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | | | - Naohiro Sata
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Joji Kitayama
- Division of Translational Research, Jichi Medical University, Tochigi, Japan
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
3
|
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
|
4
|
Yamada S, Yamada K, Sugawara-Narutaki A, Baba Y, Yukawa H. Near-infrared-II fluorescence/magnetic resonance double modal imaging of transplanted stem cells using lanthanide co-doped gadolinium oxide nanoparticles. ANAL SCI 2024; 40:1043-1050. [PMID: 38430367 DOI: 10.1007/s44211-024-00507-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/04/2024] [Indexed: 03/03/2024]
Abstract
To ensure maximum therapeutic safety and efficacy of stem cell transplantation, it is essential to observe the kinetics of behavior, accumulation, and engraftment of transplanted stem cells in vivo. However, it is difficult to detect transplanted stem cells with high sensitivity by conventional in vivo imaging technologies. To diagnose the kinetics of transplanted stem cells, we prepared multifunctional nanoparticles, Gd2O3 co-doped with Er3+ and Yb3+ (Gd2O3: Er, Yb-NPs), and developed an in vivo double modal imaging technique with near-infrared-II (NIR-II) fluorescence imaging and magnetic resonance imaging (MRI) of stem cells using Gd2O3: Er, Yb-NPs. Gd2O3: Er, Yb-NPs were transduced into adipose tissue-derived stem cells (ASCs) through a simple incubation process without cytotoxicity under certain concentrations of Gd2O3: Er, Yb-NPs and were found not to affect the morphology of ASCs. ASCs labeled with Gd2O3: Er, Yb-NPs were transplanted subcutaneously onto the backs of mice, and successfully imaged with good contrast using an in vivo NIR-II fluorescence imaging and MRI system. These data suggest that Gd2O3: Er, Yb-NPs may be useful for in vivo double modal imaging with NIR-II fluorescence imaging and MRI of transplanted stem cells.
Collapse
Affiliation(s)
- Shota Yamada
- Department of Energy Engineering, Graduate School of Engineering, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8603, Japan.
| | - Kaori Yamada
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8603, Japan
| | - Ayae Sugawara-Narutaki
- Department of Energy Engineering, Graduate School of Engineering, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8603, Japan
| | - Yoshinobu Baba
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8603, Japan
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8603, Japan
- Institute for Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Anagawa 4-9-1, Inage-Ku, Chiba, 263-8555, Japan
- Department of Medical-Engineering Collaboration Supported by SEI Group CSR Foundation, Nagoya University, Tsurumai 65, Showa-Ku, Nagoya, 466-8550, Japan
| | - Hiroshi Yukawa
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8603, Japan.
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8603, Japan.
- Institute for Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Anagawa 4-9-1, Inage-Ku, Chiba, 263-8555, Japan.
- Department of Medical-Engineering Collaboration Supported by SEI Group CSR Foundation, Nagoya University, Tsurumai 65, Showa-Ku, Nagoya, 466-8550, Japan.
- B-3Frontier, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), Institute for Advanced Research, Nagoya University, Tsurumai 65, Showa-Ku, Nagoya, 466-8550, Japan.
- Department of Quantum Life Science, Graduate School of Science, Chiba University, Chiba, Japan.
| |
Collapse
|
5
|
Egorikhina MN, Rubtsova YP, Linkova DD, Charykova IN, Farafontova EA, Aleinik DY. Specifics of Cryopreservation of Hydrogel Biopolymer Scaffolds with Encapsulated Mesenchymal Stem Cells. Polymers (Basel) 2024; 16:247. [PMID: 38257046 PMCID: PMC10820988 DOI: 10.3390/polym16020247] [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: 11/03/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
The demand for regenerative medicine products is growing rapidly in clinical practice. Unfortunately, their use has certain limitations. One of these, which significantly constrains the widespread distribution and commercialization of such materials, is their short life span. For products containing suspensions of cells, this issue can be solved by using cryopreservation. However, this approach is rarely used for multicomponent tissue-engineered products due to the complexity of selecting appropriate cryopreservation protocols and the lack of established criteria for assessing the quality of such products once defrosted. Our research is aimed at developing a cryopreservation protocol for an original hydrogel scaffold with encapsulated MSCs and developing a set of criteria for assessing the quality of their functional activity in vitro. The scaffolds were frozen using two alternative types of cryocontainers and stored at either -40 °C or -80 °C. After cryopreservation, the external state of the scaffolds was evaluated in addition to recording the cell viability, visible changes during subsequent cultivation, and any alterations in proliferative and secretory activity. These observations were compared to those of scaffolds cultivated without cryopreservation. It was shown that cryopreservation at -80 °C in an appropriate type of cryocontainer was optimal for the hydrogels/adipose-derived stem cells (ASCs) tested if it provided a smooth temperature decrease during freezing over a period of at least three hours until the target values of the cryopreservation temperature regimen were reached. It was shown that evaluating a set of indicators, including the viability, the morphology, and the proliferative and secretory activity of the cells, enables the characterization of the quality of a tissue-engineered construct after its withdrawal from cryopreservation, as well as indicating the effectiveness of the cryopreservation protocol.
Collapse
Affiliation(s)
| | | | - Daria D. Linkova
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation (FSBEI HE PRMU MOH), 603600 Nizhny Novgorod, Russia; (M.N.E.); (Y.P.R.); (I.N.C.); (D.Y.A.)
| | | | | | | |
Collapse
|
6
|
Inoue O, Goten C, Hashimuko D, Yamaguchi K, Takeda Y, Nomura A, Ootsuji H, Takashima S, Iino K, Takemura H, Halurkar M, Lim HW, Hwa V, Sanchez-Gurmaches J, Usui S, Takamura M. Single-cell transcriptomics identifies adipose tissue CD271 + progenitors for enhanced angiogenesis in limb ischemia. Cell Rep Med 2023; 4:101337. [PMID: 38118404 PMCID: PMC10772587 DOI: 10.1016/j.xcrm.2023.101337] [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: 02/13/2023] [Revised: 07/10/2023] [Accepted: 11/21/2023] [Indexed: 12/22/2023]
Abstract
Therapeutic angiogenesis using mesenchymal stem/stromal cell grafts have shown modest and controversial effects in preventing amputation for patients with critical limb ischemia. Through single-cell transcriptomic analysis of human tissues, we identify CD271+ progenitors specifically from subcutaneous adipose tissue (AT) as having the most prominent pro-angiogenic gene profile distinct from other stem cell populations. AT-CD271+ progenitors demonstrate robust in vivo angiogenic capacity over conventional adipose stromal cell grafts, characterized by long-term engraftment, augmented tissue regeneration, and significant recovery of blood flow in a xenograft model of limb ischemia. Mechanistically, the angiogenic capacity of CD271+ progenitors is dependent on functional CD271 and mTOR signaling. Notably, the number and angiogenic capacity of CD271+ progenitors are strikingly reduced in insulin-resistant donors. Our study highlights the identification of AT-CD271+ progenitors with in vivo superior efficacy for limb ischemia. Furthermore, we showcase comprehensive single-cell transcriptomics strategies for identification of suitable grafts for cell therapy.
Collapse
Affiliation(s)
- Oto Inoue
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan; Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Chiaki Goten
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Daiki Hashimuko
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kosei Yamaguchi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yusuke Takeda
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Ayano Nomura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Ootsuji
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shinichiro Takashima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kenji Iino
- Department of Thoracic, Cardiovascular and General Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hirofumi Takemura
- Department of Thoracic, Cardiovascular and General Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Manasi Halurkar
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Vivian Hwa
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Premium Research Institute for Human Medicine (WPI-PRIMe), Osaka University, Osaka, Japan; Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joan Sanchez-Gurmaches
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Soichiro Usui
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
7
|
Farhana S, Kai YC, Kadir R, Sulaiman WAW, Nordin NA, Nasir NAM. The fate of adipose tissue and adipose-derived stem cells in allograft. Cell Tissue Res 2023; 394:269-292. [PMID: 37624425 DOI: 10.1007/s00441-023-03827-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023]
Abstract
Utilizing adipose tissue and adipose-derived stem cells (ADSCs) turned into a promising field of allograft in recent years. The therapeutic potential of adipose tissue and ADSCs is governed by their molecular secretions, ability to sustain multi-differentiation and self-renewal which are pivotal in reconstructive, genetic diseases, and cosmetic goals. However, revisiting the existing functional capacity of adipose tissue and ADSCs and their intricate relationship with allograft is crucial to figure out the remarkable question of safety to use in allograft due to the growing evidence of interactions between tumor microenvironment and ADSCs. For instance, the molecular secretions of adipose tissue and ADSCs induce angiogenesis, create growth factors, and control the inflammatory response; it has now been well determined. Though the existing preclinical allograft studies gave positive feedback, ADSCs and adipose tissue are attracted by some factors of tumor stroma. Moreover, allorecognition is pivotal to allograft rejection which is carried out by costimulation in a complement-dependent way and leads to the destruction of the donor cells. However, extensive preclinical trials of adipose tissue and ADSCs in allograft at molecular level are still limited. Hence, comprehensive immunomodulatory analysis could ensure the successful allograft of adipose tissue and ADSCs avoiding the oncological risk.
Collapse
Affiliation(s)
- Sadia Farhana
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Yew Chun Kai
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Ramlah Kadir
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Wan Azman Wan Sulaiman
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Nor Asyikin Nordin
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Nur Azida Mohd Nasir
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia.
| |
Collapse
|
8
|
Sharqawi A, Mansour MF, Elatrash GA, Ismail EA, Ralph D, El-Sakka AI. Role of adipose-derived stem cells in healing surgically induced trauma of the rat's tunica albuginea. Sex Med 2023; 11:qfad058. [PMID: 38028732 PMCID: PMC10661659 DOI: 10.1093/sexmed/qfad058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Background Injection of adipose-derived stem cells (ADSCs) into the injured tunica albuginea (TA) may prevent fibrosis, restore the balance between pro- and antifibrotic pathways, and potentially mitigate erectile dysfunction caused by abnormal TA healing. Aim To assess the potential role of ADSC injection on structural, ultrastructural, functional, and molecular changes in surgically induced trauma of the rat's TA. Methods Forty adult male albino Wistar rats were divided into 5 groups of 8 rats each: group 1, sham; group 2, injury to TA without treatment; group 3, injury to TA and suture repair; group 4, injury to TA and injection of ADSCs without suture repair; group 5, injury to TA followed by injection of ADSCs and suture repair. Outcomes After 6 weeks, all groups were subjected to functional, histologic, and ultrastructural examination and molecular expression of healing growth factors. Results The intracavernous pressure (ICP; mean ± SD) was 114 ± 2, 32 ± 2, 65 ± 2, 68 ± 2, and 111 ± 2 mm Hg in groups 1 to 5, respectively. There were significant differences in ICP between each of groups 3 to 5 and group 2 (P < .05), and groups 3 and 4 each had significant differences with group 1 (P < .05). No significant difference in ICP occurred between groups 3 and 4 (P > .05). There were significant histologic and ultrastructural alterations in tunical tissues from group 2; however, these changes were markedly less in group 5 in terms of lower levels of fibrotic changes, elastosis, and superior overall neuroendothelial expression. Groups 3 and 4 showed improved structural and ultrastructural parameters when compared with group 2. Group 5 demonstrated lower levels of transforming growth factor β1 and basic fibroblast growth factor expression. Clinical Implications This experimental model may encourage administration of ADSCs to prevent the deleterious effects of trauma to the TA. Strengths and Limitations Injecting ADSCs can improve the healing process and erectile dysfunction in a rat model following TA injury, and combining ADSC injection with surgical suturing resulted in superior outcomes. The main limitation was the absence of long-term ICP measurements and a longer follow-up period that may provide further insight into the chronic phase of the healing process. Conclusion ADSC injection may prevent structural, ultrastructural, functional, and molecular alterations in surgically induced trauma of the rat's TA and enhance the effect of tunical suturing after trauma.
Collapse
Affiliation(s)
| | - Mona F Mansour
- Department of Physiology, Suez Canal University, Ismailia 4111, Egypt
| | - Gamal A Elatrash
- Department of Urology, Suez Canal University, Ismailia 4111, Egypt
| | - Ezzat A Ismail
- Department of Urology, Suez Canal University, Ismailia 4111, Egypt
| | - David Ralph
- Institute of Urology, University College of London Hospital, London W1G 8PH, United Kingdom
| | - Ahmed I El-Sakka
- Department of Urology, Suez Canal University, Ismailia 4111, Egypt
| |
Collapse
|
9
|
Yang H, Chen J, Li J. Isolation, culture, and delivery considerations for the use of mesenchymal stem cells in potential therapies for acute liver failure. Front Immunol 2023; 14:1243220. [PMID: 37744328 PMCID: PMC10513107 DOI: 10.3389/fimmu.2023.1243220] [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: 06/20/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Acute liver failure (ALF) is a high-mortality syndrome for which liver transplantation is considered the only effective treatment option. A shortage of donor organs, high costs and surgical complications associated with immune rejection constrain the therapeutic effects of liver transplantation. Recently, mesenchymal stem cell (MSC) therapy was recognized as an alternative strategy for liver transplantation. Bone marrow mesenchymal stem cells (BMSCs) have been used in clinical trials of several liver diseases due to their ease of acquisition, strong proliferation ability, multipotent differentiation, homing to the lesion site, low immunogenicity and anti-inflammatory and antifibrotic effects. In this review, we comprehensively summarized the harvest and culture expansion strategies for BMSCs, the development of animal models of ALF of different aetiologies, the critical mechanisms of BMSC therapy for ALF and the challenge of clinical application.
Collapse
Affiliation(s)
| | | | - Jun Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
10
|
Clemente-Suárez VJ, Martín-Rodríguez A, Redondo-Flórez L, López-Mora C, Yáñez-Sepúlveda R, Tornero-Aguilera JF. New Insights and Potential Therapeutic Interventions in Metabolic Diseases. Int J Mol Sci 2023; 24:10672. [PMID: 37445852 DOI: 10.3390/ijms241310672] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Endocrine homeostasis and metabolic diseases have been the subject of extensive research in recent years. The development of new techniques and insights has led to a deeper understanding of the mechanisms underlying these conditions and opened up new avenues for diagnosis and treatment. In this review, we discussed the rise of metabolic diseases, especially in Western countries, the genetical, psychological, and behavioral basis of metabolic diseases, the role of nutrition and physical activity in the development of metabolic diseases, the role of single-cell transcriptomics, gut microbiota, epigenetics, advanced imaging techniques, and cell-based therapies in metabolic diseases. Finally, practical applications derived from this information are made.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | | | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Tajo Street s/n, 28670 Villaviciosa de Odon, Spain
| | - Clara López-Mora
- Facultad de Ciencias Biomédicas y de la Salud, Universidad Europea de Valencia, Pg. de l'Albereda, 7, 46010 València, Spain
| | - Rodrigo Yáñez-Sepúlveda
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2520000, Chile
| | | |
Collapse
|
11
|
Ishiuchi N, Nakashima A, Maeda S, Miura Y, Miyasako K, Sasaki K, Uchiki T, Sasaki A, Nagamatsu S, Nakao N, Nagao M, Masaki T. Comparison of therapeutic effects of mesenchymal stem cells derived from superficial and deep subcutaneous adipose tissues. Stem Cell Res Ther 2023; 14:121. [PMID: 37143086 PMCID: PMC10161523 DOI: 10.1186/s13287-023-03350-3] [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: 09/01/2022] [Accepted: 04/19/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Fibrosis is a common histological feature in the process from chronic organ injury to organ failure. Chronic tissue injury causes inflammatory cell infiltration into the injured tissue. The persistence of this inflammatory cell infiltration leads to fibrosis and organ failure. Adipose-derived mesenchymal stem cells (ASCs) have received much attention as a regenerative therapeutic tool to prevent progression from organ injury to failure. Subcutaneous abdominal adipose tissue is divided into superficial and deep layers by a superficial fascia. Adipose tissue easily collected by liposuction is usually obtained from a deep layer, so ASCs derived from a deep layer are generally used for regenerative medicine. However, no research has been conducted to investigate differences in the therapeutic effects of ASCs from the superficial and deep layers (Sup-ASCs and Deep-ASCs, respectively). Therefore, we compared the therapeutic potencies of Sup-ASCs and Deep-ASCs. METHODS ASCs were isolated from superficial and deep subcutaneous abdominal adipose tissues collected from patients who underwent breast reconstruction. We first compared cell characteristics, such as morphology, cell proliferation, cell surface markers, adipogenic and osteogenic differentiation, cell senescence markers, and expression of coagulation and anticoagulant factors between Sup-ASCs and Deep-ASCs. Furthermore, we compared their ability to promote polarization of M2 macrophages and to inhibit transforming growth factor (TGF)-β/Smad signaling using THP-1 cells and TGF-β1 stimulated HK-2 cells incubated with conditioned media from Sup-ASCs or Deep-ASCs. In in vivo experiments, after renal ischemia-reperfusion injury (IRI) procedure, Sup-ASCs or Deep-ASCs were injected through the abdominal aorta. At 21 days post-injection, the rats were sacrificed and their left kidneys were collected to evaluate fibrosis. Finally, we performed RNA-sequencing analysis of Sup-ASCs and Deep-ASCs. RESULTS Sup-ASCs had greater proliferation and adipogenic differentiation compared with Deep-ASCs, whereas both ASC types had similar morphology, cell surface markers, senescence markers, and expression of coagulation and anticoagulant factors. Conditioned media from Sup-ASCs and Deep-ASCs equally promoted polarization of M2 macrophages and suppressed TGF-β/Smad signaling. Moreover, administration of Sup-ASCs and Deep-ASCs equally ameliorated renal fibrosis induced by IRI in rats. RNA-sequencing analysis revealed no significant difference in the expression of genes involved in anti-inflammatory and anti-fibrotic effects between Sup-ASCs and Deep-ASCs. CONCLUSIONS These results indicate that both Sup-ASCs and Deep-ASCs can be used effectively and safely as an intravascular ASC therapy for organ injury.
Collapse
Affiliation(s)
- Naoki Ishiuchi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
- Center for Cause of Death Investigation Research, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- Department of Forensic Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Satoshi Maeda
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- TWOCELLS Company, Limited, 16-35 Hijiyama-honmachi, Minami-ku, Hiroshima, 732-0816, Japan
| | - Yoshie Miura
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- TWOCELLS Company, Limited, 16-35 Hijiyama-honmachi, Minami-ku, Hiroshima, 732-0816, Japan
| | - Kisho Miyasako
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kensuke Sasaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Toshio Uchiki
- Department of Plastic and Reconstructive Surgery, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ayano Sasaki
- Department of Plastic and Reconstructive Surgery, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shogo Nagamatsu
- Department of Plastic and Reconstructive Surgery, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Naoki Nakao
- Department of Forensic Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Masataka Nagao
- Center for Cause of Death Investigation Research, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- Department of Forensic Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| |
Collapse
|
12
|
Inoue O, Goten C, Hashimuko D, Yamaguchi K, Takeda Y, Nomura A, Ootsuji H, Takashima S, Iino K, Takemura H, Halurkar M, Lim HW, Hwa V, Sanchez-Gurmaches J, Usui S, Takamura M. Single cell transcriptomics identifies adipose tissue CD271+ progenitors for enhanced angiogenesis in limb ischemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.09.527726. [PMID: 36865239 PMCID: PMC9980009 DOI: 10.1101/2023.02.09.527726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Therapeutic angiogenesis using mesenchymal stem/stromal cell grafts have shown modest and controversial effects in preventing amputation for patients with critical limb ischemia. Through single-cell transcriptomic analysis of human tissues, we identified CD271 + progenitors specifically from subcutaneous adipose tissue (AT) as having the most prominent pro-angiogenic gene profile distinct from other stem cell populations. AT-CD271 + progenitors demonstrated robust in vivo angiogenic capacity, over conventional adipose stromal cell grafts, characterized by long-term engraftment, augmented tissue regeneration, and significant recovery of blood flow in a xenograft model of limb ischemia. Mechanistically, the angiogenic capacity of CD271 + progenitors is dependent on functional CD271 and mTOR signaling. Notably, the number and angiogenic capacity of CD271 + progenitors was strikingly reduced in insulin resistant donors. Our study highlights the identification of AT-CD271 + progenitors with in vivo superior efficacy for limb ischemia. Furthermore, we showcase comprehensive single-cell transcriptomics strategies for identification of suitable grafts for cell therapy. HIGHLIGHTS Adipose tissue stromal cells have a distinct angiogenic gene profile among human cell sources. CD271 + progenitors in adipose tissue have a prominent angiogenic gene profile. CD271 + progenitors show superior therapeutic capacities for limb ischemia. CD271 + progenitors are reduced and functionally impaired in insulin resistant donors. GRAPHICAL ABSTRACT
Collapse
|
13
|
Teratani T, Kasahara N, Fujimoto Y, Sakuma Y, Miki A, Goto M, Sata N, Kitayama J. Mesenchymal Stem Cells Secretions Enhanced ATP Generation on Isolated Islets during Transplantation. Islets 2022; 14:69-81. [PMID: 35034568 PMCID: PMC8765074 DOI: 10.1080/19382014.2021.2022423] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The success of islet transplantation in both basic research and clinical settings has proven that cell therapy has the potential to cure diabetes. Islets intended for transplantation are inevitably subjected to damage from a number of sources, including ischemic injury during removal and delivery of the donor pancreas, enzymatic digestion during islet isolation, and reperfusion injury after transplantation in the recipient. Here, we found that protein factors secreted by porcine adipose-tissue mesenchymal stem cells (AT-MSCs) were capable of activating preserved porcine islets. A conditioned medium was prepared from the supernatant obtained by culturing porcine AT-MSCs for 2 days in serum-free medium. Islets were preserved at 4°C in University of Wisconsin solution during transportation and then incubated at 37°C in RPMI-1620 medium with fractions of various molecular weights prepared from the conditioned medium. After treatment with certain fractions of the AT-MSC secretions, the intracellular ATP levels of the activated islets had increased to over 160% of their initial values after 4 days of incubation. Our novel system may be able to restore the condition of isolated islets after transportation or preservation and may help to improve the long-term outcome of islet transplantation.Abbreviations: AT-MSC, adipose-tissue mesenchymal stem cell; Cas-3, caspase-3; DAPI, 4,6-diamidino-2-phenylindole; DTZ, dithizone; ES cell, embryonic stem cell; FITC, fluorescein isothiocyanate; IEQ, islet equivalent; INS, insulin; iPS cell, induced pluripotent stem cell; Luc-Tg rat, luciferase-transgenic rat; PCNA, proliferating cell nuclear antigen; PDX1, pancreatic and duodenal homeobox protein-1; UW, University of Wisconsin; ZO1, zona occludens 1.
Collapse
Affiliation(s)
- Takumi Teratani
- Division of Translational Research, Jichi Medical University, Tochigi, Japan
- Department of Surgery, Jichi Medical University, Tochigi, Japan
- CONTACT Takumi Teratani Division of Clinical Investigation, Jichi Medical University, 3311-1, Yakushiji, Shimotsukeshi, Tochigi329-0498, Japan
| | - Naoya Kasahara
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | | | - Yasunaru Sakuma
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Atsushi Miki
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Masafumi Goto
- New Industry Creation Hatchery Center, Tohoku University, Miyagi, Japan
| | - Naohiro Sata
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| | - Joji Kitayama
- Division of Translational Research, Jichi Medical University, Tochigi, Japan
- Department of Surgery, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
14
|
Chen YC, Fu YS, Tsai SW, Wu PK, Chen CM, Chen WM, Chen CF. IL-1b in the Secretomes of MSCs Seeded on Human Decellularized Allogeneic Bone Promotes Angiogenesis. Int J Mol Sci 2022; 23:15301. [PMID: 36499629 PMCID: PMC9737155 DOI: 10.3390/ijms232315301] [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: 10/12/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis plays an important role in the development of bone and bone regeneration to provide the required molecules. Mesenchymal stem cells (MSCs) are pluripotent, self-renewing, and spindle-shaped cells, which can differentiate into multiple lineages such as chondrocytes, osteocytes, and adipocytes. MSCs derived from bone marrow (BMMSCs), adipose tissue (ADMSCs), and Wharton's jelly (UCMSCs) are popular in the field of tissue regeneration. MSCs have been proposed that can promote bone regeneration by enhancing vascularization. In this study, the angiogenic potential of secretomes of undifferentiated and osteo-differentiated BMMSCs, ADMSCs, and UCMSCs seeded on human decellularized allogeneic bone were compared. Human umbilical vein endothelial cells (HUVECs) were treated with MSC secretomes. Cell growth, cell migration, and angiogenesis of HUVECs were analyzed by MTT, wound healing, and tube formation assays. Angiogenic gene expression levels of MSCs were evaluated using real-time quantitative PCR. Antibody neutralization was performed to validate the candidate target. Our study demonstrates that the angiogenic gene expression profile is tissue-dependent and the angiogenic ability of secretomes is independent of the state of differentiation. We also explore that IL-1b is important for MSC angiogenic potential. Taken together, this study proves that IL-1b in the secretomes plays a vital role in angiogenesis.
Collapse
Affiliation(s)
- Yi-Chun Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yu-Show Fu
- Department of Anatomy and Cell Biology, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Shang-Wen Tsai
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Orthopaedics, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Po-Kuei Wu
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Orthopaedics, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chao-Ming Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Orthopaedics, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Wei-Ming Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Orthopaedics, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Cheng-Fong Chen
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Orthopaedics, School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
15
|
Shah JS, Macaitis J, Lundquist B, Johnstone B, Coleman M, Jefferson MA, Glaser J, Rodriguez AR, Cardin S, Wang HC, Burdette A. Evaluating Thera-101 as a Low-Volume Resuscitation Fluid in a Model of Polytrauma. Int J Mol Sci 2022; 23:ijms232012664. [PMID: 36293520 PMCID: PMC9604349 DOI: 10.3390/ijms232012664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Traumatic brain injury (TBI) and hemorrhage remain challenging to treat in austere conditions. Developing a therapeutic to mitigate the associated pathophysiology is critical to meet this treatment gap, especially as these injuries and associated high mortality are possibly preventable. Here, Thera-101 (T-101) was evaluated as low-volume resuscitative fluid in a rat model of TBI and hemorrhage. The therapeutic, T-101, is uniquely situated as a TBI and hemorrhage intervention. It contains a cocktail of proteins and microvesicles from the secretome of adipose-derived mesenchymal stromal cells that can act on repair and regenerative mechanisms associated with poly-trauma. T-101 efficacy was determined at 4, 24, 48, and 72 h post-injury by evaluating blood chemistry, inflammatory chemo/cytokines, histology, and diffusion tensor imaging. Blood chemistry indicated that T-101 reduced the markers of liver damage to Sham levels while the levels remained elevated with the control (saline) resuscitative fluid. Histology supports the potential protective effects of T-101 on the kidneys. Diffusion tensor imaging showed that the injury caused the most damage to the corpus callosum and the fimbria. Immunohistochemistry suggests that T-101 may mitigate astrocyte activation at 72 h. Together, these data suggest that T-101 may serve as a potential field deployable low-volume resuscitation therapeutic.
Collapse
Affiliation(s)
- Jessica Stukel Shah
- Naval Medical Research Unit San Antonio, Fort Sam Houston, San Antonio, TX 78234, USA
| | - Joseph Macaitis
- Naval Medical Research Unit San Antonio, Fort Sam Houston, San Antonio, TX 78234, USA
| | - Bridney Lundquist
- Naval Medical Research Unit San Antonio, Fort Sam Houston, San Antonio, TX 78234, USA
| | | | | | - Michelle A. Jefferson
- Air Force Research Laboratory, 711th Human Performance Wing, Airman Systems Directorate, Bioeffects Division, Veterinary Science Branch, San Antonio, TX 78234, USA
| | - Jacob Glaser
- Naval Medical Research Unit San Antonio, Fort Sam Houston, San Antonio, TX 78234, USA
| | - Annette R. Rodriguez
- Naval Medical Research Unit San Antonio, Fort Sam Houston, San Antonio, TX 78234, USA
| | - Sylvain Cardin
- Naval Medical Research Unit San Antonio, Fort Sam Houston, San Antonio, TX 78234, USA
| | - Heuy-Ching Wang
- Naval Medical Research Unit San Antonio, Fort Sam Houston, San Antonio, TX 78234, USA
- Correspondence: (H.-C.W.); (A.B.); Tel.: +1-210-539-7017 (H.-C.W.); +1-210-325-2668 (A.B.)
| | - Alexander Burdette
- Naval Medical Research Unit San Antonio, Fort Sam Houston, San Antonio, TX 78234, USA
- Correspondence: (H.-C.W.); (A.B.); Tel.: +1-210-539-7017 (H.-C.W.); +1-210-325-2668 (A.B.)
| |
Collapse
|
16
|
CHEN Z, RUAN B, LONG G, LIN W. Adipose tissue-derived mesenchymal stem cells attenuate lung inflammation and fibrosis in the bleomycin-induced pulmonary fibrosis rat model via caveolin-1/NF-kB signaling axis. Physiol Res 2022; 71:657-666. [PMID: 36047729 PMCID: PMC9841806 DOI: 10.33549/physiolres.934892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Stem cells have emerged as promising therapeutic options for several human diseases, including pulmonary fibrosis (PF). In this study, we investigated the therapeutic effects of adipose tissue-derived mesenchymal stem cells (ADMSCs) in the bleomycin-induced PF model rats and the underlying mechanisms. The PF model rats were generated by intratracheal injections of 5 mg/kg bleomycin sulfate. The ADMSC group rats were generated by injecting 2×10(6) ADMSCs via the tail vein at 0, 12, and 24 h after bleomycin injection. The control, PF, and ADMSC group rats were sacrificed on day 21 after bleomycin injections and the changes in lung histology and the levels of pro-inflammatory cytokines, collagen I, and caveolin-1 (Cav-1), and the activity of the NF-kappaB signaling pathway in the lung tissues was assessed by hematoxylin-eosin staining, ELISA, and western blotting assays. The lung tissues of the PF model rats showed significant infiltration of neutrophils, tissue destruction, and collagen deposition, but these effects were abrogated by the ADMSCs. The levels of pro-inflammatory cytokines such as IL-6, IL-1beta, and TGF-beta1 were elevated in the lung tissues and the bronchoalveolar lavage fluid (BALF) of the bleomycin-induced PF model rats, but these effects were reversed by the ADMSCs. The lung tissues of the PF model rats showed significant downregulation of Cav-1 and significantly higher activation of the pro-inflammatory NF-kappaB pathway. However, administration of the ADMSCs restored the expression levels of Cav-1 and suppressed the NF-kappaB signaling pathway in the lungs of the bleomycin-induced PF model rats. In conclusion, this study demonstrated that the ADMSCs protected against bleomycin-induced PF in the rat model by modulating the Cav-1/NF-kappaB axis.
Collapse
Affiliation(s)
- Zhe CHEN
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Wenling, Zhejiang, China
| | - Bingqing RUAN
- Department of Internal Medicine, Wenling Women’s and Children’s Hospital, Zhejiang, China
| | - Guangyan LONG
- Department of Infectious Diseases, The First People’s Hospital of Wenling, Zhejiang, China
| | - Wei LIN
- Department of Respirology, The First People’s Hospital of Wenling, Zhejiang, China
| |
Collapse
|
17
|
Hernandez JC, Yeh DW, Marh J, Choi HY, Kim J, Chopra S, Ding L, Thornton M, Grubbs B, Makowka L, Sher L, Machida K. Activated and nonactivated MSCs increase survival in humanized mice after acute liver injury through alcohol binging. Hepatol Commun 2022; 6:1549-1560. [PMID: 35246968 PMCID: PMC9234635 DOI: 10.1002/hep4.1924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 01/15/2022] [Accepted: 01/31/2022] [Indexed: 11/08/2022] Open
Abstract
The ability of the liver to regenerate after injury makes it an ideal organ to study for potential therapeutic interventions. Mesenchymal stem cells (MSCs) possess self-renewal and differentiation properties, as well as anti-inflammatory properties that make them an ideal candidate for therapy of acute liver injury. The primary aim of this study is to evaluate the potential for reversal of hepatic injury using human umbilical cord-derived MSCs. Secondary aims include comparison of various methods of administration as well as comparison of activated versus nonactivated human umbilical cord stem cells. To induce liver injury, humanized mice were fed high-cholesterol high-fat liquid diet with alcohol binge drinking. Mice were then treated with either umbilical cord MSCs, activated umbilical cord MSCs, or a placebo and followed for survival. Blood samples were obtained at the end of the binge drinking and at the time of death to measure alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. Histology of all mouse livers was reported at time of death. Activated MSCs that were injected intravenously, intraperitoneally, or both routes had superior survival compared with nonactivated MSCs and with placebo-treated mice. AST and ALT levels were elevated in all mice before treatment and improved in the mice treated with stem cells. Conclusion: Activated stem cells resulted in marked improvement in survival and in recovery of hepatic chemistries. Activated umbilical cord MSCs should be considered an important area of investigation in acute liver injury.
Collapse
Affiliation(s)
- Juan Carlos Hernandez
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Da-Wei Yeh
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Joel Marh
- PrimeGenUS Inc.Santa AnaCaliforniaUSA
| | - Hye Yeon Choi
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Julia Kim
- PrimeGenUS Inc.Santa AnaCaliforniaUSA
| | - Shefali Chopra
- Department of PathologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Li Ding
- Department of Population and PublicHealth Sciences University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Matthew Thornton
- Department of SurgeryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA.,Childrens Hospital Los AngelesLos AngelesCaliforniaUSA
| | - Brendan Grubbs
- Department of SurgeryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA.,Childrens Hospital Los AngelesLos AngelesCaliforniaUSA
| | | | - Linda Sher
- PrimeGenUS Inc.Santa AnaCaliforniaUSA.,Department of SurgeryUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Keigo Machida
- Departments of Molecular Microbiology and ImmunologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA.,Southern California Research Center for ALPD and CirrhosisLos AngelesCaliforniaUSA
| |
Collapse
|
18
|
Yan S, Zhang C, Ji X, Wu G, Huang X, Zhang Y, Zhang Y. MSC-ACE2 Ameliorates Streptococcus uberis-Induced Inflammatory Injury in Mammary Epithelial Cells by Upregulating the IL-10/STAT3/SOCS3 Pathway. Front Immunol 2022; 13:870780. [PMID: 35677060 PMCID: PMC9167935 DOI: 10.3389/fimmu.2022.870780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
In the dairy industry, Streptococcus uberis (S. uberis) is one of the most important pathogenic bacteria associated with mastitis in milk-producing cows, causing vast economic loss. To date, the only real effective method of treating and preventing streptococcal mastitis is antimicrobial therapy. In many inflammatory diseases, mesenchymal stem cells (MSCs) and angiotensin-converting enzyme 2 (ACE2) play an anti-inflammatory and anti-injurious role. Accordingly, we hypothesized that MSCs overexpressing ACE2 (MSC-ACE2) would ameliorate the inflammatory injury caused by S. uberis in mammary epithelial cells more efficiently than MSC alone. By activating the transcription 3/suppressor of cytokine signaling 3 (IL-10/STAT3/SOCS3) signaling pathway, MSC-ACE2 inhibited the NF-κB, MAPKs, apoptosis, and pyroptosis passways. Moreover, MSC-ACE2 overturned the downregulation of Occludin, Zonula occludens 1 (ZO-1), and Claudin-3 expression levels caused by S. uberis, suggesting that MSC-ACE2 promotes the repair of the blood-milk barrier. MSC-ACE2 demonstrated greater effectiveness than MSC alone, as expected. Based on these results, MSC-ACE2 effectively inhibits EpH4-Ev cell's inflammatory responses induced by S. uberis, and would be an effective therapeutic tool for treating streptococcal mastitis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuanshu Zhang
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
19
|
Robinson AM, Stavely R, Miller S, Eri R, Nurgali K. Mesenchymal stem cell treatment for enteric neuropathy in the Winnie mouse model of spontaneous chronic colitis. Cell Tissue Res 2022; 389:41-70. [PMID: 35536444 DOI: 10.1007/s00441-022-03633-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 04/26/2022] [Indexed: 11/30/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic gut inflammation with periods of acute flares and remission. Beneficial effects of a single dose of mesenchymal stem cell (MSC)-based treatment have been demonstrated in acute models of colitis. No studies investigated therapeutic effects of MSCs for the attenuation of enteric neuropathy in a chronic model of colitis. The short and long-term effects of MSC treatment in modulating inflammation and damage to the enteric nervous system (ENS) were studied in the Winnie mouse model of spontaneous chronic colitis highly representative of human IBD. Winnie mice received a single dose of either 1 × 106 human bone marrow-derived MSCs or 100µL PBS by intracolonic enema. C57BL/6 mice received 100µL PBS. Colon tissues were collected at 3 and 60 days post MSC administration to evaluate the short-term and long-term effects of MSCs on inflammation and enteric neuropathy by histological and immunohistochemical analyses. In a separate set of experiments, multiple treatments with 4 × 106 and 2 × 106 MSCs were performed and tissue collected at 3 days post treatment. Chronic intestinal inflammation in Winnie mice was associated with persistent diarrhea, perianal bleeding, morphological changes, and immune cell infiltration in the colon. Significant changes to the ENS, including impairment of cholinergic, noradrenergic and sensory innervation, and myenteric neuronal loss were prominent in Winnie mice. Treatment with a single dose of bone marrow-derived MSCs was ineffective in attenuating chronic inflammation and enteric neuropathy in Winnie.
Collapse
Affiliation(s)
- Ainsley M Robinson
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia
| | - Rhian Stavely
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia.,Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Sarah Miller
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia
| | - Rajaraman Eri
- University of Tasmania, School of Health Sciences, Launceston, TAS, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia. .,Department of Medicine Western Health, The University of Melbourne, Melbourne, VIC, Australia. .,Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia.
| |
Collapse
|
20
|
Mikłosz A, Nikitiuk BE, Chabowski A. Using adipose-derived mesenchymal stem cells to fight the metabolic complications of obesity: Where do we stand? Obes Rev 2022; 23:e13413. [PMID: 34985174 PMCID: PMC9285813 DOI: 10.1111/obr.13413] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 12/15/2022]
Abstract
Obesity is a critical risk factor for the development of metabolic diseases, and its prevalence is increasing worldwide. Stem cell-based therapies have become a promising tool for therapeutic intervention. Among them are adipose-derived mesenchymal stem cells (ADMSCs), secreting numerous bioactive molecules, like growth factors, cytokines, and chemokines. Their unique features, including immunosuppressive and immunomodulatory properties, make them an ideal candidates for clinical applications. Numerous experimental studies have shown that ADMSCs can improve pancreatic islet cell viability and function, ameliorate hyperglycemia, improve insulin sensitivity, restore liver function, counteract dyslipidemia, lower pro-inflammatory cytokines, and reduce oxidative stress in the animal models. These results prompted scientists to use ADMSCs clinically. However, up to date, there have been few clinical studies or ongoing trails using ADMSCs to treat metabolic disorders such as type 2 diabetes mellitus (T2DM) or liver cirrhosis. Most human studies have implemented autologous ADMSCs with minimal risk of cellular rejection. Because the functionality of ADMSCs is significantly reduced in subjects with obesity and/or metabolic syndrome, their efficacy is questioned. ADMSCs transplantation may offer a potential therapeutic approach for the treatment of metabolic complications of obesity, but randomized controlled trials are required to establish their safety and efficacy in humans prior to routine clinical use.
Collapse
Affiliation(s)
- Agnieszka Mikłosz
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | | | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
21
|
Patel A, Aslam R, Jamil M, Ansari A, Khan S. The Effects of Growth Factors and Cytokines on Hepatic Regeneration: A Systematic Review. Cureus 2022; 14:e24539. [PMID: 35651436 PMCID: PMC9138487 DOI: 10.7759/cureus.24539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2022] [Indexed: 01/10/2023] Open
Abstract
The incidence of liver disease increases throughout the years due to many lifestyle factors; thus, the only definite treatment available for chronic liver disease is a liver transplant. However, the liver has a natural ability to repair itself and regenerate its hepatic tissue from stem cells. It is hypothesized that by inducing the liver with specific growth factors and cytokines such as interleukin 6 (IL-6) compared to general growth factors like growth differentiation factor 15 (GDF-15), it can regenerate, decreasing the need for liver transplant procedures. MEDLINE, the Journal of Hepatology, and Google Scholar were used to find articles. Various studies, including epidemiological studies dated from the year 2000 and greater, were used for the introduction. The results used only randomized control trials, experimental studies, and primary articles published since 2000. This compared the results of manipulating variables to determine the effects of hepatic regeneration by either specific hepatocyte growth factors or general growth factors like GDF-15. A total of 10 collected studies showed increased levels of gene expression and function, improved gross morphology, and histological appearance of the liver when induced by cytokines and specific growth factors versus general growth factors. Overall, the hypothesis was proven. The effects of GDF-15 were not significant compared to the effects of hepatocyte-specific growth factors and cytokines like IL-6 because they have two different effects on the liver after liver injury. Future studies should investigate this topic on the human hepatic regenerative ability, plus compare the effects of general growth factors like GDF-15 and specific hepatocyte growth factors and cytokines such as IL-6 in human liver tissue.
Collapse
|
22
|
Zhang L, Ma XJN, Fei YY, Han HT, Xu J, Cheng L, Li X. Stem cell therapy in liver regeneration: Focus on mesenchymal stem cells and induced pluripotent stem cells. Pharmacol Ther 2022; 232:108004. [PMID: 34597754 DOI: 10.1016/j.pharmthera.2021.108004] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/11/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023]
Abstract
The liver has the ability to repair itself after injury; however, a variety of pathological changes in the liver can affect its ability to regenerate, and this could lead to liver failure. Mesenchymal stem cells (MSCs) are considered a good source of cells for regenerative medicine, as they regulate liver regeneration through different mechanisms, and their efficacy has been demonstrated by many animal experiments and clinical studies. Induced pluripotent stem cells, another good source of MSCs, have also made great progress in the establishment of organoids, such as liver disease models, and in drug screening. Owing to the recent developments in MSCs and induced pluripotent stem cells, combined with emerging technologies including graphene, nano-biomaterials, and gene editing, precision medicine and individualized clinical treatment may be realized in the near future.
Collapse
Affiliation(s)
- Lu Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Xiao-Jing-Nan Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Yuan-Yuan Fei
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Heng-Tong Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Jun Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Lu Cheng
- Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Xun Li
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
23
|
Wang P, Cui Y, Wang J, Liu D, Tian Y, Liu K, Wang X, Liu L, He Y, Pei Y, Li L, Sun L, Zhu Z, Chang D, Jia J, You H. Mesenchymal stem cells protect against acetaminophen hepatotoxicity by secreting regenerative cytokine hepatocyte growth factor. Stem Cell Res Ther 2022; 13:94. [PMID: 35246254 PMCID: PMC8895877 DOI: 10.1186/s13287-022-02754-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/26/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Acetaminophen (APAP) overdose is a major cause of the morbidity of acute liver failure. The current clinically approved treatment for APAP poisoning, N-acetylcysteine (NAC), has a limited therapeutic window, and prolonged treatment with NAC delays liver regeneration. Mesenchymal stem cells (MSCs) also have therapeutic effects on APAP-induced mouse liver failure, but whether the effects are comparable to those of NAC has not been determined, and the mechanism still needs further exploration. METHODS Fasted C57BL/6 mice that received 500 mg/kg APAP were treated intravenously with 300 mg/kg NAC or different amounts of MSCs at 2 h after APAP to investigate survival, hepatocyte necrosis and neutrophil/macrophage recruitment. In vitro co-culture was performed to study the anti-necrotic effects of MSCs on the APAP-injured hepatocyte cell line L-O2. RESULTS MSCs dose-dependently rescued the C57BL/6J mice from APAP-induced liver failure, with 87.5% of MSCs (1 × 106) surviving similar to that of NAC (90%). MSC has similar effects on reduced hepatocyte necrosis and granulocytic myeloid-derived suppressor cells (MDSC) infiltration but enhanced the proportion of regenerative monocytic MDSC when compared to NAC. Mechanistically, MSCs attenuate hepatocyte necrosis by secreting hepatocyte growth factor (HGF). When HGF was knocked down, the protective effects of MSCs were reduced on APAP-induced hepatocyte necrosis and mouse liver failure. CONCLUSIONS MSCs are comparable to NAC against APAP-induced liver failure by secreting HGF with less regenerative retardation concerns, thus facilitating the application of MSCs in clinical therapy for APAP liver failure.
Collapse
Affiliation(s)
- Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis and National Clinical Research Center for Digestive Diseases, No. 95 Yong-An Road, Beijing, 100050, China
| | - Yan Cui
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis and National Clinical Research Center for Digestive Diseases, No. 95 Yong-An Road, Beijing, 100050, China
| | - Jing Wang
- BOE Regenerative Medicine Technology Co., Ltd., Beijing, 100015, China
| | - Donghua Liu
- BOE Regenerative Medicine Technology Co., Ltd., Beijing, 100015, China
| | - Yue Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation and National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Kai Liu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation and National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Xue Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis and National Clinical Research Center for Digestive Diseases, No. 95 Yong-An Road, Beijing, 100050, China
| | - Lin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis and National Clinical Research Center for Digestive Diseases, No. 95 Yong-An Road, Beijing, 100050, China
| | - Yu He
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis and National Clinical Research Center for Digestive Diseases, No. 95 Yong-An Road, Beijing, 100050, China
| | - Yufeng Pei
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis and National Clinical Research Center for Digestive Diseases, No. 95 Yong-An Road, Beijing, 100050, China
| | - Li Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis and National Clinical Research Center for Digestive Diseases, No. 95 Yong-An Road, Beijing, 100050, China
| | - Liying Sun
- Division of Liver Transplantation Surgery, Department of Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Zhijun Zhu
- Division of Liver Transplantation Surgery, Department of Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Dehua Chang
- Department of Cell Therapy in Regenerative Medicine, University of Tokyo Hospital, Tokyo, 113-8655, Japan.
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis and National Clinical Research Center for Digestive Diseases, No. 95 Yong-An Road, Beijing, 100050, China. .,Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation and National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis and National Clinical Research Center for Digestive Diseases, No. 95 Yong-An Road, Beijing, 100050, China. .,Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation and National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| |
Collapse
|
24
|
Khalighi S, Saadatmand M. Bioprinting a thick and cell-laden partially oxidized alginate-gelatin scaffold with embedded micro-channels as future soft tissue platform. Int J Biol Macromol 2021; 193:2153-2164. [PMID: 34800519 DOI: 10.1016/j.ijbiomac.2021.11.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 11/24/2022]
Abstract
Despite all the advancements in tissue engineering, one of the unsolved challenges is the mass transfer limitation. Therefore, the subject of pre-vascularization in the engineered tissues gets more attention to avoid necrotic core formation. In this study, we considered a design for interconnected channels with a muscle tissue-like structure, in silico and in vitro. A sequence of simple steps make it possible for us to use the same material, gelatin, as both a sacrificial material and one of the main components of the scaffold simultaneously. We defined a new approach to quantify the repeatability of a new combination of hydrogels (Partially Oxidized Alginate + Gelatin) for extrusion-based bioprinting. Additionally, the mechanical properties, hydrogel porosity, degradation time, and swelling ratio were also evaluated. Based on all these test results, the scaffold with the optimum properties was chosen for the bioprinting of adipose derived mesenchymal stem cells (ADMSCs) in the scaffolds with and without the channels. This bioprinted scaffold with microchannels showed promising mimicry of the microenvironment, leading to higher survival and proliferation rates of the cells by up to 250%. Based on these results, it has the potential to serve as a platform for further research in vascularization, healthy/disease modelling, and stem cell differentiation.
Collapse
Affiliation(s)
- Sadaf Khalighi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Maryam Saadatmand
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
25
|
Secretome Analysis of Rabbit and Human Mesenchymal Stem and Endothelial Progenitor Cells: A Comparative Study. Int J Mol Sci 2021; 22:ijms222212283. [PMID: 34830165 PMCID: PMC8625496 DOI: 10.3390/ijms222212283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022] Open
Abstract
Human adipose tissue-derived mesenchymal stem cells (AT-MSCs) have been studied several years for their immunomodulatory effect through the paracrine mechanism and cytokine secretion. In combination with endothelial progenitor cells (EPCs), MSCs have great therapeutical potential for the repair of endothelium and wound healing. However, little is known about the cytokine profile of rabbit AT-MSCs or even EPCs. The aim of this study was to analyze the secretomes of these rabbit stem/progenitor cells. A large-scale human cytokine array (up to 80 cytokines) was used to identify and compare cytokines secreted into conditioned media of human and rabbit AT-MSCs as well as HUVECs and rabbit EPCs. Few cytokines were highly expressed by human AT-MSCs (TIMP-2, TIMP-1), HUVECs (MCP-1, TIMP-2, GRO, Angiogenin, IL-8, TIMP-1), or by rabbit EPCs (TIMP-2). Several cytokines have moderate expression by human (MCP-1, GRO, Angiogenin, TGF-β 2, IL-8, LIF, IL-6, Osteopontin, Osteoprotegerin) and rabbit AT-MSCs (TIMP-2, TGF-β 2, LIF, Osteopontin, IL-8, IL-5, IL-3) or by HUVECs (IL-6, MIF, TGF-β 2, GCP-2, IGFBP-2, Osteoprotegerin, EGF, LIF, PDGF-BB, MCP-3, Osteopontin, Leptin, IL-5, ENA-78, TNF-β) and rabbit EPCs (TGF-β 2, Osteopontin, GRO, LIF, IL-8, IL-5, IL-3). In conclusion, the proposed method seems to be useful for the secretome analysis of rabbit stem/progenitor cells.
Collapse
|
26
|
Łach K, Cebulski J, Chaber R, Kocan B, Wojnarowska-Nowak R, Banaś-Ząbczyk A. First Identification of the Effects of Low Frequency Electromagnetic Field on the Micromolecular Changes in Adipose Tissue-Derived Mesenchymal Stem Cells by Fourier Transform Infrared Spectroscopy. J Med Phys 2021; 46:253-262. [PMID: 35261495 PMCID: PMC8853457 DOI: 10.4103/jmp.jmp_57_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose: In this study, we hypothesize that exposure of adipose tissue-mesenchymal stem cells (AT-MSCs) to electromagnetic field (EMF) may impact adipose stem cells' micromolecular structure (analyzed using Fourier transform infrared spectroscopy [FTIR]). Materials and Methods: The AT-MSCs were exposed to continuous vertically applied sinusoidal EMF with a frequency of 50 Hz and a flux density of 1.5 mT for 24, 48, and 72 h. After an appropriate time (24, 48, 72 h) cells were washed with PBS, scrubbed, and immediately taken into FTIR analyses. Results: EMFs affect AT-MSCs. The greatest differences were in the range of nucleic acids and proteins in the fingerprint region which occurred after 24 and 48 h of EMF exposure. However, in the case of 72 h of EMF exposure, no significant differences were noticed in the FTIR spectra towards the control. Conclusions: FTIR spectra show differences between samples under the influence of EMF before they will be manifested at the morphological level. The largest differences in the range of nucleic acids and proteins in the fingerprint region occurred at 24 and 48 h of EMF exposure. That means it was during the first 48 h after EMF exposure a great number of dynamic changes occurred. However, in the case of AT-MSCs in 72 h EMF and 72 h control, no significant differences were noted in the FTIR spectra, which means that the chemical composition in these two cases is similar. EMF is not neutral for stem cells, especially in the in the first hours of interaction (24 h, 48 h).
Collapse
Affiliation(s)
- Kornelia Łach
- Department of Pediatrics, Institute of Medical Sciences, Medical College of Rzeszow University, University of Rzeszow, Warzywna 1A, Rzeszow, Poland
| | - Józef Cebulski
- Institute of Physics, College of Natural Sciences, University of Rzeszow, Pigonia 1, Rzeszow, Poland
| | - Radosław Chaber
- Department of Pediatrics, Institute of Medical Sciences, Medical College of Rzeszow University, University of Rzeszow, Warzywna 1A, Rzeszow, Poland
| | - Beata Kocan
- Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, University of Rzeszow, Warzywna 1A, 35-310 Rzeszow, Poland
| | - Renata Wojnarowska-Nowak
- Institute of Material Engineering, College of Natural Sciences, University of Rzeszow, Pigonia 1
| | - Agnieszka Banaś-Ząbczyk
- Department of Biology, Institute of Medical Sciences, Medical College of Rzeszow University, Al. Mjr. W. Kopisto 2a, 35-959 Rzeszow, Poland
| |
Collapse
|
27
|
Therapeutic approach of adipose-derived mesenchymal stem cells in refractory peptic ulcer. Stem Cell Res Ther 2021; 12:515. [PMID: 34565461 PMCID: PMC8474857 DOI: 10.1186/s13287-021-02584-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/04/2021] [Indexed: 12/14/2022] Open
Abstract
Peptic ulcer is one of the most common gastrointestinal tract disorders worldwide, associated with challenges such as refractory morbidity, bleeding, interference with use of anticoagulants, and potential side effects associated with long-term use of proton pump inhibitors. A peptic ulcer is a defect in gastric or duodenal mucosa extending from muscularis mucosa to deeper layers of the stomach wall. In most cases, ulcers respond to standard treatments. However, in some people, peptic ulcer becomes resistant to conventional treatment or recurs after initially successful therapy. Therefore, new and safe treatments, including the use of stem cells, are highly favored for these patients. Adipose-derived mesenchymal stem cells are readily available in large quantities with minimal invasive intervention, and isolation of adipose-derived mesenchymal stromal stem cells (ASC) produces large amounts of stem cells, which are essential for cell-based and restorative therapies. These cells have high flexibility and can differentiate into several types of cells in vitro. This article will investigate the effects and possible mechanisms and signaling pathways of adipose tissue-derived mesenchymal stem cells in patients with refractory peptic ulcers.
Collapse
|
28
|
Shang Z, Jiang Y, Guan X, Wang A, Ma B. Therapeutic Effects of Stem Cells From Different Source on Renal Ischemia- Reperfusion Injury: A Systematic Review and Network Meta-analysis of Animal Studies. Front Pharmacol 2021; 12:713059. [PMID: 34539400 PMCID: PMC8444551 DOI: 10.3389/fphar.2021.713059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/17/2021] [Indexed: 12/20/2022] Open
Abstract
Objective: Although stem cell therapy for renal ischemia-reperfusion injury (RIRI) has made immense progress in animal studies, conflicting results have been reported by the investigators. Therefore, we aimed to systematically evaluate the effects of different stem cells on renal function of animals with ischemia-reperfusion injury and to compare the efficacies of stem cells from various sources. Methods: PubMed, Web of Science, Embase, Cochrane, CNKI, VIP, CBM, and WanFang Data were searched for records until April 2021. Two researchers independently conducted literature screening, data extraction, and literature quality evaluation. Results and conclusion: Seventy-two animal studies were included for data analysis. Different stem cells significantly reduced serum creatinine and blood urea nitrogen levels in the early and middle stages (1 and 7 days) compared to the negative control group, however there was no significant difference in the late stage among all groups (14 days); In the early stage (1 day), the renal histopathological score in the stem cell group was significantly lower than that in the negative control group, and there was no significant difference among these stem cells. In addition, there was no significant difference between stem cell and negative control in proliferation of resident cells, however, significantly less apoptosis of resident cells than negative control. In conclusion, the results showed that stem cells from diverse sources could improve the renal function of RIRI animals. ADMSCs and MDMSCs were the most-researched stem cells, and they possibly hold the highest therapeutic potential. However, the quality of evidence included in this study is low, and there are many risks of bias. The exact efficacy of the stem cells and the requirement for further clinical studies remain unclear.
Collapse
Affiliation(s)
- Zhizhong Shang
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.,The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yanbiao Jiang
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.,The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xin Guan
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Anan Wang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Bin Ma
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| |
Collapse
|
29
|
Prządka P, Buczak K, Frejlich E, Gąsior L, Suliga K, Kiełbowicz Z. The Role of Mesenchymal Stem Cells (MSCs) in Veterinary Medicine and Their Use in Musculoskeletal Disorders. Biomolecules 2021; 11:1141. [PMID: 34439807 PMCID: PMC8391453 DOI: 10.3390/biom11081141] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
Regenerative medicine is a dynamically developing field of human and veterinary medicine. The animal model was most commonly used for mesenchymal stem cells (MSCs) treatment in experimental and preclinical studies with a satisfactory therapeutic effect. Year by year, the need for alternative treatments in veterinary medicine is increasing, and other applications for promising MSCs and their biological derivatives are constantly being sought. There is also an increase in demand for other methods of treating disease states, of which the classical treatment methods did not bring the desired results. Cell therapy can be a realistic option for treating human and animal diseases in the near future and therefore additional research is needed to optimize cell origins, numbers, or application methods in order to standardize the treatment process and assess its effects. The aim of the following work was to summarize available knowledge about stem cells in veterinary medicine and their possible application in the treatment of chosen musculoskeletal disorders in dogs and horses.
Collapse
Affiliation(s)
- Przemysław Prządka
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Science, Pl. Grunwadzki 51, 50-366 Wroclaw, Poland; (K.B.); (Z.K.)
| | - Krzysztof Buczak
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Science, Pl. Grunwadzki 51, 50-366 Wroclaw, Poland; (K.B.); (Z.K.)
| | - Ewelina Frejlich
- 2nd Department of General Surgery and Surgical Oncology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Ludwika Gąsior
- Vets & Pets Veterinary Clinic, Zakladowa 11N, 50-231 Wroclaw, Poland;
| | - Kamil Suliga
- Student Veterinary Surgical Society “LANCET”, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Science, Pl. Grunwaldzki 51, 50-366 Wroclaw, Poland;
| | - Zdzisław Kiełbowicz
- Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Science, Pl. Grunwadzki 51, 50-366 Wroclaw, Poland; (K.B.); (Z.K.)
| |
Collapse
|
30
|
He X, Hong W, Yang J, Lei H, Lu T, He C, Bi Z, Pan X, Liu Y, Dai L, Wang W, Huang C, Deng H, Wei X. Spontaneous apoptosis of cells in therapeutic stem cell preparation exert immunomodulatory effects through release of phosphatidylserine. Signal Transduct Target Ther 2021; 6:270. [PMID: 34262012 PMCID: PMC8280232 DOI: 10.1038/s41392-021-00688-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/08/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cell (MSC)-mediated immunomodulation has been harnessed for the treatment of human diseases, but its underlying mechanism has not been fully understood. Dead cells, including apoptotic cells have immunomodulatory properties. It has been repeatedly reported that the proportion of nonviable MSCs in a MSC therapeutic preparation varied from 5~50% in the ongoing clinical trials. It is conceivable that the nonviable cells in a MSC therapeutic preparation may play a role in the therapeutic effects of MSCs. We found that the MSC therapeutic preparation in the present study had about 5% dead MSCs (DMSCs), characterized by apoptotic cells. Namely, 1 × 106 MSCs in the preparation contained about 5 × 104 DMSCs. We found that the treatment with even 5 × 104 DMSCs alone had the equal therapeutic effects as with 1 × 106 MSCs. This protective effect of the dead MSCs alone was confirmed in four mouse models, including concanavalin A (ConA)- and carbon tetrachloride (CCl4)-induced acute liver injury, LPS-induced lung injury and spinal cord injury. We also found that the infused MSCs died by apoptosis in vivo. Furthermore, the therapeutic effect was attributed to the elevated level of phosphatidylserine (PS) upon the injection of MSCs or DMSCs. The direct administration of PS liposomes (PSLs) mimic apoptotic cell fragments also exerted the protective effects as MSCs and DMSCs. The Mer tyrosine kinase (MerTK) deficiency or the knockout of chemokine receptor C-C motif chemokine receptor 2 (CCR2) reversed these protective effects of MSCs or DMSCs. These results revealed that DMSCs alone in the therapeutic stem cell preparation or the apoptotic cells induced in vivo may exert the same immunomodulatory property as the "living MSCs preparation" through releasing PS, which was further recognized by MerTK and participated in modulating immune cells.
Collapse
Affiliation(s)
- Xuemei He
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.488387.8Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan People’s Republic of China
| | - Weiqi Hong
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Jingyun Yang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Hong Lei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Tianqi Lu
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Cai He
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Zhenfei Bi
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Xiangyu Pan
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Yu Liu
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Lunzhi Dai
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Wei Wang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Canhua Huang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Hongxin Deng
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Xiawei Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| |
Collapse
|
31
|
LIU L, YANG F. Application of Modified Mesenchymal Stem Cells Transplantation in the Treatment of Liver Injury. Physiol Res 2021. [DOI: 10.33549/physiolres.934623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Acute and chronic hepatitis, cirrhosis, and other liver diseases pose a serious threat to human health; however, liver transplantation is the only reliable treatment for the terminal stage of liver diseases. Previous researchers have shown that mesenchymal stem cells (MSCs) are characterized by differentiation and paracrine effects, as well as anti-oxidative stress and immune regulation functions. When MSCs are transplanted into animals, they migrate to the injured liver tissue along with the circulation, to protect the liver and alleviate the injury through the paracrine, immune regulation and other characteristics, making mesenchymal stem cell transplantation a promising alternative therapy for liver diseases. Although the efficacy of MSCs transplantation has been confirmed in various animal models of liver injury, many researchers have also proposed various pretreatment methods to improve the efficacy of mesenchymal stem cell transplantation, but there is still lack a set of scientific methods system aimed at improving the efficacy of transplantation therapy in scientific research and clinical practice. In this review, we summarize the possible mechanisms of MSCs therapy and compare the existing methods of MSCs modification corresponding to the treatment mechanism, hoping to provide as a reference to help future researchers explore a safe and simple transplantation strategy.
Collapse
Affiliation(s)
- L LIU
- School of Basic Medicine, Yangtze University Health Science Center, Jingzhou, China
| | - F YANG
- School of Basic Medicine, Yangtze University Health Science Center, Jingzhou, China
| |
Collapse
|
32
|
Adipose-Derived Stem Cells Secretome and Its Potential Application in "Stem Cell-Free Therapy". Biomolecules 2021; 11:biom11060878. [PMID: 34199330 PMCID: PMC8231996 DOI: 10.3390/biom11060878] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived stem cells (ASCs) secrete many cytokines, proteins, growth factors, and extracellular vesicles with beneficial outcomes that can be used in regenerative medicine. It has great potential, and the development of new treatment strategies using the ASCs secretome is of global interest. Besides cytokines, proteins, and growth factors, the therapeutic effect of secretome is hidden in non-coding RNAs such as miR-21, miR-24, and miR-26 carried via exosomes secreted by adequate cells. The whole secretome, including ASC-derived exosomes (ASC-exos) has been proven in many studies to have immunomodulatory, proangiogenic, neurotrophic, and epithelization activity and can potentially be used for neurodegenerative, cardiovascular, respiratory, inflammatory, and autoimmune diseases as well as wound healing treatment. Due to limitations in the use of stem cells in cell-based therapy, its secretome with emphasis on exosomes seems to be a reasonable and safer alternative with increased effectiveness and fewer side effects. Moreover, the great advantage of cell-free therapy is the possibility of biobanking the ASCs secretome. In this review, we focus on the current state of knowledge on the use of the ASCs secretome in stem cell-free therapy.
Collapse
|
33
|
Luan Y, Kong X, Feng Y. Mesenchymal stem cells therapy for acute liver failure: Recent advances and future perspectives. LIVER RESEARCH 2021; 5:53-61. [PMID: 39959343 PMCID: PMC11791815 DOI: 10.1016/j.livres.2021.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/16/2021] [Accepted: 03/24/2021] [Indexed: 12/21/2022]
Abstract
Acute liver failure (ALF) is a life-threatening disease characterized by the rapid development of hepatocyte death and a systemic inflammatory response, which leads to high mortality. Despite the prevention of ALF complications, therapeutic effectiveness remains limited because of the rapid disease progression. Thus, there is a need to explore various therapeutic approaches. Currently, the only effective treatment is liver transplantation; However, the lack of donors, surgical complications, immunosuppression, and high medical costs limit its clinical application. Recently, mesenchymal stem cells (MSCs) have been found to exert hepatoprotective effects in ALF through suppression of inflammation, immunoregulation, promotion of mitosis, anti-apoptosis effects, and alleviation of the metabolic and oxidative stress imbalance. In this review, we summarize the advantages and disadvantages of MSCs from different sources and their molecular mechanisms in ALF treatment, along with future perspectives that may provide guidance to improve the current status of MSCs therapy for ALF.
Collapse
Affiliation(s)
- Yuling Luan
- Department of General Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoni Kong
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Feng
- Department of General Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
34
|
Zhang J, Mohsin A, Peng Y, Dai Y, Zhuang Y, Guo M, Zhao P. Sandwich-Type Near-Infrared Conjugated Polymer Nanoparticles for Revealing the Fate of Transplanted Human Umbilical Cord Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:3512-3520. [PMID: 33435676 DOI: 10.1021/acsami.0c13815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Near-infrared conjugated polymer nanoparticles (NIR-CPNs) have been widely used in in vivo imaging fields. However, most of them face the aggregation-induced fluorescence quenching (ACQ) dilemma and serious dye leakage behavior, which impedes the long-term monitoring of transplanted cells in vivo. In the present work, a novel strategy of sandwich-type encapsulation of the conjugated polymer interlayer in the crystalline SiO2 core + shell (SSiO2@SPFTBT@CSiO2) is developed, which works well to avoid the ACQ problem by homogeneously dispersing poly((9,9-dioctylfluorene-2,7-diyl)-alt-(4,7-di(thiophene-2-yl)-2,1,3-benzothiadiazole)-5',5″-diyl) (PFTBT) and suppressing intermolecular π-π stacking. Furthermore, the unparalleled nanostructure efficiently stabilizes nanoparticles and successfully achieves long-term biocompatibility without interfering the biological characteristics of stem cells, indicating the potential of SSiO2@SPFTBT@CSiO2 in cell labeling. In addition, the fate of human umbilical cord mesenchymal stem cells (hucMSCs) in a mouse model with acute liver injury was disclosed. We found that the hucMSCs mainly migrated from the lungs to the injured liver and most transplanted hucMSCs were cleared up by the liver at 8 days post-injection. Revelation of the shuttle process and period will benefit in improving the clinical efficacy of hucMSCs, and the sandwich-type encapsulation strategy could also open a new avenue to obtain bright and robust NIR-CPNs for long-term fluorescence imaging.
Collapse
Affiliation(s)
- Junhong Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Ali Mohsin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Yan Peng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Yichen Dai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Yingping Zhuang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Peng Zhao
- School of Chemistry and Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, P. R. China
| |
Collapse
|
35
|
Shimasaki M, Ueda S, Ichiseki T, Hirata H, Kawahara N, Ueda Y. Resistance of bone marrow mesenchymal stem cells in a stressed environment - Comparison with osteocyte cells. Int J Med Sci 2021; 18:1375-1381. [PMID: 33628093 PMCID: PMC7893571 DOI: 10.7150/ijms.52104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/17/2020] [Indexed: 11/05/2022] Open
Abstract
Introduction: Recently, the efficacy of mesenchymal stem cells (MSCs) mediated by their tissue repair and anti-inflammatory actions in the prevention and therapy of various disorders has been reported. In this research, our attention was focused specifically on the prevention and therapy of glucocorticoid-induced osteonecrosis. We investigated the stress resistance of MSC against glucocorticoid administration and hypoxic stress, which are factors known to induce osteocytic cell death. Materials and Methods: Mouse bone cells (MLO-Y4) and bone-marrow derived mouse MSCs were exposed to dexamethasone (Dex), hypoxia of 1% oxygen or both in vitro. Mitochondrial membrane potentials were estimated by mitochondria labeling with a cell-permeant probe (Mito tracker red); expression of these apoptosis-inducing molecules, oxidative stress marker (8-hydroxy-2'-deoxyguanosine), caspase-3, -9, and two apoptosis-inhibiting molecules, energy-producing ATP synthase (ATP5A) and X-linked inhibitor of apoptosis protein (XIAP), were analyzed by both immunofluorescence and western blot. Results: With exposure to either dexamethasone or hypoxia, MLO-Y4 showed reduced mitochondrial membrane potential, ATP5A and upregulation of 8-OHdG, cleaved caspases and XIAP. Those changes were significantly enhanced by treatment with dexamethasone plus hypoxia. In MSCs, however, mitochondrial membrane potentials were preserved, while no significant changes in the pro-apoptosis or anti-apoptosis molecules analyzed were found even with exposure to both dexamethasone and hypoxia. No such effects induced by treatment with dexamethasone, hypoxia, or both were demonstrated in MSCs at all. Discussion: In osteocyte cells subjected to the double stresses of glucocorticoid administration and a hypoxic environment osteocytic cell death was mediated via mitochondria. In contrast, MSC subjected to the same stressors showed preservation of mitochondrial function and reduced oxidative stress. Accordingly, even under conditions sufficiently stressful to cause the osteocytic cell death in vivo, it was thought that the function of MSC could be preserved, suggesting that in the case of osteonecrosis preventative and therapeutic strategies incorporating their intraosseous implantation may be promising.
Collapse
Affiliation(s)
- Miyako Shimasaki
- Department of Pathology 2, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku-gun, Ishikawa, 920-0293, Japan
| | - Shusuke Ueda
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Toru Ichiseki
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Hiroaki Hirata
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Norio Kawahara
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Yoshimichi Ueda
- Department of Pathology 2, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku-gun, Ishikawa, 920-0293, Japan
| |
Collapse
|
36
|
Three Cases of Alcohol-Induced Acute-On-Chronic Liver Failure With Successful Support by Adipose-Derived Stem Cells. Clin Transl Gastroenterol 2020; 10:e00095. [PMID: 31789934 PMCID: PMC6970561 DOI: 10.14309/ctg.0000000000000095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES: Acute liver failure (ALF) and acute-on-chronic liver failure (AOCLF) are critical medical conditions with urgent therapy requirements. When ALF or AOCLF are due to alcohol intoxication or based on chronic alcohol abuse, virtually, no therapeutic options are available as liver transplantation is prohibited. In this case series, treatment of alcohol-induced ALF/AOCLF with adipose--derived stem cells (ASC) was tested under compassionate use. METHODS: ASC from 2 donors were isolated, cultured, and expanded by established protocols. ASC were administered to 3 individuals with either ALF or AOCLF due to alcohol abuse under compassionate use. Clinical presentation, serum measurements, and other diagnostic methods were compiled before ASC treatment and during the disease course after ASC administration. RESULTS: Three patients were admitted to the Department of Gastroenterology, Hepatology, and Infectious Diseases (University Hospital Magdeburg) with acute or AOCLF due to alcohol abuse. All 3 patients presented in impaired general condition and with elevated, in 1 case drastically elevated, serum liver enzyme concentrations. Treatment with ASC led to improvements in general condition and reduction of serum transaminases. In 2 cases, reduction of liver stiffness and increase of liver function by the C13 methacetin breath test were observed after ASC treatment. Recovery to a normal condition was achieved between 1 and 2 months after ASC treatment. No adverse effects associated to ASC treatment were observed. DISCUSSION: ASC treatment may be a feasible option to enhance recovery from alcohol-induced ALF or AOCLF. ASC treatment seems safe in the presented cases.
Collapse
|
37
|
Yang F, Zhang Q, Huang S, Ma D. Recent advances of near infrared inorganic fluorescent probes for biomedical applications. J Mater Chem B 2020; 8:7856-7879. [PMID: 32749426 DOI: 10.1039/d0tb01430c] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Near infrared (NIR)-excitable and NIR-emitting probes have fuelled advances in biomedical applications owing to their power in enabling deep tissue imaging, offering high image contrast and reducing phototoxicity. There are essentially three NIR biological windows, i.e., 700-950 nm (NIR I), 1000-1350 nm (NIR II) and 1550-1870 nm (NIR III). Recently emerging optical probes that can be excited by an 800 nm laser and emit in the NIR II or III windows, denoted as NIR I-to-NIR II/III, are particularly attractive. That is because the longer wavelengths in the NIR II and NIR III windows offer deeper penetration and higher signal to noise ratio than those in the NIR I window. NIR imaging has indeed become a quickly evolving field and, simultaneously, stimulated the further development of new classes of NIR I-to-NIR II/III inorganic fluorescent probes, which include PbS, Ag2S-based quantum dots (QDs) and rare earth (RE) doped NPs (RENPs) that possess quite diverse optical properties and follow different emission mechanisms. This review summarizes the recent progress on material merits, synthetic routes, the rational choice of excitation in the NIR I window, NIR II/III emission optimization, and surface modification of aforementioned fluorescent probes. We also introduce the latest notable accomplishments enabled by these probes in fluorescence imaging, lifetime-based multiplexed imaging and photothermal therapy (PTT), together with a critical discussion of forthcoming challenges and perspectives for clinic use.
Collapse
Affiliation(s)
- Fan Yang
- Institut National de la Recherche Scientifique, Centre Énergie, Matériaux et Télécommunications, 1650 Boul. Lionel-Boulet, Varennes, Québec J3X 1S2, Canada.
| | | | | | | |
Collapse
|
38
|
Autologous transplantation of adipose-derived stromal cells combined with sevoflurane ameliorates acute lung injury induced by cecal ligation and puncture in rats. Sci Rep 2020; 10:13760. [PMID: 32792558 PMCID: PMC7426944 DOI: 10.1038/s41598-020-70767-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
Adipose-derived stromal cells (ADSCs) have excellent capacities for regeneration and tissue protection, while sevoflurane, as a requisite component of surgical procedures, has shown therapeutic benefit in animal models of sepsis. This study therefore determined if the combination of sevoflurane and ADSCs exerted additional protective effects against acute lung injury (ALI) induced by cecal ligation and puncture (CLP) in rats. The animals were randomized into five groups: (sham operation (group I), CLP followed by mechanical ventilation (group II), CLP plus sevoflurane at 0.5 minimum alveolar concentration (group III), CLP plus intravenous autologous 5 × 106 ADSCs (group IV), and CLP plus sevoflurane and ADSCs (group V). Levels of the pro-inflammatory cytokines tumor necrosis factor-α, transforming growth factor-β1, interleukin-1β and interleukin-6 were significantly increased in CLP rats. Moreover, epithelial sodium channel expression levels and activities of Na/K-ATPase and alveolar fluid clearance were significantly reduced in CLP-induced ALI rats. ADSCs improved all these parameters, and these effects were further enhanced by the addition of sevoflurane. In conclusion, combined treatment with ADSCs and sevoflurane is superior to either ADSCs or sevoflurane therapy alone for preventing ALI. This beneficial effect may be partly due to improved alveolar fluid clearance by the paracrine or systemic production of keratinocyte growth factor and via anti-inflammatory properties.
Collapse
|
39
|
Nazari-Shafti TZ, Neuber S, Garcia Duran A, Xu Z, Beltsios E, Seifert M, Falk V, Stamm C. Human mesenchymal stromal cells and derived extracellular vesicles: Translational strategies to increase their proangiogenic potential for the treatment of cardiovascular disease. Stem Cells Transl Med 2020; 9:1558-1569. [PMID: 32761804 PMCID: PMC7695640 DOI: 10.1002/sctm.19-0432] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) offer great potential for the treatment of cardiovascular diseases (CVDs) such as myocardial infarction and heart failure. Studies have revealed that the efficacy of MSCs is mainly attributed to their capacity to secrete numerous trophic factors that promote angiogenesis, inhibit apoptosis, and modulate the immune response. There is growing evidence that MSC‐derived extracellular vesicles (EVs) containing a cargo of lipids, proteins, metabolites, and RNAs play a key role in this paracrine mechanism. In particular, encapsulated microRNAs have been identified as important positive regulators of angiogenesis in pathological settings of insufficient blood supply to the heart, thus opening a new path for the treatment of CVD. In the present review, we discuss the current knowledge related to the proangiogenic potential of MSCs and MSC‐derived EVs as well as methods to enhance their biological activities for improved cardiac tissue repair. Increasing our understanding of mechanisms supporting angiogenesis will help optimize future approaches to CVD intervention.
Collapse
Affiliation(s)
- Timo Z Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Neuber
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ana Garcia Duran
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Zhiyi Xu
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eleftherios Beltsios
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Martina Seifert
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt- Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Division of Cardiovascular Surgery, University of Zurich, Zurich, Switzerland
| | - Christof Stamm
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
40
|
Liang Z, Yin X, Sun W, Zhang S, Chen X, Pei L, Zhao N. Enhanced protection against lipopolysaccharide-induced acute lung injury by autologous transplantation of adipose-derived stromal cells combined with low tidal volume ventilation in rats. J Cell Physiol 2020; 236:1295-1308. [PMID: 32662079 DOI: 10.1002/jcp.29936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/02/2020] [Indexed: 12/30/2022]
Abstract
Adipose-derived stromal cells (ADSCs) showed excellent capacity in regeneration and tissue protection. Low tidal volume ventilation (LVT) strategy demonstrates a therapeutic benefit on the treatment of acute lung injury/acute respiratory distress syndrome (ALI/ARDS). This study, therefore, aimed to undertaken determine whether the combined LVT and ADSCs treatment exerts additional protection against lipopolysaccharide (LPS)-induced ALI in rats. The animals were randomized into seven groups: Group I (control), Group II (instillation of LPS at 10 mg/kg intratracheally), Group III (LPS+LVT 6 ml/kg), Group IV (LPS+intravenous autologous 5 × 106 ADSCs which were pretreated with a scrambled small interfering RNA [siRNA] of keratinocyte growth factor [KGF] negative control), Group V (LPS+ADSCs which were pretreated with a scrambled siRNA of KGF, Group VI (LPS+LVT and ADSCs as in the Group IV), and Group VII (LPS+LVT and ADSCs as in the Group V). We found that levels of tumor necrosis factor-α, transforming growth factor-β1, and interleukin (IL)-1β and IL-6, the proinflammatory cytokines, were remarkably increased in LPS rats. Moreover, the expressions of ENaC, activity of Na, K-ATPase, and alveolar fluid clearance (AFC) were obviously reduced by LPS-induced ALI. The rats treated by ADSCs showed improved effects in all these changes of ALI and further enhanced by ADSCs combined with LVT treatment. Importantly, the treatment of ADSCs with siRNA-mediated knockdown of KGF partially eliminated the therapeutic effects. In conclusion, combined treatment with ADSCs and LVT not only is superior to either ADSCs or LVT therapy alone in the prevention of ALI. Evidence of the beneficial effect may be partly due to improving AFC by paracrine or systemic production of KGF and anti-inflammatory properties.
Collapse
Affiliation(s)
- Zuodi Liang
- Department of Anesthesiology, The First Hospital Affiliated at China Medical University, Shenyang, China
| | - Xiuru Yin
- Department of Anesthesiology, The First Hospital Affiliated at China Medical University, Shenyang, China
| | - Wenchong Sun
- Department of Anesthesiology, The First Hospital Affiliated at China Medical University, Shenyang, China
| | - Shuo Zhang
- Department of Anesthesiology, The First Hospital Affiliated at China Medical University, Shenyang, China
| | - Xiaohuan Chen
- Department of Anesthesiology, The First Hospital Affiliated at China Medical University, Shenyang, China
| | - Ling Pei
- Department of Anesthesiology, The First Hospital Affiliated at China Medical University, Shenyang, China
| | - Ning Zhao
- Department of ENT, The First Hospital Affiliated at China Medical University, Shenyang, China
| |
Collapse
|
41
|
Adiponectin Stimulates Exosome Release to Enhance Mesenchymal Stem-Cell-Driven Therapy of Heart Failure in Mice. Mol Ther 2020; 28:2203-2219. [PMID: 32652045 PMCID: PMC7351027 DOI: 10.1016/j.ymthe.2020.06.026] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/18/2020] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are cultured adult stem cells that originally reside in virtually all tissues, and the gain of MSCs by transplantation has become the leading form of cell therapy in various diseases. However, there is limited knowledge on the alteration of its efficacy by factors in recipients. Here, we report that the cardioprotective properties of intravenously injected MSCs in a mouse model of pressure-overload heart failure largely depend on circulating adiponectin, an adipocyte-secreted factor. The injected MSCs exert their function through exosomes, extracellular vesicles of endosome origin. Adiponectin stimulated exosome biogenesis and secretion through binding to T-cadherin, a unique glycosylphosphatidylinositol-anchored cadherin, on MSCs. A pharmacological or adenovirus-mediated genetic increase in plasma adiponectin enhanced the therapeutic efficacy of MSCs. Our findings provide novel insights into the importance of adiponectin in mesenchymal-progenitor-mediated organ protections.
Collapse
|
42
|
Fujiwara O, Prasai A, Perez-Bello D, El Ayadi A, Petrov IY, Esenaliev RO, Petrov Y, Herndon DN, Finnerty CC, Prough DS, Enkhbaatar P. Adipose-derived stem cells improve grafted burn wound healing by promoting wound bed blood flow. BURNS & TRAUMA 2020; 8:tkaa009. [PMID: 32346539 PMCID: PMC7175768 DOI: 10.1093/burnst/tkaa009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 01/22/2020] [Accepted: 01/30/2000] [Indexed: 01/08/2023]
Abstract
BACKGROUND Researchers have explored the use of adipose-derived stem cells (ASCs) as a cell-based therapy to cover wounds in burn patients; however, underlying mechanistic aspects are not completely understood. We hypothesized that ASCs would improve post-burn wound healing after eschar excision and grafting by increasing wound blood flow via induction of angiogenesis-related pathways. METHODS To test the hypothesis, we used an ovine burn model. A 5 cm2 full thickness burn wound was induced on each side of the dorsum. After 24 hours, the burned skin was excised and a 2 cm2 patch of autologous donor skin was grafted. The wound sites were randomly allocated to either topical application of 7 million allogeneic ASCs or placebo treatment (phosphate-buffered saline [PBS]). Effects of ASCs culture media was also compared to those of PBS. Wound healing was assessed at one and two weeks following the application of ASCs. Allogeneic ASCs were isolated, cultured and characterized from non-injured healthy sheep. The identity of the ASCs was confirmed by flow cytometry analysis, differentiation into multiple lineages and gene expression via real-time polymerase chain reaction. Wound blood flow, epithelialization, graft size and take and the expression of vascular endothelial growth factor (VEGF) were determined via enzyme-linked immunosorbent assay and Western blot. RESULTS Treatment with ASCs accelerated the patch graft growth compared to the control (p < 0.05). Topical application of ASCs significantly increased wound blood flow (p < 0.05). Expression of VEGF was significantly higher in the wounds treated with ASCs compared to control (p < 0.05). CONCLUSIONS ASCs accelerated grafted skin growth possibly by increasing the blood flow via angiogenesis induced by a VEGF-dependent pathway.
Collapse
Affiliation(s)
- Osamu Fujiwara
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Anesh Prasai
- Department of Surgery, University of Texas Medical Branch, Galveston, 301 University BLVD TX 77555, USA
- Shriners Hospitals for Children – Galveston, 815 Market Street Galveston, TX 77555, USA
| | - Dannelys Perez-Bello
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Amina El Ayadi
- Department of Surgery, University of Texas Medical Branch, Galveston, 301 University BLVD TX 77555, USA
- Shriners Hospitals for Children – Galveston, 815 Market Street Galveston, TX 77555, USA
- Sealy Center for Molecular Medicine, and the Institute for Translational Sciences, University of Texas Medical Branch, 301 University BLVD Galveston, TX 77555, USA
| | - Irene Y Petrov
- Center for Biomedical Engineering, University of Texas Medical Branch, 601 Harbor Side Dr. Galveston, TX 77555, USA
| | - Rinat O Esenaliev
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
- Center for Biomedical Engineering, University of Texas Medical Branch, 601 Harbor Side Dr. Galveston, TX 77555, USA
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, 301 University BLVD Galveston, TX 77555, USA
| | - Yuriy Petrov
- Center for Biomedical Engineering, University of Texas Medical Branch, 601 Harbor Side Dr. Galveston, TX 77555, USA
| | - David N Herndon
- Department of Surgery, University of Texas Medical Branch, Galveston, 301 University BLVD TX 77555, USA
- Shriners Hospitals for Children – Galveston, 815 Market Street Galveston, TX 77555, USA
| | - Celeste C Finnerty
- Department of Surgery, University of Texas Medical Branch, Galveston, 301 University BLVD TX 77555, USA
- Shriners Hospitals for Children – Galveston, 815 Market Street Galveston, TX 77555, USA
- Sealy Center for Molecular Medicine, and the Institute for Translational Sciences, University of Texas Medical Branch, 301 University BLVD Galveston, TX 77555, USA
| | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
- Shriners Hospitals for Children – Galveston, 815 Market Street Galveston, TX 77555, USA
| |
Collapse
|
43
|
de Souza BM, Rodrigues M, de Oliveira FS, da Silva LPA, Bouças AP, Portinho CP, Dos Santos BP, Camassola M, Rocha D, Lysakowski S, Martini J, Leitão CB, Nardi NB, Bauer AC, Crispim D. Improvement of human pancreatic islet quality after co-culture with human adipose-derived stem cells. Mol Cell Endocrinol 2020; 505:110729. [PMID: 31972330 DOI: 10.1016/j.mce.2020.110729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/30/2019] [Accepted: 01/17/2020] [Indexed: 01/08/2023]
Abstract
The aim of this study was to investigate whether co-culture of human islets with adipose-derived stem cells (ASCs) can improve islet quality and to evaluate which factors play a role in the protective effect of ASCs against islet dysfunction. Islets and ASCs were cultured in three experimental groups for 24 h, 48 h, and 72 h: 1) indirect co-culture of islets with ASC monolayer (Islets/ASCs); 2) islets alone; and 3) ASCs alone. Co-culture with ASCs improved islet viability and function in all culture time-points analyzed. VEGFA, HGF, IL6, IL8, IL10, CCL2, IL1B, and TNF protein levels were increased in supernatants of islet/ASC group compared to islets alone, mainly after 24 h. Moreover, VEGFA, IL6, CCL2, HIF1A, XIAP, CHOP, and NFKBIA genes were differentially expressed in islets from the co-culture condition compared to islets alone. In conclusion, co-culture of islets with ASCs promotes improvements in islet quality.
Collapse
Affiliation(s)
- Bianca M de Souza
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, RS, Brazil.
| | - Michelle Rodrigues
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil
| | - Fernanda S de Oliveira
- Laboratory of Cell Differentiation, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Liana P A da Silva
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil
| | - Ana P Bouças
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, RS, Brazil
| | - Ciro P Portinho
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil
| | - Bruno P Dos Santos
- Laboratory for Tissue Bioengineering (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Melissa Camassola
- Laboratory for Stem Cells and Tissue Engineering, Post-Graduation Program in Cellular and Molecular Biology Applied to Health, Universidade Luterana do Brasil, Canoas, RS, Brazil
| | - Dagoberto Rocha
- Post-Graduation Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Simone Lysakowski
- Organ Procurement Organization, Santa Casa de Misericórdia de Porto Alegre. Porto Alegre, RS, Brazil
| | - Juliano Martini
- Transplant Center, Surgery Department, Hospital Dom Vicente Scherer, Santa Casa de Misericórdia de Porto Alegre. Porto Alegre, RS, Brazil
| | - Cristiane B Leitão
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, RS, Brazil
| | - Nance B Nardi
- Laboratory for Stem Cells and Tissue Engineering, Post-Graduation Program in Cellular and Molecular Biology Applied to Health, Universidade Luterana do Brasil, Canoas, RS, Brazil
| | - Andrea C Bauer
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, RS, Brazil
| | - Daisy Crispim
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, RS, Brazil
| |
Collapse
|
44
|
Adipose-Derived Stem Cell Transplantation Attenuates Inflammation and Promotes Liver Regeneration after Ischemia-Reperfusion and Hemihepatectomy in Swine. Stem Cells Int 2019; 2019:2489584. [PMID: 31827526 PMCID: PMC6885808 DOI: 10.1155/2019/2489584] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/17/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023] Open
Abstract
Aim To study the anti-inflammatory and liver regenerative effects of adipose-derived mesenchymal stem cells (ADSCs) on a porcine model of ischemia-reperfusion (IR) and hemihepatectomy. Methods Eighteen healthy Bama miniature pigs were randomly divided into the sham-operated (sham), untreated IR injury (IRI), and ADSC-transplanted (ADSC) groups. Hepatic IR was established by laparoscopic hemihepatectomy. ADSCs were transplanted directly into the liver parenchyma after the surgery. Hepatic inflammation and liver regeneration were evaluated by histopathological examination and assessment of relevant cytokines and other factors. Results ADSC transplantation successfully ameliorated the IRI-induced histopathological damage and the high levels of pro-inflammatory cytokines like IL-1β, IL-6, and TNF-α. In addition, the ADSCs enhanced the expression of the anti-inflammatory IL-10, regenerative factors including HGF, Cyclin D1, and proliferating cell nuclear antigen (PCNA), and angiogenic factors like VEGF, ANG-1, and ANG-2. Conclusions ADSCs attenuated the hepatic IRI-induced inflammatory response and promoted liver regeneration.
Collapse
|
45
|
Gentile P, Calabrese C, De Angelis B, Pizzicannella J, Kothari A, Garcovich S. Impact of the Different Preparation Methods to Obtain Human Adipose-Derived Stromal Vascular Fraction Cells (AD-SVFs) and Human Adipose-Derived Mesenchymal Stem Cells (AD-MSCs): Enzymatic Digestion Versus Mechanical Centrifugation. Int J Mol Sci 2019; 20:5471. [PMID: 31684107 PMCID: PMC6862236 DOI: 10.3390/ijms20215471] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/27/2019] [Accepted: 11/01/2019] [Indexed: 12/16/2022] Open
Abstract
Autologous therapies using adipose-derived stromal vascular fraction (AD-SVFs) and adult adipose-derived mesenchymal stem cells (AD-MSCs) warrant careful preparation of the harvested adipose tissue. Currently, no standardized technique for this preparation exists. Processing quantitative standards (PQSs) define manufacturing quantitative variables (such as time, volume, and pressure). Processing qualitative standards (PQLSs) define the quality of the materials and methods in manufacturing. The purpose of the review was to use PQSs and PQLSs to report the in vivo and in vitro results obtained by different processing kits that use different procedures (enzymatic vs. non-enzymatic) to isolate human AD-SVFs/AD-MSCs. PQSs included the volume of fat tissue harvested and reagents used, the time/gravity of centrifugation, and the time, temperature, and tilt level/speed of incubation and/or centrifugation. PQLSs included the use of a collagenase, a processing time of 30 min, kit weight, transparency of the kit components, the maintenance of a closed sterile processing environment, and the use of a small centrifuge and incubating rocker. Using a kit with the PQSs and PQLSs described in this study enables the isolation of AD-MSCs that meet the consensus quality criteria. As the discovery of new critical quality attributes (CQAs) of AD-MSCs evolve with respect to purity and potency, adjustments to these benchmark PQSs and PQLs will hopefully isolate AD-MSCs of various CQAs with greater reproducibility, quality, and safety. Confirmatory studies will no doubt need to be completed.
Collapse
Affiliation(s)
- Pietro Gentile
- Surgical Science Department, Plastic and Reconstructive Surgery, University of Rome "Tor Vergata", 00179 Rome, Italy.
| | | | - Barbara De Angelis
- Surgical Science Department, Plastic and Reconstructive Surgery, University of Rome "Tor Vergata", 00179 Rome, Italy.
| | | | - Ashutosh Kothari
- Chief of Breast Surgery Unit, Guy's Hospital, Guy's and St. Thomas' NHS Foundation Trust, London SE1 9RT, UK.
| | - Simone Garcovich
- Institute of Dermatology, F. Policlinico Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
46
|
Mesenchymal Stem Cells From Bone Marrow, Adipose Tissue, and Lung Tissue Differentially Mitigate Lung and Distal Organ Damage in Experimental Acute Respiratory Distress Syndrome. Crit Care Med 2019; 46:e132-e140. [PMID: 29116998 DOI: 10.1097/ccm.0000000000002833] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Mesenchymal stem cells-based therapies have shown promising effects in experimental acute respiratory distress syndrome. Different mesenchymal stem cells sources may result in diverse effects in respiratory diseases; however, there is no information regarding the best source of mesenchymal stem cells to treat pulmonary acute respiratory distress syndrome. We tested the hypothesis that mesenchymal stem cells derived from bone marrow, adipose tissue, and lung tissue would lead to different beneficial effects on lung and distal organ damage in experimental pulmonary acute respiratory distress syndrome. DESIGN Animal study and primary cell culture. SETTING Laboratory investigation. SUBJECTS Seventy-five Wistar rats. INTERVENTIONS Wistar rats received saline (control) or Escherichia coli lipopolysaccharide (acute respiratory distress syndrome) intratracheally. On day 2, acute respiratory distress syndrome animals were further randomized to receive saline or bone marrow, adipose tissue, or lung tissue mesenchymal stem cells (1 × 10 cells) IV. Lung mechanics, histology, and protein levels of inflammatory mediators and growth factors were analyzed 5 days after mesenchymal stem cells administration. RAW 264.7 cells (a macrophage cell line) were incubated with lipopolysaccharide followed by coculture or not with bone marrow, adipose tissue, and lung tissue mesenchymal stem cells (10 cells/mL medium). MEASUREMENTS AND MAIN RESULTS Regardless of mesenchymal stem cells source, cells administration improved lung function and reduced alveolar collapse, tissue cellularity, collagen, and elastic fiber content in lung tissue, as well as decreased apoptotic cell counts in liver. Bone marrow and adipose tissue mesenchymal stem cells administration also reduced levels of tumor necrosis factor-α, interleukin-1β, keratinocyte-derived chemokine, transforming growth factor-β, and vascular endothelial growth factor, as well as apoptotic cell counts in lung and kidney, while increasing expression of keratinocyte growth factor in lung tissue. Additionally, mesenchymal stem cells differently modulated the secretion of biomarkers by macrophages depending on their source. CONCLUSIONS Mesenchymal stem cells from different sources led to variable responses in lungs and distal organs. Bone marrow and adipose tissue mesenchymal stem cells yielded greater beneficial effects than lung tissue mesenchymal stem cells. These findings may be regarded as promising in clinical trials.
Collapse
|
47
|
Hu C, Zhao L, Li L. Current understanding of adipose-derived mesenchymal stem cell-based therapies in liver diseases. Stem Cell Res Ther 2019; 10:199. [PMID: 31287024 PMCID: PMC6613269 DOI: 10.1186/s13287-019-1310-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The liver, the largest organ with multiple synthetic and secretory functions in mammals, consists of hepatocytes, cholangiocytes, hepatic stellate cells (HSCs), sinusoidal endothelial cells, Kupffer cells (KCs), and immune cells, among others. Various causative factors, including viral infection, toxins, autoimmune defects, and genetic disorders, can impair liver function and result in chronic liver disease or acute liver failure. Mesenchymal stem cells (MSCs) from various tissues have emerged as a potential candidate for cell transplantation to promote liver regeneration. Adipose-derived MSCs (ADMSCs) with high multi-lineage potential and self-renewal capacity have attracted great attention as a promising means of liver regeneration. The abundance source and minimally invasive procedure required to obtain ADMSCs makes them superior to bone marrow-derived MSCs (BMMSCs). In this review, we comprehensively analyze landmark studies that address the isolation, proliferation, and hepatogenic differentiation of ADMSCs and summarize the therapeutic effects of ADMSCs in animal models of liver diseases. We also discuss key points related to improving the hepatic differentiation of ADMSCs via exposure of the cells to cytokines and growth factors (GFs), extracellular matrix (ECM), and various physical parameters in in vitro culture. The optimization of culturing methods and of the transplantation route will contribute to the further application of ADMSCs in liver regeneration and help improve the survival rate of patients with liver diseases. To this end, ADMSCs provide a potential strategy in the field of liver regeneration for treating acute or chronic liver injury, thus ensuring the availability of ADMSCs for research, trial, and clinical applications in various liver diseases in the future.
Collapse
Affiliation(s)
- Chenxia Hu
- 0000 0004 1759 700Xgrid.13402.34Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang People’s Republic of China
| | - Lingfei Zhao
- 0000 0004 1759 700Xgrid.13402.34Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang People’s Republic of China
| | - Lanjuan Li
- 0000 0004 1759 700Xgrid.13402.34Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University; Key Laboratory of Kidney Disease Prevention and Control Technology, Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang People’s Republic of China
| |
Collapse
|
48
|
Scioli MG, Storti G, D'Amico F, Gentile P, Kim BS, Cervelli V, Orlandi A. Adipose-Derived Stem Cells in Cancer Progression: New Perspectives and Opportunities. Int J Mol Sci 2019; 20:3296. [PMID: 31277510 PMCID: PMC6651808 DOI: 10.3390/ijms20133296] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
Growing importance has been attributed to interactions between tumors, the stromal microenvironment and adult mesenchymal stem cells. Adipose-derived stem cells (ASCs) are routinely employed in regenerative medicine and in autologous fat transfer procedures. To date, clinical trials have failed to demonstrate the potential pro-oncogenic role of ASC enrichment. Nevertheless, some pre-clinical studies from in vitro and in vivo models have suggested that ASCs act as a potential tumor promoter for different cancer cell types, and support tumor progression and invasiveness through the activation of several intracellular signals. Interaction with the tumor microenvironment and extracellular matrix remodeling, the exosomal release of pro-oncogenic factors as well as the induction of epithelial-mesenchymal transitions are the most investigated mechanisms. Moreover, ASCs have also demonstrated an elective tumor homing capacity and this tumor-targeting capacity makes them a suitable carrier for anti-cancer drug delivery. New genetic and applied nanotechnologies may help to design promising anti-cancer cell-based approaches through the release of loaded intracellular nanoparticles. These new anti-cancer therapies can more effectively target tumor cells, reaching higher local concentrations even in pharmacological sanctuaries, and thus minimizing systemic adverse drug effects. The potential interplay between ASCs and tumors and potential ASCs-based therapeutic approaches are discussed.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Gabriele Storti
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Federico D'Amico
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Bong-Sung Kim
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy.
| |
Collapse
|
49
|
Gentile P, Garcovich S. Concise Review: Adipose-Derived Stem Cells (ASCs) and Adipocyte-Secreted Exosomal microRNA (A-SE-miR) Modulate Cancer Growth and proMote Wound Repair. J Clin Med 2019; 8:855. [PMID: 31208047 PMCID: PMC6616456 DOI: 10.3390/jcm8060855] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/05/2019] [Accepted: 06/12/2019] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ASCs) have been routinely used from several years in regenerative surgery without any definitive statement about their potential pro-oncogenic or anti-oncogenic role. ASCs has proven to favor tumor progression in several experimental cancer models, playing a central role in regulating tumor invasiveness and metastatic potential through several mechanisms, such as the paracrine release of exosomes containing pro-oncogenic molecules and the induction of epithelial-mesenchymal transition. However, the high secretory activity and the preferential tumor-targeting make also ASCs a potentially suitable vehicle for delivery of new anti-cancer molecules in tumor microenvironment. Nanotechnologies, viral vectors, drug-loaded exosomes, and micro-RNAs (MiR) represent additional new tools that can be applied for cell-mediated drug delivery in a tumor microenvironment. Recent studies revealed that the MiR play important roles in paracrine actions on adipose-resident macrophages, and their dysregulation has been implicated in the pathogenesis of obesity, diabetes, and diabetic complications as wounds. Numerous MiR are present in adipose tissues, actively participating in the regulation of adipogenesis, adipokine secretion, inflammation, and inter-cellular communications in the local tissues. These results provide important insights into Adipocyte-secreted exosomal microRNA (A-SE-MiR) function and they suggest evaluating the potential role of A-SE-MiR in tumor progression, the mechanisms underlying ASCs-cancer cell interplay and clinical safety of ASCs-based therapies.
Collapse
Affiliation(s)
- Pietro Gentile
- Surgical Science Department, Plastic and Reconstructive Surgery Unit, University of "Tor Vergata", 00133 Rome, Italy.
| | - Simone Garcovich
- Institute of Dermatology, F. Policlinico Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
50
|
Born S, Dörfel MJ, Hartjen P, Haschemi Yekani SA, Luecke J, Meutsch JK, Westphal JK, Birkelbach M, Köhnke R, Smeets R, Krueger M. A short-term plastic adherence incubation of the stromal vascular fraction leads to a predictable GMP-compliant cell-product. ACTA ACUST UNITED AC 2019; 9:161-172. [PMID: 31508331 PMCID: PMC6726751 DOI: 10.15171/bi.2019.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/14/2022]
Abstract
![]()
Introduction:Mesenchymal stromal/stem cells (MSCs) derived from fat tissue are an encouraging tool for regenerative medicine. They share properties similar to the bone marrow-derived MSCs, but the amount of MSCs per gram of fat tissue is 500x higher. The fat tissue can easily be digested by collagenase, releasing a heterogeneous cell fraction called stromal vascular fraction (SVF) which contains a variable amount of stromal/stem cells. In Europe, cell products like the SVF derived from fat tissue are considered advanced therapy medicinal product (ATMPs). As a consequence, the manufacturing process has to be approved via GMP-compliant process validation. The problem of the process validation for SVF is the heterogeneity of this fraction.
Methods: Here, we modified existing purification strategies by adding an additional plastic adherence incubation of maximal 20 hours after SVF isolation. The resulting cell fraction was characterized and compared to SVF as well as cultivated adipose-derived stromal/stem cells (ASCs) with respect to viability and cell yield, the expression of surface markers, differentiation potential and cytokine expression.
Results: Short-term incubation significantly reduced the heterogeneity of the resulting cell fraction compared to SVF. The cells were able to differentiate into adipocytes, chondrocytes, and osteoblasts. More importantly, they expressed trophic proteins which have been previously associated with the beneficial effects of MSCs. Furthermore, GMP compliance of the production process described herein was acknowledged by the national regulatory agencies (DE_BB_01_GMP_2017_1018).
Conclusion: Addition of a short purification-step after the SVF isolation is a cheap and fast strategy to isolate a homogeneous uncultivated GMP-compliant cell fraction of ASCs.
Collapse
Affiliation(s)
| | | | - Philip Hartjen
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | - Moritz Birkelbach
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Robert Köhnke
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany.,Division, Regenerative Orofacial Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|