1
|
Wong YS, Mançanares AC, Navarrete F, Poblete P, Mendez-Pérez L, Rodriguez-Alvarez L, Castro FO. Short preconditioning with TGFβ of equine adipose tissue-derived mesenchymal stem cells predisposes towards an anti-fibrotic secretory phenotype: A possible tool for treatment of endometrosis in mares. Theriogenology 2024; 225:119-129. [PMID: 38805994 DOI: 10.1016/j.theriogenology.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
Endometrosis in mares is a disease resulting from chronic inflammation characterized by peri glandular fibrosis. There is no effective treatment so far, which opens the door for exploring the use of stem cells as a candidate. Transforming growth factor beta (TGFβ) is crucial for the establishment and progression of fibrosis in mare's endometrosis. We aimed to develop regenerative approaches to treat endometrosis by using mesenchymal stem cells (MSC), for which understanding the effect of TGFβ on exogenous MSC is crucial. We isolated and characterized equine adipose MSC from six donors. Cells were pooled and exposed to 10 ng/ml of TGFβ for 0, 4, and 24 h, after which cells were analyzed for proliferation, migration, mesodermal differentiation, expression of fibrosis-related mRNAs, and prostaglandin E2 secretion. At 24 h of exposition to TGFβ, there was a progressive increase in the contraction of the monolayer, leading to nodular structures, while cell viability did not change. Exposure to TGFβ impaired adipogenic and osteogenic differentiation after 4 h of treatment, which was more marked at 24 h, represented by a decrease in Oil red and Alizarin red staining, as well as a significant drop (p < 0.05) in the expression of key gene regulators of differentiation processes (PPARG for adipose and RUNX2 for osteogenic differentiation). TGFβ increased chondrogenic differentiation as shown by the upsurge in size of the resulting 3D cell pellet and intensity of Alcian Blue staining, as well as the significant up-regulation of SOX9 expression (p < 0.05) at 4 h, which reached a maximum peak at 24 h (p < 0.01), indicative of up-regulation of glycosaminoglycan synthesis. Preconditioning MSC with TGFβ led to a significant increase (p < 0.05) in the expression of myofibroblast gene markers aSMA, COL1A1, and TGFβ at 24 h exposition time. In contrast, the expression of COL3A1 did not change with respect to the control but registered a significant downregulation compared to 4 h (p < 0.05). TGFβ also affected the expression of genes involved in PGE2 synthesis and function; COX2, PTGES, and the PGE2 receptor EP4 were all significantly upregulated early at 4 h (p < 0.05). Cells exposed to TGFβ showed a significant upregulation of PGE2 secretion at 4 h compared to untreated cells (p < 0.05); conversely, at 24 h, the PGE2 values decreased significantly compared to control cells (p < 0.05). Preconditioning MSC for 4 h led to an anti-fibrotic secretory phenotype, while a longer period (24 h) led to a pro-fibrotic one. It is tempting to propose a 4-h preconditioning of exogenous MSC with TGFβ to drive them towards an anti-fibrotic phenotype for cellular and cell-free therapies in fibrotic diseases such as endometrosis of mares.
Collapse
Affiliation(s)
- Yat Sen Wong
- Ph.D Program in Veterinary Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Ana Carolina Mançanares
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Felipe Navarrete
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Pamela Poblete
- Ph.D Program in Veterinary Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | - Lidice Mendez-Pérez
- Ph.D Program in Veterinary Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile
| | | | - Fidel Ovidio Castro
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Chillán, Chile.
| |
Collapse
|
2
|
Gao Y, Chen L, Li Y, Sun S, Ran X. HUC-MSCs combined with platelet lysate treat diabetic chronic cutaneous ulcers in Bama miniature pig. Regen Ther 2024; 26:1138-1149. [PMID: 39640920 PMCID: PMC11617409 DOI: 10.1016/j.reth.2024.11.005] [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: 04/16/2024] [Revised: 07/04/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024] Open
Abstract
Human umbilical cord mesenchymal stem cells (HUC-MSCs) and platelet lysate (PL) shows potential of wound healing. However, MSCs in combination with PL for wound healing is still lacking. In this study, we presented high glucose cultured wound related cells to mimic diabetic chronic ulcers (DCU) cells, wound healing indicators and the TGFβ/Smad signaling pathway were detected by PL cultured HUC-MSC supernatant (MSC-Sp) in vitro. In vivo study, diabetes was induced in pigs feeding a high-energy diet and multiple injections of streptozotocin (125 mg/kg). Chronic wounds were created on both sides of the backs of seven pigs by surgical creation and foreign body compression for eight weeks before treatment. The wounds were treated with saline control (N = 11), PL (N = 11), HUC- MSCs (N = 18, 6 × 106/mL/cm2), and PL + HUC-MSCs (N = 18, 6 × 106/mL/cm2) respectively. Tissue samples were collected to observe new collagen, neovascularization, wound healing factors, and the TGFβ/Smad signaling pathway. The resulting PL-cultured MSC-Sp promoted the proliferation of keratinocytes, fibroblasts, and vascular endothelial cells and inhibited the TGFβ1/TGFβ3 ratio, upregulated VEGF-α and PDGF-BB production by keratinocytes and fibroblasts, and downregulated the expression of CD86, IL-6, and TNF-α in RAW264.7 cells. PL + HUC-MSCs had the best wound healing rate in vivo, and promoted collagen formation, neovascularization, and inflammation, regulated the balance between IL-6/TGFβ1 and IL-6/Arg-1 and upregulated the expression of VEGF-α and TGFβ1. In summary, PL + HUC-MSCs had a better wound healing effect than HUC-MSCs or PL treatment alone by regulating the IL-6/Arg-1 and IL-6/TGFβ1 balance and upregulating TGFβ1, VEGF-α, Col1, and α-SMA.
Collapse
Affiliation(s)
- Yunyi Gao
- Department of Endocrinology & Metabolism, West China Hospital of Sichuan University, Chengdu, China
- Innovation Research Center for Diabetic Foot, Diabetic Foot Care Center, West China Hospital of Sichuan University, Chengdu, China
- Department of Medical Affairs, West China Hospital of Sichuan University, Chengdu, China
| | - Lihong Chen
- Department of Endocrinology & Metabolism, West China Hospital of Sichuan University, Chengdu, China
- Innovation Research Center for Diabetic Foot, Diabetic Foot Care Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yan Li
- Department of Endocrinology & Metabolism, West China Hospital of Sichuan University, Chengdu, China
- Innovation Research Center for Diabetic Foot, Diabetic Foot Care Center, West China Hospital of Sichuan University, Chengdu, China
| | - Shiyi Sun
- Department of Endocrinology & Metabolism, West China Hospital of Sichuan University, Chengdu, China
- Innovation Research Center for Diabetic Foot, Diabetic Foot Care Center, West China Hospital of Sichuan University, Chengdu, China
| | - XingWu Ran
- Department of Endocrinology & Metabolism, West China Hospital of Sichuan University, Chengdu, China
- Innovation Research Center for Diabetic Foot, Diabetic Foot Care Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Yang X, Xiong M, Fu X, Sun X. Bioactive materials for in vivo sweat gland regeneration. Bioact Mater 2024; 31:247-271. [PMID: 37637080 PMCID: PMC10457517 DOI: 10.1016/j.bioactmat.2023.07.025] [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: 04/20/2023] [Revised: 07/30/2023] [Accepted: 07/30/2023] [Indexed: 08/29/2023] Open
Abstract
Loss of sweat glands (SwGs) commonly associated with extensive skin defects is a leading cause of hyperthermia and heat stroke. In vivo tissue engineering possesses the potential to take use of the body natural ability to regenerate SwGs, making it more conducive to clinical translation. Despite recent advances in regenerative medicine, reconstructing SwG tissue with the same structure and function as native tissue remains challenging. Elucidating the SwG generation mechanism and developing biomaterials for in vivo tissue engineering is essential for understanding and developing in vivo SwG regenerative strategies. Here, we outline the cell biology associated with functional wound healing and the characteristics of bioactive materials. We critically summarize the recent progress in bioactive material-based cell modulation approaches for in vivo SwG regeneration, including the recruitment of endogenous cells to the skin lesion for SwG regeneration and in vivo cellular reprogramming for SwG regeneration. We discussed the re-establishment of microenvironment via bioactive material-mediated regulators. Besides, we offer promising perspectives for directing in situ SwG regeneration via bioactive material-based cell-free strategy, which is a simple and effective approach to regenerate SwG tissue with both fidelity of structure and function. Finally, we discuss the opportunities and challenges of in vivo SwG regeneration in detail. The molecular mechanisms and cell fate modulation of in vivo SwG regeneration will provide further insights into the regeneration of patient-specific SwGs and the development of potential intervention strategies for gland-derived diseases.
Collapse
Affiliation(s)
- Xinling Yang
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Mingchen Xiong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| |
Collapse
|
4
|
Administration of stem cells against cardiovascular diseases with a focus on molecular mechanisms: Current knowledge and prospects. Tissue Cell 2023; 81:102030. [PMID: 36709696 DOI: 10.1016/j.tice.2023.102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023]
Abstract
Cardiovascular diseases (CVDs) are a serious global concern for public and human health. Despite the emergence of significant therapeutic advances, it is still the leading cause of death and disability worldwide. As a result, extensive efforts are underway to develop practical therapeutic approaches. Stem cell-based therapies could be considered a promising strategy for the treatment of CVDs. The efficacy of stem cell-based therapeutic approaches is demonstrated through recent laboratory and clinical studies due to their inherent regenerative properties, proliferative nature, and their capacity to differentiate into different cells such as cardiomyocytes. These properties could improve cardiovascular functioning leading to heart regeneration. The two most common types of stem cells with the potential to cure heart diseases are induced pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs). Several studies have demonstrated the use, efficacy, and safety of MSC and iPSCs-based therapies for the treatment of CVDs. In this study, we explain the application of stem cells, especially iPSCs and MSCs, in the treatment of CVDs with a focus on cellular and molecular mechanisms and then discuss the advantages, disadvantages, and perspectives of using this technology in the treatment of these diseases.
Collapse
|
5
|
Lee AH, Ghosh D, Koh IL, Dawson MR. Senescence-associated exosomes transfer miRNA-induced fibrosis to neighboring cells. Aging (Albany NY) 2023; 15:1237-1256. [PMID: 36842089 PMCID: PMC10042705 DOI: 10.18632/aging.204539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023]
Abstract
Radiation-induced fibrosis is a common side effect of radiotherapy, which is the most common treatment for cancer. However, radiation also causes p53-mediated cell cycle arrest, prolonged expression of p21, and the development of senescence in normal cells that reside in irradiated tissues. Bone marrow-derived mesenchymal stem cells (MSCs) accumulate in primary tumor sites because of their natural tropism for inflammatory and fibrotic tissues. MSCs are extremely sensitive to low doses of ionizing radiation and acquire senescence as a result of bystander radiation effects. Senescent cells remain metabolically active but develop a potent senescence-associated secretory phenotype (SASP) that correlates to hyperactive secretion of cytokines, pro-fibrotic growth factors, and exosomes (EXOs). Integrative pathway analysis highlighted that radiation-induced senescence significantly enriched cell-cycle, extracellular matrix, transforming growth factor-β (TGF-β) signaling, and vesicle-mediated transport genes in MSCs. EXOs are cell-secreted nanovesicles (a subclass of small extracellular vesicles) that contain biomaterials-proteins, RNAs, microRNAs (miRNAs)-that are critical in cell-cell communication. miRNA content analysis of secreted EXOs further revealed that radiation-induced senescence uniquely altered miRNA profiles. In fact, several of the standout miRNAs directly targeted TGF-β or downstream genes. To examine bystander effects of radiation-induced senescence, we further treated normal MSCs with senescence-associated EXOs (SA-EXOs). These modulated genes related to TGF-β pathway and elevated both alpha smooth muscle actin (protein increased in senescent, activated cells) and Ki-67 (proliferative marker) expression in SA-EXO treated MSCs compared to untreated MSCs. We revealed SA-EXOs possess unique miRNA content that influence myofibroblast phenotypes via TGF-β pathway activation. This highlights that SA-EXOs are potent SASP factors that play a large role in cancer-related fibrosis.
Collapse
Affiliation(s)
- Amy H Lee
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA
| | - Deepraj Ghosh
- Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Ivy L Koh
- Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Michelle R Dawson
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA
- Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| |
Collapse
|
6
|
Trends in using mesenchymal stromal/stem cells (MSCs) in treating corneal diseases. Ocul Surf 2022; 26:255-267. [DOI: 10.1016/j.jtos.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 12/05/2022]
|
7
|
Kim S, Kim K. Lipid-mediated ex vivo cell surface engineering for augmented cellular functionalities. BIOMATERIALS ADVANCES 2022; 140:213059. [PMID: 35961186 DOI: 10.1016/j.bioadv.2022.213059] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/23/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
Once administrated, intercellular adhesion to recognize and/or arrest target cells is essential for specific treatments, especially for cancer or tumor. However, immune cells administrated into the tumor-microenvironment could lose their intrinsic functionalities such as target recognition ability, resulting in an ineffective cancer immunotherapy. Various manipulation techniques for decorating functional moieties onto cell surface and enhancing target recognition have been developed. A hydrophobic interaction-mediated ex-vivo cell surface engineering using lipid-based biomaterials could be a state-of-the-art engineering technique that could achieve high-efficiency cell surface modification by a single method without disturbance of intrinsic characteristics of cells. In this regard, this review provides design principles for the development of lipid-based biomaterials with a linear structure of lipid, polyethylene glycol, and functional group, strategies for the synthesis process, and their practical applications in biomedical engineering. Especially, we provide new insights into the development of a novel surface coating techniques for natural killer (NK) cells with engineering decoration of cancer targeting moieties on their cell surfaces. Among immune cells, NK cells are interesting cell population for substituting T cells because of their excellent safety and independent anticancer efficacy. Thus, optimal strategies to select cancer-type-specific targeting moieties and present them onto the surface of immune cells (especially, NK cells) using lipid-based biomaterials could provide additional tools to capture cancer cells for developing novel immune cell therapy products. Enhanced anticancer efficacies by surface-engineered NK cells have been demonstrated both in vitro and in vivo. Therefore, it could be speculated that recent progresses in cell surface modification technology via lipid-based biomaterials could strengthen immune surveillance and immune synapses for utilization in a next-generation cancer immunotherapy, beyond currently available genetic engineering tool such as chimeric antigen receptor-mediated immune cell modulation.
Collapse
Affiliation(s)
- Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Romagano MP, Sherman LS, Shadpoor B, El-Far M, Souayah S, Pamarthi SH, Kra J, Hood-Nehra A, Etchegaray JP, Williams SF, Rameshwar P. Aspirin-Mediated Reset of Preeclamptic Placental Stem Cell Transcriptome - Implication for Stabilized Placental Function. Stem Cell Rev Rep 2022; 18:3066-3082. [PMID: 35908144 DOI: 10.1007/s12015-022-10419-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 11/24/2022]
Abstract
Preeclampsia (PE) is a pregnancy-specific disease, occurring in ~ 2-10% of all pregnancies. PE is associated with increased maternal and perinatal morbidity and mortality, hypertension, proteinuria, disrupted artery remodeling, placental ischemia and reperfusion, and inflammation. The mechanism of PE pathogenesis remains unresolved explaining limited treatment. Aspirin is used to reduce the risk of developing PE. This study investigated aspirin's effect on PE-derived placenta mesenchymal stem cells (P-MSCs). P-MSCs from chorionic membrane (CM), chorionic villi, membranes from the maternal and amniotic regions, and umbilical cord were similar in morphology, phenotype and multipotency. Since CM-derived P-MSCs could undergo long-term passages, the experimental studies were conducted with this source of P-MSCs. Aspirin (1 mM) induced significant functional and transcriptomic changes in PE-derived P-MSCs, similar to healthy P-MSCs. These include cell cycle quiescence, improved angiogenic pathways, and immune suppressor potential. The latter indicated that aspirin could induce an indirect program to mitigate PE-associated inflammation. As a mediator of activating the DNA repair program, aspirin increased p53, and upregulated genes within the basic excision repair pathway. The robust ability for P-MSCs to maintain its function with high dose aspirin contrasted bone marrow (M) MSCs, which differentiated with eventual senescence/aging with 100 fold less aspirin. This difference cautions how data from other MSC sources are extrapolated to evaluate PE pathogenesis. Dysfunction among P-MSCs in PE involves a network of multiple pathways that can be restored to an almost healthy functional P-MSC. The findings could lead to targeted treatment for PE.
Collapse
Affiliation(s)
- Matthew P Romagano
- Department of Obstetrics, Gynecology and Reproductive Health, D-Maternal Fetal Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Lauren S Sherman
- Department of Medicine-Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA.,Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA
| | - Bobak Shadpoor
- Department of Medicine-Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA.,Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA
| | - Markos El-Far
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA
| | - Sami Souayah
- Department of Medicine-Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Sri Harika Pamarthi
- Department of Medicine-Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Joshua Kra
- Department of Medicine-Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA.,Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
| | - Anupama Hood-Nehra
- Department of Medicine-Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA.,Rutgers Cancer Institute of New Jersey, Newark, NJ, USA
| | | | - Shauna F Williams
- Department of Obstetrics, Gynecology and Reproductive Health, D-Maternal Fetal Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA.
| | - Pranela Rameshwar
- Department of Medicine-Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
9
|
Salehipour Bavarsad S, Jalali MT, Bijan Nejad D, Alypoor B, Babaahmadi Rezaei H, Mohammadtaghvaei N. TGFβ1-Pretreated Exosomes of Wharton Jelly Mesenchymal Stem Cell as a Therapeutic Strategy for Improving Liver Fibrosis. HEPATITIS MONTHLY 2022; 22. [DOI: 10.5812/hepatmon-123416] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/14/2022] [Accepted: 05/26/2022] [Indexed: 01/03/2025]
Abstract
Background: Mesenchymal stem cells (MSCs) are the most promising tools for cell treatment and human tissue regeneration, e.g., in liver fibrosis. Mesenchymal stem cells repair tissue damage through paracrine mediators such as exosomes. Types and concentrations of inflammatory mediators, including transforming growth factor-beta (TGFβ1), in MSCs microenvironment can affect MSCs’ function and therapeutic potency. Objectives: This experimental study aimed to explore the effects of Wharton jelly MSCs (WJ-MSCs) exosomes on fibrotic gene expression and Smad2/3 phosphorylation (phospho-Smad2/3 (p-Smad2/3)). Moreover, we further investigated whether WJ-MSCs pretreatment with different concentrations of TGFβ1 changes the anti-fibrotic properties of their exosomes. Methods: After isolation from the umbilical cord, WJ-MSCs were characterized by observing differentiation and measuring surface biomarkers using flowcytometry. The WJ-MSC-derived exosomes were extracted and identified using transmission electron microscopy (TEM), dynamic light scattering (DLS), and western blotting. Real-time PCR and western blot for extracellular matrix (ECM) and p-Smad2/3 expression detection were used to investigate the effect of exosomes from untreated and TGFβ1-pretreated WJ-MSCs on activated hepatic stellate cells (HSCs). Results: Phospho-Smad2/3, α-smooth muscle actin (α-SMA), and collagen1α1 levels were enhanced following treatment with TGFβ1, whereas E-cadherin was decreased. However, the outcomes were reversed after treatment with WJ-MSC-derived exosomes. Exosomes from TGFβ1-pretreated WJ-MSCs induced a significant decrease in p-Smad2/3 levels in activated HSCs, accompanied by the upregulation of E-cadherin gene expression and downregulation of α-SMA and collagen1α1 when compared to untreated WJ-MSC-derived exosomes. The p-Smad2/3 proteins were significantly decreased (fold change: 0.23, P-value < 0.0001) after exposure to low-dose TGFβ1-pretreated WJ-MSC-derived exosomes (0.1 ng/mL), showing the best effect on activated HSCs. Conclusions: Exosomes derived from untreated WJ-MSCs could regress TGFβ-Smad2/3 signaling and the expression of fibrotic markers in activated LX-2 cells. However, these effects were significantly profound with applying exosomes derived from 0.1 ng/mL TGFβ-pretreated WJ-MSCs. We also observed the dose-response effects of TGFβ on WJ-MSCs-derived exosomes. Therefore, exosomes derived from TGFβ-pretreated WJ-MSCs may be critical in improving fibrosis and benefit liver fibrosis patients.
Collapse
|
10
|
Aulanni’am A, Raissa R, Riawan W, Wuragil DK, Permata FS, Beltran MAG. Epidermal Stem Cell in Wound Healing of Gliricidia sepium Leaves from Indonesia and the Philippines in Rats (Rattus norvegicus). Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
AIM: This study intended to investigate the regenerate wound, due to the ointment therapy containing Gliricidia sepium leaves that has potential-induced epidermal stem cells producing. It determined its effect on the expression of transforming growth factor-β1 (TGF-β1), Smad-3, β-catenin, LGR-6.
MATERIALS AND METHODS: About 16 Wistar male rats aged approximately 2 months (150–200g) were used and were divided into four treatment groups (T1, positive control; T2, negative control; T3, wounds treated with G. sepium from Indonesia; and T4, wounds treated with G. sepium from the Philippines). The treatment of ointment was applied to the wound for 3 days. The expression of TGF-β1, Smad-3, β-catenin, and LGR-6 was observed by immunohistochemistry staining.
RESULTS: G. sepium leaves significantly (p < 0.05) upregulated the expression of TGF-β1, Smad-3, β-catenin, and LGR-6 in the group treated with Indonesian G. sepium leaves were higher than that in the group treated with G. sepium leaves from the Philippines.
CONCLUSIONS: Both leaves Varian contain flavonoids, saponins, and tannins, which act as producing epidermal stem cell agents to enhance the wound healing process. It can be concluded that both Gl. sepium Varian Indonesia and the Philippines have a potential effect on wound healing.
Collapse
|
11
|
Choudhery MS. Strategies to improve regenerative potential of mesenchymal stem cells. World J Stem Cells 2021; 13:1845-1862. [PMID: 35069986 PMCID: PMC8727227 DOI: 10.4252/wjsc.v13.i12.1845] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/31/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
In the last few decades, stem cell-based therapies have gained attention worldwide for various diseases and disorders. Adult stem cells, particularly mesenchymal stem cells (MSCs), are preferred due to their significant regenerative potential in cellular therapies and are currently involved in hundreds of clinical trials. Although MSCs have high self-renewal as well as differentiation potential, such abilities are compromised with “advanced age” and “disease status” of the donor. Similarly, cell-based therapies require high cell number for clinical applications that often require in vitro expansion of cells. It is pertinent to note that aged individuals are the main segment of population for stem cell-based therapies, however; autologous use of stem cells for such patients (aged and diseased) does not seem to give optimal results due to their compromised potential. In vitro expansion to obtain large numbers of cells also negatively affects the regenerative potential of MSCs. It is therefore essential to improve the regenerative potential of stem cells compromised due to “in vitro expansion”, “donor age” and “donor disease status” for their successful autologous use. The current review has been organized to address the age and disease depleted function of resident adult stem cells, and the strategies to improve their potential. To combat the problem of decline in the regenerative potential of cells, this review focuses on the strategies that manipulate the cell environment such as hypoxia, heat shock, caloric restriction and preconditioning with different factors.
Collapse
Affiliation(s)
- Mahmood S Choudhery
- Department of Biomedical Sciences, King Edward Medical University, Lahore 54000, Punjab, Pakistan
- Department of Genetics and Molecular Biology, University of Health Sciences, Lahore 54600, Punjab, Pakistan
| |
Collapse
|
12
|
Marofi F, Alexandrovna KI, Margiana R, Bahramali M, Suksatan W, Abdelbasset WK, Chupradit S, Nasimi M, Maashi MS. MSCs and their exosomes: a rapidly evolving approach in the context of cutaneous wounds therapy. Stem Cell Res Ther 2021; 12:597. [PMID: 34863308 PMCID: PMC8642895 DOI: 10.1186/s13287-021-02662-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/14/2021] [Indexed: 12/18/2022] Open
Abstract
Currently, mesenchymal stem/stromal stem cell (MSC) therapy has become a promising option for accelerating cutaneous wound healing. In vivo reports have outlined the robust competences of MSCs to offer a solid milieu by inhibition of inflammatory reactions, which in turn, enables skin regeneration. Further, due to their great potential to stimulate angiogenesis and also facilitate matrix remodeling, MSCs hold substantial potential as future therapeutic strategies in this context. The MSCs-induced wound healing is thought to mainly rely on the secretion of a myriad of paracrine factors in addition to their direct differentiation to skin-resident cells. Besides, MSCs-derived exosomes as nanoscale and closed membrane vesicles have recently been suggested as an effective and cell-free approach to support skin regeneration, circumventing the concerns respecting direct application of MSCs. The MSCs-derived exosomes comprise molecular components including lipid, proteins, DNA, microRNA, and also mRNA, which target molecular pathways and also biological activities in recipient cells (e.g., endothelial cell, keratinocyte, and fibroblast). The secreted exosome modifies macrophage activation, stimulates angiogenesis, and instigates keratinocytes and dermal fibroblast proliferations as well as migrations concurrently regulate inherent potential of myofibroblast for adjustment of turnover of the ECM. In the present review, we will focus on the recent findings concerning the application of MSCs and their derivative exosome to support wound healing and skin regeneration, with special focus on last decade in vivo reports.
Collapse
Affiliation(s)
- Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master’s Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Mahta Bahramali
- Biotechnology Department, University of Tehran, Tehran, Iran
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210 Thailand
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200 Thailand
| | | | - Marwah Suliman Maashi
- Stem Cells and Regenerative Medicine Unit at King Fahad Medical Research Centre, Jeddah, Saudi Arabia
| |
Collapse
|
13
|
Li A, Guo F, Pan Q, Chen S, Chen J, Liu HF, Pan Q. Mesenchymal Stem Cell Therapy: Hope for Patients With Systemic Lupus Erythematosus. Front Immunol 2021; 12:728190. [PMID: 34659214 PMCID: PMC8516390 DOI: 10.3389/fimmu.2021.728190] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/14/2021] [Indexed: 12/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease. Although previous studies have demonstrated that SLE is related to the imbalance of cells in the immune system, including B cells, T cells, and dendritic cells, etc., the mechanisms underlying SLE pathogenesis remain unclear. Therefore, effective and low side-effect therapies for SLE are lacking. Recently, mesenchymal stem cell (MSC) therapy for autoimmune diseases, particularly SLE, has gained increasing attention. This therapy can improve the signs and symptoms of refractory SLE by promoting the proliferation of Th2 and Treg cells and inhibiting the activity of Th1, Th17, and B cells, etc. However, MSC therapy is also reported ineffective in some patients with SLE, which may be related to MSC- or patient-derived factors. Therefore, the therapeutic effects of MSCs should be further confirmed. This review summarizes the status of MSC therapy in refractory SLE treatment and potential reasons for the ineffectiveness of MSC therapy from three perspectives. We propose various MSC modification methods that may be beneficial in enhancing the immunosuppression of MSCs in SLE. However, their safety and protective effects in patients with SLE still need to be confirmed by further experimental and clinical evidence.
Collapse
Affiliation(s)
- Aifen Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fengbiao Guo
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Quanren Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuxian Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaxuan Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
14
|
Youssef JR, Boraie NA, Ibrahim HF, Ismail FA, El-Moslemany RM. Glibenclamide Nanocrystal-Loaded Bioactive Polymeric Scaffolds for Skin Regeneration: In Vitro Characterization and Preclinical Evaluation. Pharmaceutics 2021; 13:1469. [PMID: 34575545 PMCID: PMC8469322 DOI: 10.3390/pharmaceutics13091469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 01/02/2023] Open
Abstract
Skin restoration following full-thickness injury poses significant clinical challenges including inflammation and scarring. Medicated scaffolds formulated from natural bioactive polymers present an attractive platform for promoting wound healing. Glibenclamide was formulated in collagen/chitosan composite scaffolds to fulfill this aim. Glibenclamide was forged into nanocrystals with optimized colloidal properties (particle size of 352.2 nm, and polydispersity index of 0.29) using Kolliphor as a stabilizer to allow loading into the hydrophilic polymeric matrix. Scaffolds were prepared by the freeze drying method using different total polymer contents (3-6%) and collagen/chitosan ratios (0.25-2). A total polymer content of 3% at a collagen/chitosan ratio of 2:1 (SCGL3-2) was selected based on the results of in vitro characterization including the swelling index (1095.21), porosity (94.08%), mechanical strength, rate of degradation and in vitro drug release. SCGL3-2 was shown to be hemocompatible based on the results of protein binding, blood clotting and percentage hemolysis assays. In vitro cell culture studies on HSF cells demonstrated the biocompatibility of nanocrystals and SCGL3-2. In vivo studies on a rat model of a full-thickness wound presented rapid closure with enhanced histological and immunohistochemical parameters, revealing the success of the scaffold in reducing inflammation and promoting wound healing without scar formation. Hence, SCGL3-2 could be considered a potential dermal substitute for skin regeneration.
Collapse
Affiliation(s)
- Julie R. Youssef
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21523, Egypt; (J.R.Y.); (N.A.B.); (F.A.I.)
| | - Nabila A. Boraie
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21523, Egypt; (J.R.Y.); (N.A.B.); (F.A.I.)
| | - Heba F. Ibrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria 21523, Egypt;
| | - Fatma A. Ismail
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21523, Egypt; (J.R.Y.); (N.A.B.); (F.A.I.)
| | - Riham M. El-Moslemany
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21523, Egypt; (J.R.Y.); (N.A.B.); (F.A.I.)
| |
Collapse
|
15
|
Ashfaq R, Mehmood A, Ramzan A, Hussain I, Tarar MN, Riazuddin S. Antioxidant pretreatment enhances umbilical cord derived stem cells survival in response to thermal stress in vitro. Regen Med 2020; 15:1441-1453. [PMID: 32339058 DOI: 10.2217/rme-2019-0090] [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] [Indexed: 01/08/2023] Open
Abstract
Aim: Pretreatment of stem cells with antioxidants accelerates their ability to counter oxidative stress and is associated with the overall therapeutic outcome of their transplantation. Material & methods: Wharton Jelly derived mesenchymal stem cells (WJMSCs) were cultured and pretreated with various doses of antioxidants; Vitamin C (Vit C), Vitamin E (Vit E), Vitamin D3 (Vit D3) and their Cocktail, followed by exposure to in vitro heat injury. Assessment of WJMSCs survival, paracrine release, in vitro wound healing and expression of angiogenic and survival markers was conducted. Results: The results displayed an enhanced survival of WJMSCs especially in the case of Cocktail priming. Conclusion: Our data suggest that antioxidant pretreatment of WJMSCs strengthens the endurance of the cells, within stress conditions.
Collapse
Affiliation(s)
- Ramla Ashfaq
- Centre of Excellence in Molecular Biology (CEMB), 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Azra Mehmood
- Centre of Excellence in Molecular Biology (CEMB), 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Amna Ramzan
- Centre of Excellence in Molecular Biology (CEMB), 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan
| | - Intzar Hussain
- Department of Ophthalmology, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Moazzam Nazeer Tarar
- Department of Dermatology, Jinnah Burn & Reconstructive Surgery Centre, Lahore, Pakistan
| | - Sheikh Riazuddin
- Centre of Excellence in Molecular Biology (CEMB), 87-West Canal Bank Road, University of the Punjab, Lahore, Pakistan.,Department of Dermatology, Jinnah Burn & Reconstructive Surgery Centre, Lahore, Pakistan
| |
Collapse
|
16
|
Li M, Zhong L, He W, Ding Z, Hou Q, Zhao Y, Yuan J, Liu J, Zhu Z, Lu Q, Fu X. Concentrated Conditioned Medium-Loaded Silk Nanofiber Hydrogels with Sustained Release of Bioactive Factors To Improve Skin Regeneration. ACS APPLIED BIO MATERIALS 2019; 2:4397-4407. [PMID: 35021399 DOI: 10.1021/acsabm.9b00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Meirong Li
- Institute of Basic Medical Science, Wound Healing and Cell Biology Laboratory, Chinese PLA General Hospital, Beijing 100853, China
- Central Laboratory, Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya 572014, China
| | - Lingzhi Zhong
- Institute of Basic Medical Science, Wound Healing and Cell Biology Laboratory, Chinese PLA General Hospital, Beijing 100853, China
| | - Wenjun He
- Institute of Basic Medical Science, Wound Healing and Cell Biology Laboratory, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhaozhao Ding
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, and School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, People’s Republic of China
| | - Qian Hou
- Institute of Basic Medical Science, Wound Healing and Cell Biology Laboratory, Chinese PLA General Hospital, Beijing 100853, China
| | - Yali Zhao
- Central Laboratory, Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya 572014, China
| | - Jifang Yuan
- Institute of Basic Medical Science, Wound Healing and Cell Biology Laboratory, Chinese PLA General Hospital, Beijing 100853, China
| | - Jiejie Liu
- Institute of Basic Medical Science, Wound Healing and Cell Biology Laboratory, Chinese PLA General Hospital, Beijing 100853, China
| | - Ziying Zhu
- Institute of Basic Medical Science, Wound Healing and Cell Biology Laboratory, Chinese PLA General Hospital, Beijing 100853, China
| | - Qiang Lu
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, and School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, People’s Republic of China
| | - Xiaobing Fu
- Institute of Basic Medical Science, Wound Healing and Cell Biology Laboratory, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
17
|
Burtenshaw D, Kitching M, Redmond EM, Megson IL, Cahill PA. Reactive Oxygen Species (ROS), Intimal Thickening, and Subclinical Atherosclerotic Disease. Front Cardiovasc Med 2019; 6:89. [PMID: 31428618 PMCID: PMC6688526 DOI: 10.3389/fcvm.2019.00089] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
Arteriosclerosis causes significant morbidity and mortality worldwide. Central to this process is the development of subclinical non-atherosclerotic intimal lesions before the appearance of pathologic intimal thickening and advanced atherosclerotic plaques. Intimal thickening is associated with several risk factors, including oxidative stress due to reactive oxygen species (ROS), inflammatory cytokines and lipid. The main ROS producing systems in-vivo are reduced nicotinamide dinucleotide phosphate (NADPH) oxidase (NOX). ROS effects are context specific. Exogenous ROS induces apoptosis and senescence, whereas intracellular ROS promotes stem cell differentiation, proliferation, and migration. Lineage tracing studies using murine models of subclinical atherosclerosis have revealed the contributory role of medial smooth muscle cells (SMCs), resident vascular stem cells, circulating bone-marrow progenitors and endothelial cells that undergo endothelial-mesenchymal-transition (EndMT). This review will address the putative physiological and patho-physiological roles of ROS in controlling vascular cell fate and ROS contribution to vascular regeneration and disease progression.
Collapse
Affiliation(s)
- Denise Burtenshaw
- Vascular Biology & Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| | | | - Eileen M Redmond
- Department of Surgery, University of Rochester, Rochester, NY, United States
| | - Ian L Megson
- Centre for Health Science, UHI Institute of Health Research and Innovation, Inverness, United Kingdom
| | - Paul A Cahill
- Vascular Biology & Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
18
|
Shojaei F, Rahmati S, Banitalebi Dehkordi M. A review on different methods to increase the efficiency of mesenchymal stem cell-based wound therapy. Wound Repair Regen 2019; 27:661-671. [PMID: 31298446 DOI: 10.1111/wrr.12749] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) accelerate wound healing but the harsh environment of wound site limits the engraftment, retention, and survival rate of transplanted cells. There are multiple approaches that amplify the therapeutic potential of MSCs. The MSCs derived from medical waste material, provide comparable regenerative abilities compared to traditional sources. The application of different scaffolds increases MSC delivery and migration into the wound. The spheroid culture of MSC increases the paracrine effects of the entrapped cells and the secretion of pro-angiogenic and anti-inflammatory cytokines. The MSC pretreating and preconditioning enhances the cell migration, proliferation, and survival rate, which lead to higher angiogenesis, re-epithelialization, wound closure, and granulation tissue formation. Moreover, genetic modification has been performed in order to increase MSC angiogenesis, differentiation potential, as well as the cell life span. Herein, we review the results of aforementioned approaches and provide information accommodating to the continued development of MSC-based wound therapy in the future.
Collapse
Affiliation(s)
- Fereshteh Shojaei
- Department of Medical biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shima Rahmati
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mehdi Banitalebi Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
19
|
Fu X, Liu G, Halim A, Ju Y, Luo Q, Song AG. Mesenchymal Stem Cell Migration and Tissue Repair. Cells 2019; 8:E784. [PMID: 31357692 PMCID: PMC6721499 DOI: 10.3390/cells8080784] [Citation(s) in RCA: 556] [Impact Index Per Article: 92.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/13/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multilineage cells with the ability to self-renew and differentiate into a variety of cell types, which play key roles in tissue healing and regenerative medicine. Bone marrow-derived mesenchymal stem cells (BMSCs) are the most frequently used stem cells in cell therapy and tissue engineering. However, it is prerequisite for BMSCs to mobilize from bone marrow and migrate into injured tissues during the healing process, through peripheral circulation. The migration of BMSCs is regulated by mechanical and chemical factors in this trafficking process. In this paper, we review the effects of several main regulatory factors on BMSC migration and its underlying mechanism; discuss two critical roles of BMSCs-namely, directed differentiation and the paracrine function-in tissue repair; and provide insight into the relationship between BMSC migration and tissue repair, which may provide a better guide for clinical applications in tissue repair through the efficient regulation of BMSC migration.
Collapse
Affiliation(s)
- Xiaorong Fu
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China
| | - Ge Liu
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China
| | - Alexander Halim
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China
| | - Yang Ju
- Department of Mechanical Science and Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Qing Luo
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China
| | - And Guanbin Song
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China.
| |
Collapse
|
20
|
Mizukami A, Thomé CH, Ferreira GA, Lanfredi GP, Covas DT, Pitteri SJ, Swiech K, Faça VM. Proteomic Identification and Time-Course Monitoring of Secreted Proteins During Expansion of Human Mesenchymal Stem/Stromal in Stirred-Tank Bioreactor. Front Bioeng Biotechnol 2019; 7:154. [PMID: 31297369 PMCID: PMC6607109 DOI: 10.3389/fbioe.2019.00154] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/10/2019] [Indexed: 12/14/2022] Open
Abstract
The therapeutic potential of mesenchymal stem/stromal cells (MSC) is widely recognized for the treatment of several diseases, including acute graft-vs.-host disease (GVHD), hematological malignancies, cardiovascular, bone, and cartilage diseases. More recently, this therapeutic efficacy has been attributed to the bioactive molecules that these cells secrete (secretome), now being referred as medicinal signaling cells. This fact raises the opportunity of therapeutically using MSC-derived soluble factors rather than cells themselves, enabling their translation into the clinic. Indeed, many clinical trials are now studying the effects of MSC-secretome in the context of cell-free therapy. MSC secretome profile varies between donors, source, and culture conditions, making their therapeutic use very challenging. Therefore, identifying these soluble proteins and evaluating their production in a reproducible and scalable manner is even more relevant. In this work, we analyzed the global profile of proteins secreted by umbilical cord matrix (UCM) derived-MSC in static conditions by using mass spectrometry, enabling the identification of thousands of proteins. Afterwards, relevant proteins were chosen and monitored in the supernatant of a fully-controllable, closed and scalable system (bioreactor) by using multiple reaction monitoring (MRM) mass spectrometric technique in a time-dependent manner. The results showed that the majority of interesting proteins were enriched through time in culture, with the last day of culture being the ideal time for supernatant collection. The use of this regenerative "soup," which is frequently discarded, could represent a step toward a safe, robust and reproducible cell-free product to be used in the medical therapeutic field. The future use of chemically defined culture-media will certainly facilitate secretome production according to Good Manufacturing Practice (GMP) standards.
Collapse
Affiliation(s)
- Amanda Mizukami
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Carolina Hassibe Thomé
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Germano Aguiar Ferreira
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Guilherme Pauperio Lanfredi
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Sharon J Pitteri
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Stanford, CA, United States
| | - Kamilla Swiech
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Vitor Marcel Faça
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
21
|
Qin G, Song Y, Guo Y, Sun Y, Zeng W. LincRNA TINCR facilitates excessive proliferation and inflammation in post-burn skin fibroblasts by directly binding with SND1 protein and inducing SND1-mediated TGF-β1 expression. Biochem Biophys Res Commun 2019; 509:903-910. [DOI: 10.1016/j.bbrc.2019.01.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 01/04/2019] [Indexed: 10/27/2022]
|
22
|
Hu Y, Zhu Y, Lian N, Chen M, Bartke A, Yuan R. Metabolic Syndrome and Skin Diseases. Front Endocrinol (Lausanne) 2019; 10:788. [PMID: 31824416 PMCID: PMC6880611 DOI: 10.3389/fendo.2019.00788] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
The increasing prevalence of Metabolic syndrome (MetS) is a worldwide health problem, and the association between MetS and skin diseases has recently attracted growing attention. In this review, we summarize the associations between MetS and skin diseases, such as psoriasis, acne vulgaris, hidradenitis suppurativa, androgenetic alopecia, acanthosis nigricans, and atopic dermatitis. To discuss the potential common mechanisms underlying MetS and skin diseases, we focus on insulin signaling and insulin resistance, as well as chronic inflammation including adipokines and proinflammatory cytokines related to molecular mechanisms. A better understanding of the relationship between MetS and skin diseases contributes to early diagnosis and prevention, as well as providing clues for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Yu Hu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and Sexually Transmitted Infections, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Yun Zhu
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Ni Lian
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and Sexually Transmitted Infections, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Min Chen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and Sexually Transmitted Infections, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- *Correspondence: Min Chen
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Rong Yuan
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
- Rong Yuan
| |
Collapse
|
23
|
Cárdenes N, Álvarez D, Sellarés J, Peng Y, Corey C, Wecht S, Nouraie SM, Shanker S, Sembrat J, Bueno M, Shiva S, Mora AL, Rojas M. Senescence of bone marrow-derived mesenchymal stem cells from patients with idiopathic pulmonary fibrosis. Stem Cell Res Ther 2018; 9:257. [PMID: 30257725 PMCID: PMC6158816 DOI: 10.1186/s13287-018-0970-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/17/2018] [Accepted: 08/05/2018] [Indexed: 12/14/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease for which age is the most important risk factor. Different mechanisms associated with aging, including stem cell dysfunction, have been described to participate in the pathophysiology of IPF. We observed an extrapulmonary effect associated with IPF: increase in cell senescence of bone marrow-derived mesenchymal stem cells (B-MSCs). Methods B-MSCs were obtained from vertebral bodies procured from IPF patients and age-matched normal controls. Cell senescence was determined by cell proliferation and expression of markers of cell senescence p16INK4A, p21, and β-galactosidase activity. Mitochondrial function and DNA damage were measured. Paracrine induction of senescence and profibrotic responses were analyzed in vitro using human lung fibroblasts. The reparative capacity of B-MSCs was examined in vivo using the bleomycin-induced lung fibrosis model. Results In our study, we demonstrate for the first time that B-MSCs from IPF patients are senescent with significant differences in mitochondrial function, with accumulation of DNA damage resulting in defects in critical cell functions when compared with age-matched controls. Senescent IPF B-MSCs have the capability of paracrine senescence by inducing senescence in normal-aged fibroblasts, suggesting a possible link between senescent B-MSCs and the late onset of the disease. IPF B-MSCs also showed a diminished capacity to migrate and were less effective in preventing fibrotic changes observed in mice after bleomycin-induced injury, increasing illness severity and proinflammatory responses. Conclusions We describe extrapulmonary alterations in B-MSCs from IPF patients. The consequences of having senescent B-MSCs are not completely understood, but the decrease in their ability to respond to normal activation and the risk of having a negative impact on the local niche by inducing inflammation and senescence in the neighboring cells suggests a new link between B-MSC and the onset of the disease. Electronic supplementary material The online version of this article (10.1186/s13287-018-0970-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nayra Cárdenes
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, W1244 BST Tower 200 Lothrop Street, Pittsburgh, PA, 15261, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Diana Álvarez
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, W1244 BST Tower 200 Lothrop Street, Pittsburgh, PA, 15261, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jacobo Sellarés
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, W1244 BST Tower 200 Lothrop Street, Pittsburgh, PA, 15261, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Interstitial Lung Disease Program, Hospital Clinic, Barcelona, Spain
| | - Yating Peng
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, W1244 BST Tower 200 Lothrop Street, Pittsburgh, PA, 15261, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Research Unit of Respiratory Diseases, Central South University, Changsha, 410011, Hunan, China
| | - Catherine Corey
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Vascular Medicine Institute of the University of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sophie Wecht
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, W1244 BST Tower 200 Lothrop Street, Pittsburgh, PA, 15261, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Seyed Mehdi Nouraie
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, W1244 BST Tower 200 Lothrop Street, Pittsburgh, PA, 15261, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Swaroop Shanker
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, W1244 BST Tower 200 Lothrop Street, Pittsburgh, PA, 15261, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John Sembrat
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, W1244 BST Tower 200 Lothrop Street, Pittsburgh, PA, 15261, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marta Bueno
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Vascular Medicine Institute of the University of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sruti Shiva
- Vascular Medicine Institute of the University of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana L Mora
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Vascular Medicine Institute of the University of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mauricio Rojas
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, W1244 BST Tower 200 Lothrop Street, Pittsburgh, PA, 15261, USA. .,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. .,Vascular Medicine Institute of the University of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
24
|
Mariñas-Pardo L, García-Castro J, Rodríguez-Hurtado I, Rodríguez-García MI, Núñez-Naveira L, Hermida-Prieto M. Allogeneic Adipose-Derived Mesenchymal Stem Cells (Horse Allo 20) for the Treatment of Osteoarthritis-Associated Lameness in Horses: Characterization, Safety, and Efficacy of Intra-Articular Treatment. Stem Cells Dev 2018; 27:1147-1160. [DOI: 10.1089/scd.2018.0074] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
| | - Javier García-Castro
- Unidad de Biotecnología Celular, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | | | | | | |
Collapse
|
25
|
Liubaviciute A, Kaseta V, Vaitkuviene A, Mackiewicz Z, Biziuleviciene G. Regenerative potential of partially differentiated mesenchymal stromal cells in a mouse model of a full-thickness skin wound. EXCLI JOURNAL 2018; 17:871-888. [PMID: 30233286 PMCID: PMC6141819 DOI: 10.17179/excli2018-1504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/26/2018] [Indexed: 12/18/2022]
Abstract
Mesenchymal stromal cells (MSCs, known as mesenchymal stem cells) are considered to be a promising therapeutic tool for many diseases. But it is still unclear which cells are more efficient and safe for wound healing and tissue regeneration for clinical applications: undifferentiated, partially differentiated stem cells or differentiated cells. In this study, we modified MSCs with keratinocyte-conditioned medium (KCM) and examined MSCs, partially differentiated MSCs (PMSCs) and differentiated cell migration, accumulation in the wounded area as well as cell regenerative efficiency in a full-thickness skin wound model. In addition to that, the impact of intradermal and intravenous cell delivery methods of wound healing was evaluated. C57BL/6J mouse compact bone MSCs were treated with a KCM for 14 days. Flow cytometry analysis showed the appearance of keratinocyte surface markers which were absent in MSCs, whereas the specific markers for MSCs were lost. Cells were injected either intravenously or intradermally in C57BL/6J mice. Wound closure, cell migration and accumulation in the wounded area were further analysed. Wound healing was assessed by the rate of wound closure and by histological evaluation. Cells were monitored using optical imaging. We demonstrated that PMSCs showed morphology similar to keratinocyte cells, had enhanced migration and increased survival at the site of injury. PMSCs had a beneficial effect on wound healing and tissue regeneration. This effect was reinforced when these cells were injected intravenously. Due to their partial differentiation status, we assume that PMSCs can differentiate more rapidly into epidermal cell lineages thus causing faster and qualitatively improved wound healing.
Collapse
Affiliation(s)
- Ausra Liubaviciute
- State Research Institute Centre for Innovative Medicine, Department of Stem Cell Biology, Santariskiu str. 5, LT-08406 Vilnius, Lithuania
| | - Vytautas Kaseta
- State Research Institute Centre for Innovative Medicine, Department of Stem Cell Biology, Santariskiu str. 5, LT-08406 Vilnius, Lithuania
| | - Aida Vaitkuviene
- State Research Institute Centre for Innovative Medicine, Department of Stem Cell Biology, Santariskiu str. 5, LT-08406 Vilnius, Lithuania
| | - Zygmunt Mackiewicz
- State Research Institute Centre for Innovative Medicine, Department of Regenerative Medicine, Santariskiu str. 5, LT-08406 Vilnius, Lithuania
| | - Gene Biziuleviciene
- State Research Institute Centre for Innovative Medicine, Department of Stem Cell Biology, Santariskiu str. 5, LT-08406 Vilnius, Lithuania
| |
Collapse
|
26
|
Dou L, Yan Q, Liang P, Zhou P, Zhang Y, Ji P. iTRAQ-Based Proteomic Analysis Exploring the Influence of Hypoxia on the Proteome of Dental Pulp Stem Cells under 3D Culture. Proteomics 2018; 18. [PMID: 29327447 DOI: 10.1002/pmic.201700215] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/21/2017] [Indexed: 12/14/2022]
Abstract
Hypoxic preconditioning is commonly applied to enhance mesenchymal stem cells (MSCs) therapeutic effect before transplantation. Elucidating the effect of hypoxic preconditioning would be beneficial for improved application. However, the influence of hypoxia on dental tissue derived MSCs cultured in 3D was unknown. Thus, the present study is to investigate gene expression and proteome of dental pulp stem cells (DPSCs) after hypoxic preconditioning. DPSCs were isolated, cultured in a 3D system under the normoxic and hypoxic conditions. The gene expression was examined with reverse transcription polymerase chain reaction, and the proteome was analyzed using iTRAQ-based mass spectrometry. The expressions of HIF-1α, VEGFA, KDR at mRNA level was upregulated while BMP-2 was downregulated. Two thousand one hundred and fifteen proteins were identified and 57 proteins exhibited significant differences after hypoxic preconditioning (30 up-regulated, 27 down-regulated). Bioinformatic analysis revealed the majority of up-regulated proteins are involved in cellular process, angiogenesis, protein binding and transport, regulation of response to stimulus, metabolic processes, and immune response. Increased IL-6 and decreased TGF-1β protein expression under hypoxic condition were verified by ELISA. Hypoxic preconditioning partly affected the gene and protein expression in DPSCs under 3D culture and may enhance the efficacy of MSCs transplantation.
Collapse
Affiliation(s)
- Lei Dou
- Stomatological hospital of Chongqing medical university, Chongqing, P. R. China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, P. R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, P. R. China
| | - Qifang Yan
- Stomatological hospital of Chongqing medical university, Chongqing, P. R. China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, P. R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, P. R. China
| | - Panpan Liang
- Stomatological hospital of Chongqing medical university, Chongqing, P. R. China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, P. R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, P. R. China
| | - Pengfei Zhou
- Stomatological hospital of Chongqing medical university, Chongqing, P. R. China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, P. R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, P. R. China
| | - Yan Zhang
- Stomatological hospital of Chongqing medical university, Chongqing, P. R. China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, P. R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, P. R. China
| | - Ping Ji
- Stomatological hospital of Chongqing medical university, Chongqing, P. R. China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, P. R. China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, P. R. China
| |
Collapse
|
27
|
Zhen C, Zhu H, Li Q, Xu W. Protective effects of mesenchymal stem cell cond tional medium against inflammatory injury on human gingival fibroblast. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8263-8269. [PMID: 31966677 PMCID: PMC6965482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/20/2017] [Indexed: 06/10/2023]
Abstract
Mesenchymal stem cells (MSCs) alleviate oxidative stress in kidney, lung and heart by secreting bioactive factors. Human gingival fibroblasts (HGFs) are important for repairing periodontal tissues. This study observed protective effects of MSCs conditional medium (MSCCM) on HGFs against inflammatory injury, to investigate anti-inflammatory function and mechanism of MSCCM on HGFs. Primary cultured HGFs were identified for sources by immunohistochemistry (IHC)-SP assay. In vitro cultured MSCs were extracted for MSCCM, which was used to pre-treat HGFs with inflammatory induction by 10 μg/L IL-6 or TNF-α for 4 h. ELISA quantified transforming growth factor (TGF)-β level in the supernatant, and superoxide dismutase (SOD) plus malondialdehyde (MDA) levels were measured by colorimetry. RT-PCR measured keratocyte growth factor (KGF) mRNA expression, and flow cytometry or Western blot measured apoptosis or Caspase-3 expression in HGFs, respectively. Compared to control group, MSCCM treatment group showed no significant change of SOD, MDA, TGF-β level, cell apoptosis, KGF mRNA or Caspase-3 expression (P>0.05). Inflammation treatment elevated all those indexes but decreased SOD (P<0.05). Compared to model group, MSCCM treatment further decreased these factors but increased SOD level (P<0.05). No significant difference was found between IL-6 and TNF-α treated cells. MSCCM can partially inhibit IL-6 induced inflammatory injury of HGFs via suppressing Caspase-3 and KGF expression.
Collapse
Affiliation(s)
- Changhao Zhen
- Department of Stomatology, Affiliated Hospital of Jilin Medical UniversityJilin, China
| | - Haiyu Zhu
- Department of Stomatology, Affiliated Hospital of Jilin Medical UniversityJilin, China
| | - Qiaoling Li
- Department of Stomatology, Hongqi Hospital Affiliated to Mudanjiang Medical UniversityMudanjiang, Heilongjiang, China
| | - Wenxiu Xu
- Department of Stomatology, Hongqi Hospital Affiliated to Mudanjiang Medical UniversityMudanjiang, Heilongjiang, China
| |
Collapse
|
28
|
Menter DG, Kopetz S, Hawk E, Sood AK, Loree JM, Gresele P, Honn KV. Platelet "first responders" in wound response, cancer, and metastasis. Cancer Metastasis Rev 2017; 36:199-213. [PMID: 28730545 PMCID: PMC5709140 DOI: 10.1007/s10555-017-9682-0] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Platelets serve as "first responders" during normal wounding and homeostasis. Arising from bone marrow stem cell lineage megakaryocytes, anucleate platelets can influence inflammation and immune regulation. Biophysically, platelets are optimized due to size and discoid morphology to distribute near vessel walls, monitor vascular integrity, and initiate quick responses to vascular lesions. Adhesion receptors linked to a highly reactive filopodia-generating cytoskeleton maximizes their vascular surface contact allowing rapid response capabilities. Functionally, platelets normally initiate rapid clotting, vasoconstriction, inflammation, and wound biology that leads to sterilization, tissue repair, and resolution. Platelets also are among the first to sense, phagocytize, decorate, or react to pathogens in the circulation. These platelet first responder properties are commandeered during chronic inflammation, cancer progression, and metastasis. Leaky or inflammatory reaction blood vessel genesis during carcinogenesis provides opportunities for platelet invasion into tumors. Cancer is thought of as a non-healing or chronic wound that can be actively aided by platelet mitogenic properties to stimulate tumor growth. This growth ultimately outstrips circulatory support leads to angiogenesis and intravasation of tumor cells into the blood stream. Circulating tumor cells reengage additional platelets, which facilitates tumor cell adhesion, arrest and extravasation, and metastasis. This process, along with the hypercoagulable states associated with malignancy, is amplified by IL6 production in tumors that stimulate liver thrombopoietin production and elevates circulating platelet numbers by thrombopoiesis in the bone marrow. These complex interactions and the "first responder" role of platelets during diverse physiologic stresses provide a useful therapeutic target that deserves further exploration.
Collapse
Affiliation(s)
- David G Menter
- Department of Gastrointestinal Medical Oncology, M. D. Anderson Cancer Center, Room#: FC10.3004, 1515 Holcombe Boulevard--Unit 0426, Houston, TX, 77030, USA.
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, M. D. Anderson Cancer Center, Room#: FC10.3004, 1515 Holcombe Boulevard--Unit 0426, Houston, TX, 77030, USA
| | - Ernest Hawk
- Office of the Vice President Cancer Prevention & Population Science, M. D. Anderson Cancer Center, Unit 1370, 1515 Holcombe Boulevard, Houston, TX, 77054, USA
| | - Anil K Sood
- Gynocologic Oncology & Reproductive Medicine, M. D. Anderson Cancer Center, Unit 1362, 1515 Holcombe Boulevard, Houston, TX, 77054, USA
- Department of Cancer Biology, M. D. Anderson Cancer Center, Unit 1362, 1515 Holcombe Boulevard, Houston, TX, 77054, USA
- Center for RNA Interference and Non-Coding RNA The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Jonathan M Loree
- Department of Gastrointestinal Medical Oncology, M. D. Anderson Cancer Center, Room#: FC10.3004, 1515 Holcombe Boulevard--Unit 0426, Houston, TX, 77030, USA
| | - Paolo Gresele
- Department of Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Via E. Dal Pozzo, 06126, Perugia, Italy
| | - Kenneth V Honn
- Bioactive Lipids Research Program, Department of Pathology, Wayne State University, 431 Chemistry Bldg, 5101 Cass Avenue, Detroit, MI, 48202, USA
- Department of Pathology, Wayne State University, 431 Chemistry Bldg, 5101 Cass Avenue, Detroit, MI, 48202, USA
- Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, 5101 Cass Avenue, Detroit, MI, 48202, USA
| |
Collapse
|