1
|
Siu WS, Ma H, Leung PC. Review on Current Advancements in Facilitation of Burn Wound Healing. Bioengineering (Basel) 2025; 12:428. [PMID: 40281787 PMCID: PMC12024970 DOI: 10.3390/bioengineering12040428] [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: 03/12/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Burns are common injuries, but their treatment remains challenging due to the complex nature of the wound healing process. Burn wounds are classified into different categories based on their size and depth. Treatment modalities vary significantly across these categories, primarily focusing on the inflammation, proliferation, and remodeling phases of burn wound healing. This review summarizes current research on various approaches to enhance burn wound recovery, including advancements in wound dressings, the use of platelet-rich plasma, stem cells, their soluble factors primarily in the form of secretomes or extracellular vesicles, and nano-technologies. Additionally, advancements in modernized traditional medicine are discussed to give a new aspect for burn wound healing. This review also summarizes the barriers in translating bench research to clinical practice in burn wound treatment methods. For an effective translation, researchers and industrial partners should work more closely, while regulatory bodies should streamline the approval procedure.
Collapse
Affiliation(s)
- Wing Sum Siu
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; (H.M.); (P.C.L.)
| | | | | |
Collapse
|
2
|
Yamawaki-Ogata A, Mutsuga M, Narita Y. A review of current status of cell-based therapies for aortic aneurysms. Inflamm Regen 2023; 43:40. [PMID: 37544997 PMCID: PMC10405412 DOI: 10.1186/s41232-023-00280-8] [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: 02/25/2023] [Accepted: 05/18/2023] [Indexed: 08/08/2023] Open
Abstract
An aortic aneurysm (AA) is defined as focal aortic dilation that occurs mainly with older age and with chronic inflammation associated with atherosclerosis. The aneurysmal wall is a complex inflammatory environment characterized by endothelial dysfunction, macrophage activation, vascular smooth muscle cell (VSMC) apoptosis, and the production of proinflammatory molecules and matrix metalloproteases (MMPs) secreted by infiltrated inflammatory cells such as macrophages, T and B cells, dendritic cells, neutrophils, mast cells, and natural killer cells. To date, a considerable number of studies have been conducted on stem cell research, and growing evidence indicates that inflammation and tissue repair can be controlled through the functions of stem/progenitor cells. This review summarizes current cell-based therapies for AA, involving mesenchymal stem cells, VSMCs, multilineage-differentiating stress-enduring cells, and anti-inflammatory M2 macrophages. These cells produce beneficial outcomes in AA treatment by modulating the inflammatory environment, including decreasing the activity of proinflammatory molecules and MMPs, increasing anti-inflammatory molecules, modulating VSMC phenotypes, and preserving elastin. This article also describes detailed studies on pathophysiological mechanisms and the current progress of clinical trials.
Collapse
Affiliation(s)
- Aika Yamawaki-Ogata
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Masato Mutsuga
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yuji Narita
- Department of Cardiac Surgery, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| |
Collapse
|
3
|
Peshkova M, Korneev A, Suleimanov S, Vlasova II, Svistunov A, Kosheleva N, Timashev P. MSCs' conditioned media cytokine and growth factor profiles and their impact on macrophage polarization. Stem Cell Res Ther 2023; 14:142. [PMID: 37231519 DOI: 10.1186/s13287-023-03381-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND There is a growing body of evidence that multipotent mesenchymal stromal cells' (MSCs') remarkable therapeutic potential is attributed not only to their differentiation and regenerative capacity, but also to the paracrine effect, underlying their immunomodulatory properties. MSCs' secretome (i.e., cytokines, growth factors, and extracellular vesicles) is therefore increasingly discussed in the context of their ability to modulate inflammatory response and promote regeneration. There is evidence that 2D or 3D culturing conditions have an impact on the cells' secretome, and here we aimed to compare the secretion of cytokines and growth factors in human MSCs from different sources cultured in 2D and 3D conditions and assess their effect on human macrophages polarization in vitro. METHODS MSCs were derived from human adipose tissue, bone marrow, gingiva, placenta, and umbilical cord, cultured as monolayers or as cell spheroids. Their cytokine profiles were analyzed, and data standardization was carried out using a z-score. Human peripheral blood mononuclear cells-derived macrophages were then treated with umbilical cord-derived MSCs' conditioned media and their effect on macrophages polarization was assessed. RESULTS Our findings suggest that umbilical cord-derived MSCs' conditioned media demonstrated the highest cytokine and growth factor levels and despite mostly pro-inflammatory cytokine profile were able to promote anti-inflammatory macrophage polarization. CONCLUSIONS Umbilical cord-derived MSCs' conditioned media hold great potential for therapeutic use, demonstrating significant anti-inflammatory effect on human macrophages.
Collapse
Affiliation(s)
- Maria Peshkova
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia, 119991
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia, 119991
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia, 119991
| | - Alexander Korneev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia, 119991
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia, 119991
- Laboratory of the Polymers Synthesis for Medical Applications, Sechenov University, Moscow, Russia, 119991
| | - Shakir Suleimanov
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia, 119991
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia, 119991
| | - Irina I Vlasova
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia, 119991
| | - Andrey Svistunov
- Sechenov First Moscow State Medical University, Moscow, Russia, 119991
| | - Nastasia Kosheleva
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia, 119991
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia, 119991
- FSBSI Institute of General Pathology and Pathophysiology, Moscow, Russia, 125315
| | - Peter Timashev
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia, 119991.
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia, 119991.
- Laboratory of Clinical Smart Nanotechnologies, Sechenov University, Moscow, Russia, 119991.
| |
Collapse
|
4
|
Mesenchymal stem cells and macrophages and their interactions in tendon-bone healing. J Orthop Translat 2023; 39:63-73. [PMID: 37188000 PMCID: PMC10175706 DOI: 10.1016/j.jot.2022.12.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/24/2022] [Accepted: 12/29/2022] [Indexed: 01/21/2023] Open
Abstract
Tendon-bone insertion injuries (TBI), such as anterior cruciate ligament (ACL) and rotator cuff injuries, are common degenerative or traumatic pathologies with a negative impact on the patient's daily life, and they cause huge economic losses every year. The healing process after an injury is complex and is dependent on the surrounding environment. Macrophages accumulate during the entire process of tendon and bone healing and their phenotypes progressively transform as they regenerate. As the "sensor and switch of the immune system", mesenchymal stem cells (MSCs) respond to the inflammatory environment and exert immunomodulatory effects during the tendon-bone healing process. When exposed to appropriate stimuli, they can differentiate into different tissues, including chondrocytes, osteocytes, and epithelial cells, promoting reconstruction of the complex transitional structure of the enthesis. It is well known that MSCs and macrophages communicate with each other during tissue repair. In this review, we discuss the roles of macrophages and MSCs in TBI injury and healing. Reciprocal interactions between MSCs and macrophages and some biological processes utilizing their mutual relations in tendon-bone healing are also described. Additionally, we discuss the limitations in our understanding of tendon-bone healing and propose feasible ways to exploit MSC-macrophage interplay to develop an effective therapeutic strategy for TBI injuries. The Translational potential of this article This paper reviewed the important functions of macrophages and mesenchymal stem cells in tendon-bone healing and described the reciprocal interactions between them during the healing process. By managing macrophage phenotypes, mesenchymal stem cells and the interactions between them, some possible novel therapies for tendon-bone injury may be proposed to promote tendon-bone healing after restoration surgery.
Collapse
|
5
|
Yang CH, Lin DY, Lin YS, Hsu CY, Tung MC, Tan KT, Ou YC. The Immunological Microenvironment and the Emerging Role of Stem Cells Therapy in Peyronie's Disease: A Systematic Narrative Review. Int J Mol Sci 2023; 24:ijms24010777. [PMID: 36614220 PMCID: PMC9821411 DOI: 10.3390/ijms24010777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/25/2022] [Accepted: 12/26/2022] [Indexed: 01/03/2023] Open
Abstract
Current literature has indicated that Peyronie's disease (PD) could be initiated by microtrauma and the subsequent inflammation episodes that follow. PD could be sorted into acute or chronic status, and it can differ when selecting the clinical therapeutics. PD would cause pain and penile deformity to diseased men and impair their erectile function. Occasionally, surgical revision of the penis might be needed to correct the penile curvature. We find that there are limited effective options of intra-lesion injections for the PD plaques. By searching the databases and screening the literature with the PRISMA 2020 guideline, we observed that several preclinical studies that applied stem cell therapy in treating PD were fruitful in the acute phase. Although in the chronic phase of PD, erectile parameters were not significantly improved, and therefore, future studies might be better elevated in certain aspects, such as the sites selected for harvesting stem cells or changing the centrifugation forces. In this review, we concluded the contemporary understanding of inflammatory microenvironments in PD, the stem cell therapy in PD, and our perspectives on future studies. We concluded that there may be great potential in stem cell therapy for treating both acute and chronic phases PD.
Collapse
Affiliation(s)
- Che-Hsueh Yang
- Division of Urology, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
| | - Dian-Yu Lin
- Division of Urology, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
- Joshua Taipei Hernia Center, Central Clinic & Hospital, Taipei 106, Taiwan
- Department of Urology, College of Medicine and Shu-Tien Urological Research Center, National Yang Ming Chiao Tung University, Taipei 106, Taiwan
| | - Yi-Sheng Lin
- Division of Urology, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
| | - Chao-Yu Hsu
- Division of Urology, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
| | - Min-Che Tung
- Division of Urology, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
| | - Kok-Tong Tan
- Division of General Surgery, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
- Correspondence: (K.-T.T.); (Y.-C.O.)
| | - Yen-Chuan Ou
- Division of Urology, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 435, Taiwan
- Correspondence: (K.-T.T.); (Y.-C.O.)
| |
Collapse
|
6
|
S S, Dahal S, Bastola S, Dayal S, Yau J, Ramamurthi A. Stem Cell Based Approaches to Modulate the Matrix Milieu in Vascular Disorders. Front Cardiovasc Med 2022; 9:879977. [PMID: 35783852 PMCID: PMC9242410 DOI: 10.3389/fcvm.2022.879977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/20/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) represents a complex and dynamic framework for cells, characterized by tissue-specific biophysical, mechanical, and biochemical properties. ECM components in vascular tissues provide structural support to vascular cells and modulate their function through interaction with specific cell-surface receptors. ECM–cell interactions, together with neurotransmitters, cytokines, hormones and mechanical forces imposed by blood flow, modulate the structural organization of the vascular wall. Changes in the ECM microenvironment, as in post-injury degradation or remodeling, lead to both altered tissue function and exacerbation of vascular pathologies. Regeneration and repair of the ECM are thus critical toward reinstating vascular homeostasis. The self-renewal and transdifferentiating potential of stem cells (SCs) into other cell lineages represents a potentially useful approach in regenerative medicine, and SC-based approaches hold great promise in the development of novel therapeutics toward ECM repair. Certain adult SCs, including mesenchymal stem cells (MSCs), possess a broader plasticity and differentiation potential, and thus represent a viable option for SC-based therapeutics. However, there are significant challenges to SC therapies including, but not limited to cell processing and scaleup, quality control, phenotypic integrity in a disease milieu in vivo, and inefficient delivery to the site of tissue injury. SC-derived or -inspired strategies as a putative surrogate for conventional cell therapy are thus gaining momentum. In this article, we review current knowledge on the patho-mechanistic roles of ECM components in common vascular disorders and the prospects of developing adult SC based/inspired therapies to modulate the vascular tissue environment and reinstate vessel homeostasis in these disorders.
Collapse
|
7
|
Tran NT, Park IS, Truong MD, Park DY, Park SH, Min BH. Conditioned media derived from human fetal progenitor cells improves skin regeneration in burn wound healing. Cell Tissue Res 2022; 389:289-308. [PMID: 35624315 DOI: 10.1007/s00441-022-03638-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 05/11/2022] [Indexed: 01/06/2023]
Abstract
Stem cells are known to have excellent regenerative ability, which is primarily facilitated by indirect paracrine factors, rather than via direct cell replacement. The regenerative process is mediated by the release of extracellular matrix molecules, cytokines, and growth factors, which are also present in the media during cultivation. Herein, we aimed to demonstrate the functionality of key factors and mechanisms in skin regeneration through the analysis of conditioned media derived from fetal stem cells. A series of processes, including 3D pellet cultures, filtration and lyophilization is developed to fabricate human fetal cartilage-derived progenitor cells-conditioned media (hFCPCs-CM) and its useful properties are compared with those of human bone marrow-derived MSCs-conditioned media (hBMSCs-CM) in terms of biochemical characterization, and in vitro studies of fibroblast behavior, macrophage polarization, and burn wound healing. The hFCPCs-CM show to be devoid of cellular components but to contain large amounts of total protein, collagen, glycosaminoglycans, and growth factors, including IGFBP-2, IGFBP-6, HGF, VEGF, TGF β3, and M-CSF, and contain a specific protein, collagen alpha-1(XIV) compare with hBMSCs-CM. The therapeutic potential of hFCPCs-CM observes to be better than that of hBMSCs-CM in the viability, proliferation, and migration of fibroblasts, and M2 macrophage polarization in vitro, and efficient acceleration of wound healing and minimization of scar formation in third-degree burn wounds in a rat model. The current study shows the potential therapeutic effect of hFCPCs and provides a rationale for using the secretome released from fetal progenitor cells to promote the regeneration of skin tissues, both quantitatively and qualitatively. The ready-to-use product of human fetal cartilage-derived progenitor cells-conditioned media (hFCPCs-CM) are fabricated via a series of techniques, including a 3D culture of hFCPCs, filtration using a 3.5 kDa cutoff dialysis membrane, and lyophilization of the CM. hFCPCs-CM contains many ECM molecules and biomolecules that improves wound healing through efficient acceleration of M2 macrophage polarization and reduction of scar formation.
Collapse
Affiliation(s)
- Ngoc-Trinh Tran
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Korea
| | - In-Su Park
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Korea
| | | | - Do-Young Park
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Korea
| | - Sang-Hyug Park
- Advanced Translational Engineering and Medical Science, Seoul, Korea.
- Department of Biomecial Engineering, Pukyong National University, Busan, 48513, Korea.
| | - Byoung-Hyun Min
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea.
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Korea.
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Korea.
- Advanced Translational Engineering and Medical Science, Seoul, Korea.
| |
Collapse
|
8
|
Xu Z, Lin L, Fan Y, Huselstein C, De Isla N, He X, Chen Y, Li Y. Secretome of Mesenchymal Stem Cells from Consecutive Hypoxic Cultures Promotes Resolution of Lung Inflammation by Reprogramming Anti-Inflammatory Macrophages. Int J Mol Sci 2022; 23:ijms23084333. [PMID: 35457151 PMCID: PMC9032661 DOI: 10.3390/ijms23084333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 02/06/2023] Open
Abstract
The secretome from hypoxia-preconditioned mesenchymal stem cells (MSCs) has been shown to promote resolution of inflammation and alleviate acute lung injury (ALI) through its immunomodulatory function. However, the effects of consecutive hypoxic culture on immunomodulatory function of the MSCs secretome are largely unclarified. Here, we intend to investigate the effects of consecutive hypoxia on therapeutic efficacy of conditioned medium derived from MSCs (MSCs-CM) in alleviating ALI. Human umbilical cord-derived MSCs (UC-MSCs) were consecutively cultured in 21% O2 (Nor-MSCs) or in 1% O2 (Hypo-MSCs) from passage 0. Their conditioned medium (Nor-CM and Hypo-CM respectively) was collected and administered into ALI models. Our findings confirmed that Hypo-MSCs exhibited increased proliferation ability and decreased cell senescence compared with Nor-MSCs. Consecutive hypoxia promoted UC-MSCs to secrete immunomodulatory cytokines, such as insulin-like growth factor 1(IGF1), IL10, TNFα-stimulated gene 6(TSG6), TGFβ, and prostaglandin E2 (PGE2). Both Nor-CM and Hypo-CM could effectively limit lung inflammation, promote efferocytosis and modulate anti-inflammatory polarization of lung macrophages in ALI models. Moreover, the effects of Hypo-CM were more potent than Nor-CM. Taken together, our findings indicate that consecutive hypoxic cultures could not only promote both proliferation and quality of UC-MSCs, but also enhance the therapeutic efficacy of their secretome in mitigating lung inflammation by promoting efferocytosis and anti-inflammatory polarization of macrophages.
Collapse
Affiliation(s)
- Zhihong Xu
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
| | - Lulu Lin
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
| | - Yuxuan Fan
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
| | - Céline Huselstein
- UMR 7365 CNRS, Medical School, University of Lorraine, 54505 Nancy, France; (C.H.); (N.D.I.)
| | - Natalia De Isla
- UMR 7365 CNRS, Medical School, University of Lorraine, 54505 Nancy, France; (C.H.); (N.D.I.)
| | - Xiaohua He
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
| | - Yun Chen
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
| | - Yinping Li
- Department of Pathophysiology, Hubei Province Key Laboratory of Allergy and Immunology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan 430071, China; (Z.X.); (L.L.); (Y.F.); (X.H.); (Y.C.)
- Correspondence: ; Tel.: +86-27-6875-8727; Fax: +86-27-6875-9222
| |
Collapse
|
9
|
Li X, Wen H, Lv J, Luan B, Meng J, Gong S, Wen J, Xin S. Therapeutic efficacy of mesenchymal stem cells for abdominal aortic aneurysm: a meta-analysis of preclinical studies. Stem Cell Res Ther 2022; 13:81. [PMID: 35209940 PMCID: PMC8867868 DOI: 10.1186/s13287-022-02755-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/09/2022] [Indexed: 11/10/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA) is life-threatening, surgical treatment is currently the only clinically available intervention for the disease. Mesenchymal stem cells (MSCs) have presented eligible immunomodulatory and regenerative abilities which showed favorable therapeutic efficacy in various cardiovascular diseases. However, current evidence summarizing the effectiveness of MSCs for AAA is lacking. Thus, a meta-analysis and systematic review was necessary to be performed to assess the therapeutic efficacy of MSCs for AAA in preclinical studies. Methods Comprehensive literature search restricted in English was conducted in PubMed, Cochrane Library, EBSCO, EMBASE and Web of Science from inception to Oct 2021. The primary outcomes were parameters about aortic diameter change during MSCs intervention. The secondary outcomes included elastin content and expression level of inflammatory cytokines, matrix metalloproteinases (MMPs) and their inhibitors (TIMPs). Data were extracted and analyzed independently by two authors. The meta package with random effects model was used to calculate the pooled effect size and 95% confidence intervals in R (version 4.0.2). Results Meta-analysis of 18 included studies demonstrated that MSCs intervention has significant therapeutic effects on suppressing aortic diameter enlargement compared with the control group (diameter, SMD = − 1.19, 95% CI [− 1.47, − 0.91]; diameter change ratio, SMD = − 1.36, 95% CI [− 1.72, − 1.00]). Subgroup analysis revealed differences between MSCs and control group regarding to cell type, intervention route and cell compatibility. Moreover, the meta-analysis also showed that MSCs intervention had a significant effect on preserving aortic elastin content, reducing MCP-1, TNF-α, IL-6, MMP-2/9 and increasing TIMP-1/2 expression level compared with control group. Conclusion Our results suggested that MSC intervention is effective in AAA by suppressing aortic diameter enlargement, reducing elastin degradation, and modulating local immunoinflammatory reactions. These results are important for the systemic application of MSCs as a potential treatment candidate for AAA in further animal experiments and clinical trials. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02755-w.
Collapse
Affiliation(s)
- Xintong Li
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, No. 155, Nanjing Street, Heping District, Shenyang, 110001, China.,Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China
| | - Hao Wen
- Department of Trauma Center, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Junyuan Lv
- Department of Breast and Thyroid Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Boyang Luan
- Department of Trauma Center, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jinze Meng
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Shiqiang Gong
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Jie Wen
- Department of Ultrasonography, Inner Mongolia Baotou City Central Hospital, Baotou, China
| | - Shijie Xin
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, No. 155, Nanjing Street, Heping District, Shenyang, 110001, China. .,Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China.
| |
Collapse
|
10
|
Sant'Ana AN, Araújo AB, Gonçalves FDC, Paz AH. Effects of living and metabolically inactive mesenchymal stromal cells and their derivatives on monocytes and macrophages. World J Stem Cells 2021; 13:1160-1176. [PMID: 34630856 PMCID: PMC8474715 DOI: 10.4252/wjsc.v13.i9.1160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/01/2021] [Accepted: 09/03/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent and self-renewing stem cells that have great potential as cell therapy for autoimmune and inflammatory disorders, as well as for other clinical conditions, due to their immunoregulatory and regenerative properties. MSCs modulate the inflammatory milieu by releasing soluble factors and acting through cell-to-cell mechanisms. MSCs switch the classical inflammatory status of monocytes and macrophages towards a non-classical and anti-inflammatory phenotype. This is characterized by an increased secretion of anti-inflammatory cytokines, a decreased release of pro-inflammatory cytokines, and changes in the expression of cell membrane molecules and in metabolic pathways. The MSC modulation of monocyte and macrophage phenotypes seems to be critical for therapy effectiveness in several disease models, since when these cells are depleted, no immunoregulatory effects are observed. Here, we review the effects of living MSCs (metabolically active cells) and metabolically inactive MSCs (dead cells that lost metabolic activity by induced inactivation) and their derivatives (extracellular vesicles, soluble factors, extracts, and microparticles) on the profile of macrophages and monocytes and the implications for immunoregulatory and reparative processes. This review includes mechanisms of action exhibited in these different therapeutic approaches, which induce the anti-inflammatory properties of monocytes and macrophages. Finally, we overview several possibilities of therapeutic applications of these cells and their derivatives, with results regarding monocytes and macrophages in animal model studies and some clinical trials.
Collapse
Affiliation(s)
- Alexia Nedel Sant'Ana
- Laboratório de Células Tecidos e Genes, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Anelise Bergmann Araújo
- Centro de Processamento Celular, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil.
| | | | - Ana Helena Paz
- Laboratório de Células Tecidos e Genes, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| |
Collapse
|
11
|
Multivalent effects of heptamannosylated β-cyclodextrins on macrophage polarization to accelerate wound healing. Colloids Surf B Biointerfaces 2021; 208:112071. [PMID: 34461486 DOI: 10.1016/j.colsurfb.2021.112071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 02/05/2023]
Abstract
Macrophages have high plasticity and heterogeneity, and can suppress or mediate inflammation, depending on their cytokine secretion and phenotype. Regulating macrophage polarization into its M2 phenotype has a remarkable effect on inflammatory inhibition, inducing the regeneration of injured tissues. Here, we synthesized two heptamannosylated β-cyclodextrin derivatives (CD-Man7 and C3-CD-Man7) and demonstrated that their multivalent mannose ligands could induce M2 macrophage polarization to accelerate wound healing. Unlike hydrophilic CD-Man7, amphiphilic C3-CD-Man7 can self-assemble to form nanoparticles (CD-Man-NPs) in aqueous solution. Further, in vitro results confirmed that multivalent mannose ligands of either CD-Man7 or CD-Man-NPs stimulated RAW264.7 macrophages to differentiate into the M2 phenotype, which promoted fibroblast migration via a paracrine mechanism. In vivo results confirmed that both CD-Man7 and CD-Man-NPs reduced the inflammatory response in wound tissue and accelerated wound healing. The present study demonstrates multivalent effects of CD-Man7 and CD-Man-NPs on M2 macrophage polarization, indicating the therapeutic potential of these β-cyclodextrin glycoconjugates in the treatment of inflammatory diseases and wound healing.
Collapse
|
12
|
Verma K, Pant M, Paliwal S, Dwivedi J, Sharma S. An Insight on Multicentric Signaling of Angiotensin II in Cardiovascular system: A Recent Update. Front Pharmacol 2021; 12:734917. [PMID: 34489714 PMCID: PMC8417791 DOI: 10.3389/fphar.2021.734917] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022] Open
Abstract
The multifaceted nature of the renin-angiotensin system (RAS) makes it versatile due to its involvement in pathogenesis of the cardiovascular disease. Angiotensin II (Ang II), a multifaceted member of RAS family is known to have various potential effects. The knowledge of this peptide has immensely ameliorated after meticulous research for decades. Several studies have evidenced angiotensin I receptor (AT1 R) to mediate the majority Ang II-regulated functions in the system. Functional crosstalk between AT1 R mediated signal transduction cascades and other signaling pathways has been recognized. The review will provide an up-to-date information and recent discoveries involved in Ang II receptor signal transduction and their functional significance in the cardiovascular system for potential translation in therapeutics. Moreover, the review also focuses on the role of stem cell-based therapies in the cardiovascular system.
Collapse
Affiliation(s)
- Kanika Verma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Malvika Pant
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| |
Collapse
|
13
|
Nguyen-Truong M, Hematti P, Wang Z. Current status of myocardial restoration via the paracrine function of mesenchymal stromal cells. Am J Physiol Heart Circ Physiol 2021; 321:H112-H127. [PMID: 34085844 DOI: 10.1152/ajpheart.00217.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mesenchymal stromal cells (MSCs) have been studied for nearly two decades as a therapy for myocardial restoration. An emerging direction to repair myocardium is through their paracrine function, which includes the utilization of MSC-derived conditioned medium or extracellular vesicles. In this review, we go over the unique characteristics of MSCs that make it suitable for "off the shelf," cell-free regenerative therapy, current MSC-derived cell-free approaches including their advantages and disadvantages, and the known mechanisms of action of the paracrine effect of MSCs. With a summary of the clinical trials and preclinical studies of MSC-derived cell-free therapy, we classify the aforementioned mechanisms into angiogenesis, immunomodulation, extracellular matrix remodeling, antiapoptosis, and antioxidation. Particularly, we discuss on ways researchers have worked toward enhancing these desired properties to improve the therapeutic outcomes and the investigation of mechanobiology involved in MSC paracrine function. Lastly, we bring up the remaining challenges in this arising field and suggestions for future directions to improve our understanding and control over the potential of MSC paracrine function for myocardial restoration.
Collapse
Affiliation(s)
| | - Peiman Hematti
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado.,Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
14
|
Manganeli Polonio C, Longo de Freitas C, Garcia de Oliveira M, Rossato C, Nogueira Brandão W, Ghabdan Zanluqui N, Gomes de Oliveira L, Ayumi Nishiyama Mimura L, Braga Barros Silva M, Lúcia Garcia Calich V, Gil Nisenbaum M, Halpern S, Evangelista L, Maluf M, Perin P, Eduardo Czeresnia C, Schatzmann Peron JP. Murine endometrial-derived mesenchymal stem cells suppress experimental autoimmune encephalomyelitis depending on indoleamine-2,3-dioxygenase expression. Clin Sci (Lond) 2021; 135:1065-1082. [PMID: 33960391 DOI: 10.1042/cs20201544] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/19/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022]
Abstract
Cellular therapy with mesenchymal stem cells (MSCs) is a huge challenge for scientists, as little translational relevance has been achieved. However, many studies using MSCs have proved their suppressive and regenerative capacity. Thus, there is still a need for a better understanding of MSCs biology and the establishment of newer protocols, or to test unexplored tissue sources. Here, we demonstrate that murine endometrial-derived MSCs (meMSCs) suppress Experimental Autoimmune Encephalomyelitis (EAE). MSC-treated animals had milder disease, with a significant reduction in Th1 and Th17 lymphocytes in the lymph nodes and in the central nervous system (CNS). This was associated with increased Il27 and Cyp1a1 expression, and presence of IL-10-secreting T CD4+ cells. At EAE peak, animals had reduced CNS infiltrating cells, histopathology and demyelination. qPCR analysis evidenced the down-regulation of several pro-inflammatory genes and up-regulation of indoleamine-2,3-dioxygenase (IDO). Consistently, co-culturing of WT and IDO-/- meMSCs with T CD4+ cells evidenced the necessity of IDO on the suppression of encephalitogenic lymphocytes, and IDO-/- meMSCs were not able to suppress EAE. In addition, WT meMSCs stimulated with IL-17A and IFN-γ increased IDO expression and secretion of kynurenines in vitro, indicating a negative feedback loop. Pathogenic cytokines were increased when CD4+ T cells from AhR-/- mice were co-cultured with WT meMSC. In summary, our research evidences the suppressive activity of the unexplored meMSCs population, and shows the mechanism depends on IDO-kynurenines-Aryl hydrocarbon receptor (AhR) axis. To our knowledge this is the first report evidencing that the therapeutic potential of meMSCs relying on IDO expression.
Collapse
Affiliation(s)
- Carolina Manganeli Polonio
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Carla Longo de Freitas
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Marília Garcia de Oliveira
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Cristiano Rossato
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Wesley Nogueira Brandão
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Nágela Ghabdan Zanluqui
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo (USP), São Paulo, SP, Brazil
- Immunopathology and Allergy Post Graduate Program, School of Medicine, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Lilian Gomes de Oliveira
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo (USP), São Paulo, SP, Brazil
| | | | - Maysa Braga Barros Silva
- Clinical Biochemistry Laboratory, Clinical Analysis Department, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Vera Lúcia Garcia Calich
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | | | - Silvio Halpern
- Division of Reproductive Medicine, Halpern Clinic, São Paulo, SP, Brazil
| | | | | | - Paulo Perin
- Division of Reproductive Medicine, CEERH, São Paulo, SP, Brazil
| | | | - Jean Pierre Schatzmann Peron
- Neuroimmune Interactions Laboratory, Department of Immunology, University of São Paulo (USP), São Paulo, SP, Brazil
- Immunopathology and Allergy Post Graduate Program, School of Medicine, University of São Paulo (USP), São Paulo, SP, Brazil
| |
Collapse
|
15
|
Xu T, Wang S, Li X, Li X, Qu K, Tong H, Zhang R, Bai S, Fan J. Lithium chloride represses abdominal aortic aneurysm via regulating GSK3β/SIRT1/NF-κB signaling pathway. Free Radic Biol Med 2021; 166:1-10. [PMID: 33588051 DOI: 10.1016/j.freeradbiomed.2021.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/14/2021] [Accepted: 02/04/2021] [Indexed: 10/22/2022]
Abstract
Lithium chloride (LiCl), a pharmacological compound, was effective in reducing inflammation, but whether it can protect against abdominal aortic aneurysm (AAA) is largely unknown. This study is designed to investigate therapeutic effects of LiCl on AAA and the potential mechanism. Rat AAA models were induced by periaortic application of CaCl2. AAA rats were treated by daily intraperitoneal injection of LiCl or vehicle alone to study the protection effects of LiCl in vivo. Rat primary vascular smooth muscle cells (VSMCs) stimulated with tumor necrosis factor (TNF)-α served as an in vitro model. LiCl treatment prevented the development of AAA through inhibiting the inflammatory cells infiltration and inflammatory cytokines overproduction, as well as attenuating superoxide production and elastin degradation in aorta of AAA rats. Additionally, the downregulation of p-GSK3β(Ser9) and SIRT1, upregulation of NF-κB(p-65), MMP-2 and MMP-9 in AAA were abolished by LiCl treatment. In vitro by upregulating p-GSK3β(Ser9), LiCl significantly induced SIRT1 expression, along with inhibition of the NF-κB activation and decreased elastin level elicited in VSMCs by TNF-α stimulation. SIRT1 activator SRT1720 achieved similar repressive effects as LiCl on TNF-α-induced NF-κB activation and decreased elastin in VSMCs. Moreover, administration of LiCl also caused regression of established rats AAA. This study provided the first evidence that LiCl prevented the development of AAA through inhibiting inflammation, MMPs, and superoxide production, and facilitating the biosynthesis of elastin. The beneficial effect of LiCl may be mediated by regulation GSK3β/SIRT1/NF-κB cascade.
Collapse
Affiliation(s)
- Tong Xu
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Shoushuai Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Xiang Li
- Department of Cell Biology, Key Laboratory of Cell Biology of National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology of Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Xiuquan Li
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Kaiyun Qu
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Hao Tong
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Ruijie Zhang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Shuling Bai
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Jun Fan
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning, 110122, PR China.
| |
Collapse
|
16
|
Zhao B, Sun Q, Fan Y, Hu X, Li L, Wang J, Cui S. Transplantation of bone marrow-derived mesenchymal stem cells with silencing of microRNA-138 relieves pelvic organ prolapse through the FBLN5/IL-1β/elastin pathway. Aging (Albany NY) 2021; 13:3045-3059. [PMID: 33460398 PMCID: PMC7880387 DOI: 10.18632/aging.202465] [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: 02/18/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
Nondegradable transvaginal polypropylene meshes for treating pelvic organ prolapse (POP) are now generally unavailable or banned due to serious adverse events. New tissue engineering approaches combine degradable scaffolds with mesenchymal stem/stromal cells from human endometrium (eMSC). In this study, we investigate effect of microRNA-138 (miR-138) regulation on bone marrow-derived mesenchymal stem cells (BMSCs) and the efficacy of BMSC transplantation therapy in a rat POP model. We first identified FBLN5 as a target of miR-138. miR-138, fibulin-5 (FBLN5), interleukin-1β (IL-1β), and elastin expression in uterosacral ligament of POP patients and controls were detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. After isolation and identification, BMSCs were treated to alter their expression of miR-138 or FBLN5. Proliferation of BMSCs was analyzed by CCK-8. After establishing the rat pelvic floor dysfunction (PFD) model, we evaluated efficacy of BMSC injection by applying leak point pressure (LPP) and the conscious cystometry (CMG) tests. miR-138 inhibition resulted in increased viability of BMSCs and elevated their secretion of elastin, while downregulating IL-1β expression. BMSCs with inhibited miR-138 improved LPP and conscious CMG results in vivo. Taken together, miR-138 could be a potential therapeutic target for treating POP in conjunction with tissue engineering.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Qing Sun
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Yazhou Fan
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Xinming Hu
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Linyu Li
- Department of Scientific Research, Xinxiang Medical University Sanquan Medical College, Xinxiang 453003, Henan Province, PR China
| | - Junmin Wang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan Province, PR China
| | - Shihong Cui
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| |
Collapse
|
17
|
Li Y, Yan J, Wang M, Lv J, Yan F, Chen J. Uremic toxin indoxyl sulfate promotes proinflammatory macrophage activation by regulation of β-catenin and YAP pathways. J Mol Histol 2021; 52:197-205. [PMID: 33387144 PMCID: PMC8012310 DOI: 10.1007/s10735-020-09936-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Abstract
Evidence has been shown that indoxyl sulfate (IS) could impair kidney and cardiac functions. Moreover, macrophage polarization played important roles in chronic kidney disease and cardiovascular disease. IS acts as a nephron-vascular toxin, whereas its effect on macrophage polarization during inflammation is still not fully elucidated. In this study, we aimed to investigate the effect of IS on macrophage polarization during lipopolysaccharide (LPS) challenge. THP-1 monocytes were incubated with phorbol 12-myristate-13-acetate (PMA) to differentiate into macrophages, and then incubated with LPS and IS for 24 h. ELISA was used to detect the levels of TNFα, IL-6, IL-1β in THP-1-derived macrophages. Western blot assay was used to detect the levels of arginase1 and iNOS in THP-1-derived macrophages. Percentages of HLA-DR-positive cells (M1 macrophages) and CD206-positive cells (M2 macrophages) were detected by flow cytometry. IS markedly increased the production of the pro-inflammatory factors TNFα, IL-6, IL-1β in LPS-stimulated THP-1-derived macrophages. In addition, IS induced M1 macrophage polarization in response to LPS, as evidenced by the increased expression of iNOS and the increased proportion of HLA-DR+ macrophages. Moreover, IS downregulated the level of β-catenin, and upregulated the level of YAP in LPS-stimulated macrophages. Activating β-catenin signaling or inhibiting YAP signaling suppressed the IS-induced inflammatory response in LPS-stimulated macrophages by inhibiting M1 polarization. IS induced M1 macrophage polarization in LPS-stimulated macrophages via inhibiting β-catenin and activating YAP signaling. In addition, this study provided evidences that activation of β-catenin or inhibition of YAP could alleviate IS-induced inflammatory response in LPS-stimulated macrophages. This finding may contribute to the understanding of immune dysfunction observed in chronic kidney disease and cardiovascular disease.
Collapse
Affiliation(s)
- Ying Li
- Department of General Practice, Zhejiang Hospital, 12 Lingyin Road, West Lake District, Hangzhou, 310013, Zhejiang, People's Republic of China
| | - Jing Yan
- Department of Critical Care Medicine, Zhejiang Hospital, Hangzhou, 310013, Zhejiang, People's Republic of China
| | - Minjia Wang
- Department of Critical Care Medicine, Zhejiang Hospital, Hangzhou, 310013, Zhejiang, People's Republic of China
| | - Jing Lv
- Department of General Practice, Zhejiang Hospital, 12 Lingyin Road, West Lake District, Hangzhou, 310013, Zhejiang, People's Republic of China
| | - Fei Yan
- Department of General Practice, Zhejiang Hospital, 12 Lingyin Road, West Lake District, Hangzhou, 310013, Zhejiang, People's Republic of China
| | - Jin Chen
- Department of General Practice, Zhejiang Hospital, 12 Lingyin Road, West Lake District, Hangzhou, 310013, Zhejiang, People's Republic of China.
| |
Collapse
|
18
|
Sasaki R, Takami T, Fujisawa K, Matsumoto T, Ishikawa T, Yamamoto N, Sakaida I. Trans-portal hepatic infusion of cultured bone marrow-derived mesenchymal stem cells in a steatohepatitis murine model. J Clin Biochem Nutr 2020; 67:274-282. [PMID: 33293768 PMCID: PMC7705078 DOI: 10.3164/jcbn.20-88] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/13/2020] [Indexed: 12/23/2022] Open
Abstract
The incidence of nonalcoholic steatohepatitis-related liver cirrhosis is increasing. We used a steatohepatitis murine model fed a choline-deficient, l-amino acid-defined (CDAA) diet with a single injection of carbon tetrachloride (CCl4) to evaluate the efficacy of trans-portal hepatic infusion of bone marrow-derived mesenchymal stem cells (BMSCs) for liver fibrosis, liver steatosis, and oxidative stress. Mice were fed a CDAA diet and injected with a single intraperitoneal dose of CCl4 (0.5 ml/kg) after 4 weeks of CDAA diet. After 12 weeks of CDAA diet, 1 × 106 luciferase-positive syngeneic BMSCs (Luc-BMSCs) were infused into the animal spleen. An in vivo imaging system was used to confirm Luc-BMSC accumulation in the liver via the portal vein, and at 4 weeks after infusion, we compared liver fibrosis, liver steatosis, and oxidative stress. After the BMSC-infusion, serum albumin and serum total bilirubin were significantly improved. Liver fibrosis assessed by Sirius red staining, α-smooth muscle actin protein, and collagen 1A1 mRNA expression was significantly suppressed. Furthermore, liver steatosis area was significantly lower, the 8-hydroxy-2'-deoxyguanosine-positive cells were significantly fewer, and superoxide dismutase 2 protein expression of the liver was significantly increased. In conclusion, our data confirmed the efficacy of trans-portal hepatic infusion of BMSCs in a steatohepatitis murine model.
Collapse
Affiliation(s)
- Ryo Sasaki
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Taro Takami
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan.,Center for Regenerative and Cell Therapy, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Koichi Fujisawa
- Department of Liver Regenerative Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Toshihiko Matsumoto
- Department of Oncology and Laboratory Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Tsuyoshi Ishikawa
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Naoki Yamamoto
- Yamaguchi University Health Administration Center, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-0046, Japan
| | - Isao Sakaida
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan.,Center for Regenerative and Cell Therapy, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan.,Department of Liver Regenerative Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
19
|
Kim R, Song BW, Kim M, Kim WJ, Lee HW, Lee MY, Kim J, Chang W. Regulation of alternative macrophage activation by MSCs derived hypoxic conditioned medium, via the TGF-β1/Smad3 pathway. BMB Rep 2020; 53:600-604. [PMID: 33050988 PMCID: PMC7704222 DOI: 10.5483/bmbrep.2020.53.11.177] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/06/2020] [Accepted: 10/05/2020] [Indexed: 01/01/2023] Open
Abstract
Macrophages are re-educated and polarized in response to myocardial infarction (MI). The M2 anti-inflammatory phenotype is a known dominator of late stage MI. Mesenchymal stem cells (MSCs) represent a promising tool for cell therapy, particularly heart related diseases. In general, MSCs induce alteration of the macrophage subtype from M1 to M2, both in vitro and in vivo. We conjectured that hypoxic conditions can promote secretome productivity of MSCs. Hypoxia induces TGF-β1 expression, and TGF-β1 mediates M2 macrophage polarization for anti-inflammation and angiogenesis in infarcted areas. We hypothesized that macrophages undergo advanced M2 polarization after exposure to MSCs in hypoxia. Treatment of MSCs derived hypoxic conditioned medium (hypo-CM) promoted M2 phenotype and neovascularization through the TGF-β1/Smad3 pathway. In addition, hypo-CM derived from MSCs improved restoration of ischemic heart, such as attenuating cell apoptosis and fibrosis, and ameliorating microvessel density. Based on our results, we propose a new therapeutic method for effective MI treatment using regulation of macrophage polarization. [BMB Reports 2020; 53(11): 600-604].
Collapse
Affiliation(s)
- Ran Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Korea
| | - Byeong-Wook Song
- Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung 25601, Korea
| | - Minji Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Korea
| | - Won Jung Kim
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Korea
| | - Hee Won Lee
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Korea
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea
| | - Jongmin Kim
- Department of Life Systems, Sookmyung Women’s University, Seoul 04310, Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan 46241, Korea
| |
Collapse
|
20
|
Tutuianu R, Rosca AM, Albu Kaya MG, Pruna V, Neagu TP, Lascar I, Simionescu M, Titorencu I. Mesenchymal stromal cell-derived factors promote the colonization of collagen 3D scaffolds with human skin cells. J Cell Mol Med 2020; 24:9692-9704. [PMID: 32666712 PMCID: PMC7520263 DOI: 10.1111/jcmm.15507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/07/2020] [Accepted: 05/31/2020] [Indexed: 12/12/2022] Open
Abstract
The development of stem cell technology in combination with advances in biomaterials has opened new ways of producing engineered tissue substitutes. In this study, we investigated whether the therapeutic potential of an acellular porous scaffold made of type I collagen can be improved by the addition of a powerful trophic agent in the form of mesenchymal stromal cells conditioned medium (MSC-CM) in order to be used as an acellular scaffold for skin wound healing treatment. Our experiments showed that MSC-CM sustained the adherence of keratinocytes and fibroblasts as well as the proliferation of keratinocytes. Moreover, MSC-CM had chemoattractant properties for keratinocytes and endothelial cells, attributable to the content of trophic and pro-angiogenic factors. Also, for the dermal fibroblasts cultured on collagen scaffold in the presence of MSC-CM versus serum control, the ratio between collagen III and I mRNAs increased by 2-fold. Furthermore, the gene expression for α-smooth muscle actin, tissue inhibitor of metalloproteinase-1 and 2 and matrix metalloproteinase-14 was significantly increased by approximately 2-fold. In conclusion, factors existing in MSC-CM improve the colonization of collagen 3D scaffolds, by sustaining the adherence and proliferation of keratinocytes and by inducing a pro-healing phenotype in fibroblasts.
Collapse
Affiliation(s)
- Raluca Tutuianu
- Institute of Cellular Biology and Pathology, "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Ana-Maria Rosca
- Institute of Cellular Biology and Pathology, "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | | | - Vasile Pruna
- INCDTP-Division Leather and Footwear Research Institute, Bucharest, Romania
| | | | - Ioan Lascar
- University of Medicine and Pharmacy "Carol Davila", Bucharest, Romania
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology, "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| | - Irina Titorencu
- Institute of Cellular Biology and Pathology, "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
21
|
Guo Y, Yu Y, Hu S, Chen Y, Shen Z. The therapeutic potential of mesenchymal stem cells for cardiovascular diseases. Cell Death Dis 2020; 11:349. [PMID: 32393744 PMCID: PMC7214402 DOI: 10.1038/s41419-020-2542-9] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are derived from a wide range of sources and easily isolated and cultured. MSCs have the capacity for in vitro amplification and self-renewal, low immunogenicity and immunomodulatory properties, and under certain conditions, MSCs can be differentiated into a variety of cells. In the cardiovascular system, MSCs can protect the myocardium by reducing the level of inflammation, promoting the differentiation of myocardial cells around infarct areas and angiogenesis, increasing apoptosis resistance, and inhibiting fibrosis, which are ideal qualities for cardiovascular repair. Preclinical studies have shown that MSCs can be transplanted and improve cardiac repair, but challenges, such as their low rate of migration to the ischemic myocardium, low tissue retention, and low survival rate after transplantation, remain. This article reviews the potential and methods of MSC transplantation in the treatment of cardiovascular diseases (CVDs) and the challenges of the clinical use of MSCs.
Collapse
Affiliation(s)
- Yajun Guo
- Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China.,Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, Suzhou 215006, China
| | - Yunsheng Yu
- Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China.,Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, Suzhou 215006, China
| | - Shijun Hu
- Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China. .,Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, Suzhou 215006, China. .,State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou 215123, China. .,Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China.
| | - Yueqiu Chen
- Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China. .,Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, Suzhou 215006, China.
| | - Zhenya Shen
- Institute for Cardiovascular Science, Soochow University, Suzhou 215006, China. .,Department of Cardiovascular Surgery of The First Affiliated Hospital, Soochow University, Suzhou 215006, China.
| |
Collapse
|
22
|
MiR-144-5p limits experimental abdominal aortic aneurysm formation by mitigating M1 macrophage-associated inflammation: Suppression of TLR2 and OLR1. J Mol Cell Cardiol 2020; 143:1-14. [PMID: 32278833 DOI: 10.1016/j.yjmcc.2020.04.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/03/2020] [Accepted: 04/05/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND It has been noted that dysregulation of microRNAs (miRNAs) contributes to the formation of abdominal aortic aneurysm (AAA), a vascular disease associated with progressive aortic dilatation and degradation, and pathological infiltration and activation of inflammatory cells, such as macrophages. Our microarray data revealing that miR-144-5p was the top 1 downregulated miRNA in mouse AAA tissues as compared to normal aortas motivated us to explore its role in AAA development. METHODS We profiled miRNA and mRNA expression in Angiotensin II (Ang II)- (n = 3) and saline-infused abdominal aortas (n = 4) via Agilent microarrays, and further validated the data with real-time QPCR. In vivo, miR-144-5p or control agomirs were given to Apoe-/- mice with Ang II infusion-induced AAA. In vitro, mouse RAW 264.7 macrophages and human THP-1 macrophage-like cells were transfected with miR-144-5p or control agomirs/antagomirs, and oxidized Low Density Lipoprotein (ox-LDL) was used to stimulate M1 macrophage polarization. RESULTS Based on the microarray and real-time QPCR validation data, we identified miR-144-5p as a novel downregulated miRNA in AAA tissues. Overexpression of miR-144-5p by utilizing its specific agomirs in vivo significantly attenuated Ang II-induced aortic dilatation and elastic degradation in Apoe-/- mice and improved their survival. AAA incidence was reduced by miR-144-5p as well. MiR-144-5p polarized macrophages to M2 type in Ang II-infused aortas. Further, the expression levels of two predictive targets for miR-144-5p, Toll Like Receptor 2 (TLR2) and ox-LDL Receptor 1 (OLR1), were higher in AAA specimens, and negatively correlated to miR-144-5p (Pearson correlation coefficient r < -0.9, P < .01). These two molecules were then confirmed as novel miR-144-5p targets via dual-luciferase assay. MiR-144-5p agomirs suppressed ox-LDL-induced upregulation of M1 macrophage markers, including interleukin 1β (IL1β), tumor necrosis factor α (TNFα), prostaglandin-endoperoxide synthase 2 (PTGS2) and nitric oxide synthase 2 (NOS2), in macrophages probably by targeting TLR2. MiR-144-5p also inhibited the signaling transduction of pathways downstream to TLR2 and OLR1, including NF-κB and ERK1/2 pathways, whose abnormal activation contributed AAA formation. CONCLUSION Our work suggests miR-144-5p as a novel regulator for AAA pathology. Management of miR-144-5p and its targets TLR2 and OLR1 provides therapeutic potential for limiting AAA formation.
Collapse
|
23
|
Wu QY, Cheng Z, Zhou YZ, Zhao Y, Li JM, Zhou XM, Peng HL, Zhang GS, Liao XB, Fu XM. A novel STAT3 inhibitor attenuates angiotensin II-induced abdominal aortic aneurysm progression in mice through modulating vascular inflammation and autophagy. Cell Death Dis 2020; 11:131. [PMID: 32071300 PMCID: PMC7028955 DOI: 10.1038/s41419-020-2326-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 11/24/2022]
Abstract
Abdominal Aortic aneurysm (AAA) is associated with chronic inflammation, cells apoptosis, and impairment of autophagy. BP-1-102, a novel potent STAT3 inhibitor, has been recently reported to significantly block inflammation-related signaling pathways of JAK2/STAT3 and NF-κB, as well as regulate autophagy. However, its role in vascular inflammation and AAA progression remains to be elucidated. In the present study, the effect and potential mechanisms of BP-1-102 on angiotensin II (AngII) induced AAA in ApoE−/− mice were investigated. AAA was induced in ApoE−/− mice with infusion of AngII for 28 days. BP-1-102 was administrated orally to mice every other day. Mice were sacrificed on day 7, day 14, and day 28 to evaluate the treatment effects. BP-1-102 markedly decreased AAA incidence and aortic diameter, maintained elastin structure and volume, reduced the expression of pro-inflammatory cytokines and MMPs, and inhibited inflammatory cells infiltration. Moreover, BP-1-102 dramatically reduced the expression of JAK2, p-STAT3, p-NF-κB, and Bcl-xL but maintained the expression of LC3B and Beclin in AAA tissues. In vitro, vascular smooth muscle cells (VSMCs) were treated with AngII and/or BP-1-102 at indicated time and concentration. BP-1-102 inhibited AngII-induced JAK2/STAT3 and NF-κB signaling activation and maintained autophagy-related proteins expression in VSMCs. Taken together, our findings suggest that BP-1-102 inhibits vascular inflammation and AAA progression through decreasing JAK2/STAT3 and NF-κB activation and maintaining autophagy.
Collapse
Affiliation(s)
- Qi-Ying Wu
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Zhao Cheng
- Department of Hematology, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yang-Zhao Zhou
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yuan Zhao
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Jian-Ming Li
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xin-Min Zhou
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Hong-Ling Peng
- Department of Hematology, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Guang-Sheng Zhang
- Department of Hematology, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xiao-Bo Liao
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xian-Ming Fu
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, Hunan, P.R. China.
| |
Collapse
|
24
|
Di Somma M, Schaafsma W, Grillo E, Vliora M, Dakou E, Corsini M, Ravelli C, Ronca R, Sakellariou P, Vanparijs J, Castro B, Mitola S. Natural Histogel-Based Bio-Scaffolds for Sustaining Angiogenesis in Beige Adipose Tissue. Cells 2019; 8:cells8111457. [PMID: 31752157 PMCID: PMC6912328 DOI: 10.3390/cells8111457] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 12/17/2022] Open
Abstract
In the treatment of obesity and its related disorders, one of the measures adopted is weight reduction by controlling nutrition and increasing physical activity. A valid alternative to restore the physiological function of the human body could be the increase of energy consumption by inducing the browning of adipose tissue. To this purpose, we tested the ability of Histogel, a natural mixture of glycosaminoglycans isolated from animal Wharton jelly, to sustain the differentiation of adipose derived mesenchymal cells (ADSCs) into brown-like cells expressing UCP-1. Differentiated cells show a higher energy metabolism compared to undifferentiated mesenchymal cells. Furthermore, Histogel acts as a pro-angiogenic matrix, induces endothelial cell proliferation and sprouting in a three-dimensional gel in vitro, and stimulates neovascularization when applied in vivo on top of the chicken embryo chorioallantoic membrane or injected subcutaneously in mice. In addition to the pro-angiogenic activity of Histogel, also the ADSC derived beige cells contribute to activating endothelial cells. These data led us to propose Histogel as a promising scaffold for the modulation of the thermogenic behavior of adipose tissue. Indeed, Histogel simultaneously supports the acquisition of brown tissue markers and activates the vasculature process necessary for the correct function of the thermogenic tissue. Thus, Histogel represents a valid candidate for the development of bioscaffolds to increase the amount of brown adipose tissue in patients with metabolic disorders.
Collapse
Affiliation(s)
- Margherita Di Somma
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
| | - Wandert Schaafsma
- Histocell, S.L.Parque Tecnológico 801A, 2o 48160 Derio—BIZKAIA, Spain; (W.S.); (B.C.)
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
| | - Maria Vliora
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
- FAME Laboratory, Department of Exercise Science, University of Thessaly, 38221 Trikala, Greece;
| | - Eleni Dakou
- Laboratory of Cell Genetics, Department of Biology, Faculty of Science and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium;
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
| | - Paraskevi Sakellariou
- FAME Laboratory, Department of Exercise Science, University of Thessaly, 38221 Trikala, Greece;
| | - Jef Vanparijs
- Department of Human Physiology, Faculty of Physical Education and Physical Therapy, Vrije Universiteit Brussel, 1050 Brussels, Belgium;
| | - Begona Castro
- Histocell, S.L.Parque Tecnológico 801A, 2o 48160 Derio—BIZKAIA, Spain; (W.S.); (B.C.)
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, 25121 Brescia, Italy; (M.D.S.); (E.G.); (M.V.); (M.C.); (C.R.); (R.R.)
- Correspondence:
| |
Collapse
|
25
|
Zhou YZ, Cheng Z, Wu Y, Wu QY, Liao XB, Zhao Y, Li JM, Zhou XM, Fu XM. Mesenchymal stem cell-derived conditioned medium attenuate angiotensin II-induced aortic aneurysm growth by modulating macrophage polarization. J Cell Mol Med 2019; 23:8233-8245. [PMID: 31583844 PMCID: PMC6850971 DOI: 10.1111/jcmm.14694] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/03/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) exhibit therapeutic benefits on aortic aneurysm (AA); however, the molecular mechanisms are not fully understood. The current study aimed to investigate the therapeutic effects and potential mechanisms of murine bone marrow MSC (BM‐MSCs)–derived conditioned medium (MSCs‐CM) on angiotensin II (AngII)‐induced AA in apolipoprotein E‐deficient (apoE−/−) mice. Murine BM‐MSCs, MSCs‐CM or control medium were intravenously administrated into AngII‐induced AA in apoE−/− mice. Mice were sacrificed at 2 weeks after injection. BM‐MSCs and MSCs‐CM significantly attenuated matrix metalloproteinase (MMP)‐2 and MMP‐9 expression, aortic elastin degradation and AA growth at the site of AA. These treatments with BM‐MSCs and MSCs‐CM also decreased Ly6chigh monocytes in peripheral blood on day 7 and M1 macrophage infiltration in AA tissues on day 14, whereas they increased M2 macrophages. In addition, BM‐MSCs and MSCs‐CM reduced MCP‐1, IL‐1Ra and IL‐6 expression and increased IL‐10 expression in AA tissues. In vitro, peritoneal macrophages were co‐cultured with BM‐MSCs or fibroblasts as control in a transwell system. The mRNA and protein expression of M2 macrophage markers were evaluated. IL‐6 and IL‐1β were reduced, while IL‐10 was increased in the BM‐MSC systems. The mRNA and protein expression of M2 markers were up‐regulated in the BM‐MSC systems. Furthermore, high concentration of IGF1, VEGF and TGF‐β1 was detected in MSCs‐CM. Our results suggest that MSCs‐CM could prevent AA growth potentially through regulating macrophage polarization. These results may provide a new insight into the mechanisms of BM‐MSCs in the therapy of AA.
Collapse
Affiliation(s)
- Yang-Zhao Zhou
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, China
| | - Zhao Cheng
- Department of Hematology, The Second Xiang-ya Hospital, Central South University, Changsha, China
| | - Yin Wu
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, China
| | - Qi-Ying Wu
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, China
| | - Xiao-Bo Liao
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, China
| | - Yuan Zhao
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, China
| | - Jian-Ming Li
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, China
| | - Xin-Min Zhou
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, China
| | - Xian-Ming Fu
- Department of Cardiovascular Surgery, The Second Xiang-ya Hospital, Central South University, Changsha, China
| |
Collapse
|