1
|
Walkenhorst M, Sonner JK, Meurs N, Engler JB, Bauer S, Winschel I, Woo MS, Raich L, Winkler I, Vieira V, Unger L, Salinas G, Lantz O, Friese MA, Willing A. Protective effect of TCR-mediated MAIT cell activation during experimental autoimmune encephalomyelitis. Nat Commun 2024; 15:9287. [PMID: 39468055 DOI: 10.1038/s41467-024-53657-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells express semi-invariant T cell receptors (TCR) for recognizing bacterial and yeast antigens derived from riboflavin metabolites presented on the non-polymorphic MHC class I-related protein 1 (MR1). Neuroinflammation in multiple sclerosis (MS) is likely initiated by autoreactive T cells and perpetuated by infiltration of additional immune cells, but the precise role of MAIT cells in MS pathogenesis remains unknown. Here, we use experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, and find an accumulation of MAIT cells in the inflamed central nervous system (CNS) enriched for MAIT17 (RORγt+) and MAIT1/17 (T-bet+RORγt+) subsets with inflammatory and protective features. Results from transcriptome profiling and Nur77GFP reporter mice show that these CNS MAIT cells are activated via cytokines and TCR. Blocking TCR activation with an anti-MR1 antibody exacerbates EAE, whereas enhancing TCR activation with the cognate antigen, 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil, ameliorates EAE severity, potentially via the induction of amphiregulin (AREG). In summary, our findings suggest that TCR-mediated MAIT cell activation is protective in CNS inflammation, likely involving an induction of AREG.
Collapse
Affiliation(s)
- Mark Walkenhorst
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Meurs
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Bauer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingo Winschel
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas Raich
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Iris Winkler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Unger
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit, Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Olivier Lantz
- Institut National de la Santé et de la Recherche Médicale U932, PSL University, Institut Curie, Paris, France
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Anne Willing
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
2
|
Zhang X, Tao J, Gong S, Yu X, Shao S. Effects of Recombinant Human Granulocyte/Macrophage Colony-Stimulating Factor on Diabetic Lower Extremity Ulcers: Case Series of Nine Patients. Diabetes Metab Syndr Obes 2024; 17:1941-1956. [PMID: 38737388 PMCID: PMC11088380 DOI: 10.2147/dmso.s461349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/30/2024] [Indexed: 05/14/2024] Open
Abstract
Background Diabetic lower extremity ulcer, including diabetic foot ulcer (DFU) and leg ulcer, is one of the refractory complications of diabetes, the treatment of which is challenging, expensive, and lengthy. Recombinant Human Granulocyte/Macrophage Colony-stimulating Factor (rhGM-CSF) is an immunomodulatory cytokine that has been mainly applied in the treatment of hematological diseases. Clinical evidence regarding GM-CSF in the treatment of diabetic lower extremity ulcers is limited. This study is the first case series that investigates the repurpose effects of rhGM-CSF on diabetic ulcer healing in real clinical practice. Methods Nine patients diagnosed with diabetes and refractory lower extremity ulcer (ulcer duration ≥2 weeks) were included from September 2021 to February 2023 in the Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology. Patients with Wagner grade ≥4 and SINDAD ≥5 were excluded. The included subjects were treated with rhGM-CSF plus standard of care (SOC) including glycemic control, foot care education, debridement of necrotic tissues, topical wound dressings, offloading, and infection control when necessary. The observation endpoint was complete epithelialization. Their clinical manifestations, laboratory tests, and therapeutic effects were extracted and analyzed. Results The case series included 9 cases aged from 29 to 80 years and all the patients were male. Seven of 9 patients presented neuropathic ulcer. Only one case showed non-infected ulcer from tissue samples and one case presented ankle brachial index (ABI) <0.9. It was observed that the ulcer areas among these 9 patients gradually declined throughout the whole treatment period with the average healing velocity 0.32 ± 013 cm2/day and the mean time to complete healing 16.0 ± 3.7 days. The relative area (percentage of initial ulcer area) decreased to 66.7 ± 13.0% on average after the first treatment. Ulcers in all the 9 patients achieved complete epithelialization after 4-8 times treatments. Conclusion The case series suggests rhGM-CSF as a promising therapeutic strategy for the treatment of diabetic ulceration. More robust data from randomized controlled trials are required to further evaluate its clinical efficacy.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, People’s Republic of China
| | - Jing Tao
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, People’s Republic of China
| | - Song Gong
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, People’s Republic of China
| | - Xuefeng Yu
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, People’s Republic of China
| | - Shiying Shao
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People’s Republic of China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, People’s Republic of China
| |
Collapse
|
3
|
Zhao Z, Sun X, Tu P, Ma Y, Guo Y, Zhang Y, Liu M, Wang L, Chen X, Si L, Li G, Pan Y. Mechanisms of vascular invasion after cartilage injury and potential engineering cartilage treatment strategies. FASEB J 2024; 38:e23559. [PMID: 38502020 DOI: 10.1096/fj.202302391rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/20/2024]
Abstract
Articular cartilage injury is one of the most common diseases in orthopedic clinics. Following an articular cartilage injury, an inability to resist vascular invasion can result in cartilage calcification by newly formed blood vessels. This process ultimately leads to the loss of joint function, significantly impacting the patient's quality of life. As a result, developing anti-angiogenic methods to repair damaged cartilage has become a popular research topic. Despite this, tissue engineering, as an anti-angiogenic strategy in cartilage injury repair, has not yet been adequately investigated. This exhaustive literature review mainly focused on the process and mechanism of vascular invasion in articular cartilage injury repair and summarized the major regulatory factors and signaling pathways affecting angiogenesis in the process of cartilage injury. We aimed to discuss several potential methods for engineering cartilage repair with anti-angiogenic strategies. Three anti-angiogenic tissue engineering methods were identified, including administering angiogenesis inhibitors, applying scaffolds to manage angiogenesis, and utilizing in vitro bioreactors to enhance the therapeutic properties of cultured chondrocytes. The advantages and disadvantages of each strategy were also analyzed. By exploring these anti-angiogenic tissue engineering methods, we hope to provide guidance for researchers in related fields for future research and development in cartilage repair.
Collapse
Affiliation(s)
- Zitong Zhao
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiaoxian Sun
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Pengcheng Tu
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Yong Ma
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Yang Guo
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Yafeng Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Mengmin Liu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Lining Wang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xinyu Chen
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Lin Si
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Guangguang Li
- Orthopedics and traumatology department, Yixing Traditional Chinese Medicine Hospital, Yixing, P.R. China
| | - Yalan Pan
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| |
Collapse
|
4
|
Chen R, Li Y, Zhuang Y, Zhang Y, Wu H, Lin T, Chen S. Immune evaluation of granulocyte-macrophage colony stimulating factor loaded hierarchically 3D nanofiber scaffolds in a humanized mice model. Front Bioeng Biotechnol 2023; 11:1159068. [PMID: 37034265 PMCID: PMC10080111 DOI: 10.3389/fbioe.2023.1159068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Background: Immune evaluation of biomaterials for tissue regeneration is a critical preclinical evaluation. The current evaluation criterion (ISO 10993-1 or GB/T 16886) uses rodents to perform the immune evaluation. However, the immune system of rodents is different from humans, the obtained results may not be reliable, which could lead directly to the failure of clinical trials. Granulocyte-macrophage colony-stimulating factor (GM-CSF) shows a great potential application in tissue regeneration by regulating local immune responses. The presented work combines the advantages of GM-CSF (immunoregulation) and hierarchically 3D nanofiber scaffolds (tissue regeneration). Methods: Firstly, we fabricated GM-CSF loaded 3D radially aligned nanofiber scaffolds, and then subcutaneous implantation was performed in humanized mice. The whole scaffold and surrounding tissue were harvested at each indicated time point. Finally, the cell infiltration and local immune responses were detected by histological observations, including H&E and Masson staining and immunochemistry. Results: We found significant cell migration and extracellular matrix deposition within the 3D radially aligned nanofiber scaffold after subcutaneous implantation. The locally released GM-CSF could accelerate the expression of human dendritic cells (CD11c) only 3 days after subcutaneous implantation. Moreover, higher expression of human cytotoxic T cells (CD3+/CD8+), M1 macrophages (CD68/CCR7) was detected within GM-CSF loaded radially aligned nanofiber scaffolds and their surrounding tissues. Conclusions: The 3D radially aligned scaffold can accelerate cell migration from surrounding tissues to regenerate the wound area. And the locally released GM-CSF enhances dendritic cell recruitment and activation of cytotoxic T cells and M1 macrophages. Taken together, the GM-CSF loaded 3D radially aligned nanofiber scaffolds have a promising potential for achieving tissue regeneration.
Collapse
Affiliation(s)
- Rui Chen
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Yujie Li
- Department of Plastic, Reconstructive and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yangyang Zhuang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiming Zhang
- Department of Plastic, Reconstructive and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hailong Wu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China
- *Correspondence: Hailong Wu, ; Tao Lin, ; Shixuan Chen,
| | - Tao Lin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, China
- *Correspondence: Hailong Wu, ; Tao Lin, ; Shixuan Chen,
| | - Shixuan Chen
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
- *Correspondence: Hailong Wu, ; Tao Lin, ; Shixuan Chen,
| |
Collapse
|
5
|
Zhu H, Xing C, Dou X, Zhao Y, Peng Y, Feng C, Fang Y. Chiral Hydrogel Accelerates Re-Epithelization in Chronic Wounds via Mechanoregulation. Adv Healthc Mater 2022; 11:e2201032. [PMID: 36052735 DOI: 10.1002/adhm.202201032] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/17/2022] [Indexed: 01/28/2023]
Abstract
Chronic wounds, such as diabetic foot ulcers (DFU), are a serious clinical problem. It is a challenge for the conventional wound dressings to achieve the desirable therapeutic efficacy due to the lack of biomimetic structural environment for rapid re-epithelization. Inspired by the naturally existing chiral structures in skin, a novel amino acid-based chiral hydrogel dressing is developed, consisting of left-handed or right-handed helical fibers self-assembled by l/d-phenylalanine derivatives. Compared to the levorotatory chiral hydrogel (LH), the dextral chiral hydrogel (DH) shows the ability to enhance cell adhesion, proliferation, and migration, and strongly promotes diabetic wound healing and re-epithelialization with a drug-free mode. Interestingly, the dextral chiral hydrogel is able to actively increase adsorption of type I collagen and promote proliferation and migration of keratinocyte in an integrin and YAP-mediated manner. This study not only provides a novel strategy for treatment of chronic wounds by utilizing dextral chiral hydrogel dressings, but also unveils the molecular mechanism for effect of dextral chiral structures on the promoted proliferation of keratinocyte.
Collapse
Affiliation(s)
- Hanting Zhu
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China.,Institute of Traumatic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
| | - Chao Xing
- State Key Lab of Metal Matrix Composites, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoqiu Dou
- State Key Lab of Metal Matrix Composites, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yu Zhao
- State Key Lab of Metal Matrix Composites, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yinbo Peng
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China.,Institute of Traumatic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
| | - Chuanliang Feng
- State Key Lab of Metal Matrix Composites, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yong Fang
- Department of Burns and Plastic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China.,Institute of Traumatic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 201900, China
| |
Collapse
|
6
|
Zhou H, Zheng D, Wang H, Wu Y, Peng X, Li Q, Li T, Liu L. The protective effects of pericyte-derived microvesicles on vascular endothelial functions via CTGF delivery in sepsis. Cell Commun Signal 2021; 19:115. [PMID: 34784912 PMCID: PMC8594111 DOI: 10.1186/s12964-021-00795-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/16/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND It is well known that sepsis is a prevalent severe disease caused by infection and the treatment strategies are limited. Recently pericyte-derived microvesicles (PMVs) were confirmed to be therapeutic in many diseases, whether PMVs can protect vascular endothelial cell (VEC) injury is unknown. METHODS Pericytes were extracted from the retina of newly weaned rats, and PMVs were collected after starvation and characterized by flow-cytometry and transmission electron microscopy. First, the effect of PMVs on pulmonary vascular function in septic rats was measured via intravenous administration with HE staining, immunofluorescence, and Elisa analysis. Then, PMVs were co-incubated with VECs in the presence of lipopolysaccharide (LPS), and observed the protective effect of PMVs on VECs. Next, the proteomic analysis and further Gene Ontology (GO) enrichment analysis were performed to analyze the therapeutic mechanism of PMVs, and the angiogenesis-related protein CTGF was highly expressed in PMVs. Finally, by CTGF upregulation and downregulation in PMV, the role of PMV-carried CTGF was investigated. RESULTS PMVs restored the proliferation and angiogenesis ability of pulmonary VECs, and alleviated pulmonary vascular leakage in septic rats and LPS-stimulated VECs. Further study showed that PMVs delivered CTGF to VECs, and subsequently activated ERK1/2, and increased the phosphorylation of STAT3, thereby improving the function of VECs. The further study found CD44 mediated the absorption and internalization of PMVs to VECs, the anti-CD44 antibody inhibited the protective effect of PMVs. CONCLUSIONS PMVs may delivery CTGF to VECs, and promote the proliferation and angiogenesis ability by activating the CTGF-ERK1/2-STAT3 axis, thereby protecting pulmonary vascular function in sepsis. The therapeutic effect of PMVs was highly related to CD44-mediated absorption. Video Abstract.
Collapse
Affiliation(s)
- Henan Zhou
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Army Medical Center of PLA, Daping Hospital, Army Medical University, No.10th Daping Changjiang Road, Chongqing, 400038 China
| | - Danyang Zheng
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Army Medical Center of PLA, Daping Hospital, Army Medical University, No.10th Daping Changjiang Road, Chongqing, 400038 China
- Intensive Care Unit, General Hospital of Central Theater Command, Wuhan, 430064 China
| | - Hongchen Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Army Medical Center of PLA, Daping Hospital, Army Medical University, No.10th Daping Changjiang Road, Chongqing, 400038 China
| | - Yue Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Army Medical Center of PLA, Daping Hospital, Army Medical University, No.10th Daping Changjiang Road, Chongqing, 400038 China
| | - Xiaoyong Peng
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Army Medical Center of PLA, Daping Hospital, Army Medical University, No.10th Daping Changjiang Road, Chongqing, 400038 China
| | - Qinghui Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Army Medical Center of PLA, Daping Hospital, Army Medical University, No.10th Daping Changjiang Road, Chongqing, 400038 China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Army Medical Center of PLA, Daping Hospital, Army Medical University, No.10th Daping Changjiang Road, Chongqing, 400038 China
| | - Liangming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Army Medical Center of PLA, Daping Hospital, Army Medical University, No.10th Daping Changjiang Road, Chongqing, 400038 China
| |
Collapse
|
7
|
Li M, Liu Y, Fu Y, Gong R, Xia H, Huang X, Wu Y. Interleukin-35 inhibits lipopolysaccharide-induced endothelial cell activation by downregulating inflammation and apoptosis. Exp Cell Res 2021; 407:112784. [PMID: 34508746 DOI: 10.1016/j.yexcr.2021.112784] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 01/20/2023]
Abstract
Inflammation is an essential factor contributing to sepsis-induced endothelial cell (EC) activation. Interleukin-35 (IL-35) is an anti-inflammatory/immunosuppressive cytokine that exerts protective effects on many inflammatory diseases. In this study, we investigated the effects of IL-35 on lipopolysaccharide (LPS)-induced EC activation and the potential underlying mechanism. Human umbilical vein endothelial cells (HUVECs) were incubated with LPS (1 μg/ml) for 24 h and then cocultured with different concentrations (0, 1, 10, or 100 ng/ml) of recombinant human IL-35 (rhIL-35) for 12 h. Flow cytometry analysis revealed that IL-35 inhibited LPS-induced HUVEC apoptosis in a dose-dependent manner. RT-qPCR and Western blot analyses showed significantly higher mRNA and protein levels of the adhesion molecules intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) and the inflammatory factors IL-6 and IL-8 in the LPS group than in the control group. These changes were alleviated by IL-35 treatment, suggesting that IL-35 protects ECs by downregulating inflammation. Furthermore, IL-35 induced signal transducer and activator of transcription 1 (STAT1) and STAT4 activation and promoted their interaction. Blocking STAT1 or STAT4 expression by fludarabine (STAT1 inhibitor) treatment or siRNA-STAT4-interfering fragment transfection inhibited the protective effect of IL-35 on ECs. Moreover, we observed a similar protective effect of IL-35 treatment on ECs in a mouse sepsis model induced by intraperitoneal LPS injection. This study indicated that IL-35 exerts anti-inflammatory and antiapoptotic effects on LPS-induced EC activation by activating the STAT1 and STAT4 signaling pathways.
Collapse
Affiliation(s)
- Meng Li
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Yue Liu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Yang Fu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Ren Gong
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Huasong Xia
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Xiao Huang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
8
|
Kronstadt SM, Pottash AE, Levy D, Wang S, Chao W, Jay SM. Therapeutic Potential of Extracellular Vesicles for Sepsis Treatment. ADVANCED THERAPEUTICS 2021; 4:2000259. [PMID: 34423113 PMCID: PMC8378673 DOI: 10.1002/adtp.202000259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Indexed: 12/14/2022]
Abstract
Sepsis is a deadly condition lacking a specific treatment despite decades of research. This has prompted the exploration of new approaches, with extracellular vesicles (EVs) emerging as a focal area. EVs are nanosized, cell-derived particles that transport bioactive components (i.e., proteins, DNA, and RNA) between cells, enabling both normal physiological functions and disease progression depending on context. In particular, EVs have been identified as critical mediators of sepsis pathophysiology. However, EVs are also thought to constitute the biologically active component of cell-based therapies and have demonstrated anti-inflammatory, anti-apoptotic, and immunomodulatory effects in sepsis models. The dual nature of EVs in sepsis is explored here, discussing their endogenous roles and highlighting their therapeutic properties and potential. Related to the latter component, prior studies involving EVs from mesenchymal stem/stromal cells (MSCs) and other sources are discussed and emerging producer cells that could play important roles in future EV-based sepsis therapies are identified. Further, how methodologies could impact therapeutic development toward sepsis treatment to enhance and control EV potency is described.
Collapse
Affiliation(s)
- Stephanie M Kronstadt
- Fischell Department of Bioengineering, University of Maryland, 3102 A. James Clark Hall, College Park, MD 20742, USA
| | - Alex E Pottash
- Fischell Department of Bioengineering, University of Maryland, 3102 A. James Clark Hall, College Park, MD 20742, USA
| | - Daniel Levy
- Fischell Department of Bioengineering, University of Maryland, 3102 A. James Clark Hall, College Park, MD 20742, USA
| | - Sheng Wang
- Translational Research Program, Department of Anesthesiology and Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology and Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Steven M Jay
- Fischell Department of Bioengineering and Program in Molecular and, Cell Biology, University of Maryland, 3102 A. James Clark Hall, College Park, MD 20742, USA
| |
Collapse
|
9
|
Tang F, Tie Y, Hong W, Wei Y, Tu C, Wei X. Targeting Myeloid-Derived Suppressor Cells for Premetastatic Niche Disruption After Tumor Resection. Ann Surg Oncol 2021; 28:4030-4048. [PMID: 33258011 PMCID: PMC7703739 DOI: 10.1245/s10434-020-09371-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/29/2020] [Indexed: 02/05/2023]
Abstract
Surgical resection is a common therapeutic option for primary solid tumors. However, high cancer recurrence and metastatic rates after resection are the main cause of cancer related mortalities. This implies the existence of a "fertile soil" following surgery that facilitates colonization by circulating cancer cells. Myeloid-derived suppressor cells (MDSCs) are essential for premetastatic niche formation, and may persist in distant organs for up to 2 weeks after surgery. These postsurgical persistent lung MDSCs exhibit stronger immunosuppression compared with presurgical MDSCs, suggesting that surgery enhances MDSC function. Surgical stress and trauma trigger the secretion of systemic inflammatory cytokines, which enhance MDSC mobilization and proliferation. Additionally, damage associated molecular patterns (DAMPs) directly activate MDSCs through pattern recognition receptor-mediated signals. Surgery also increases vascular permeability, induces an increase in lysyl oxidase and extracellular matrix remodeling in lungs, that enhances MDSC mobilization. Postsurgical therapies that inhibit the induction of premetastatic niches by MDSCs promote the long-term survival of patients. Cyclooxygenase-2 inhibitors and β-blockade, or their combination, may minimize the impact of surgical stress on MDSCs. Anti-DAMPs and associated inflammatory signaling inhibitors also are potential therapies. Existing therapies under tumor-bearing conditions, such as MDSCs depletion with low-dose chemotherapy or tyrosine kinase inhibitors, MDSCs differentiation using all-trans retinoic acid, and STAT3 inhibition merit clinical evaluation during the perioperative period. In addition, combining low-dose epigenetic drugs with chemokine receptors, reversing immunosuppression through the Enhanced Recovery After Surgery protocol, repairing vascular leakage, or inhibiting extracellular matrix remodeling also may enhance the long-term survival of curative resection patients.
Collapse
Affiliation(s)
- Fan Tang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Department of Orthopeadics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yan Tie
- Department of Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, People's Republic of China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Chongqi Tu
- Department of Orthopeadics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
10
|
Li S, Wang F, Sun D. The renal microcirculation in chronic kidney disease: novel diagnostic methods and therapeutic perspectives. Cell Biosci 2021; 11:90. [PMID: 34001267 PMCID: PMC8130426 DOI: 10.1186/s13578-021-00606-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) affects 8–16% of the population worldwide and is characterized by fibrotic processes. Understanding the cellular and molecular mechanisms underpinning renal fibrosis is critical to the development of new therapeutics. Microvascular injury is considered an important contributor to renal progressive diseases. Vascular endothelium plays a significant role in responding to physical and chemical signals by generating factors that help maintain normal vascular tone, inhibit leukocyte adhesion and platelet aggregation, and suppress smooth muscle cell proliferation. Loss of the rich capillary network results in endothelial dysfunction, hypoxia, and inflammatory and oxidative effects and further leads to the imbalance of pro- and antiangiogenic factors, endothelial cell apoptosis and endothelial-mesenchymal transition. New techniques, including both invasive and noninvasive techniques, offer multiple methods to observe and monitor renal microcirculation and guide targeted therapeutic strategies. A better understanding of the role of endothelium in CKD will help in the development of effective interventions for renal microcirculation improvement. This review focuses on the role of microvascular injury in CKD, the methods to detect microvessels and the novel treatments to ameliorate renal fibrosis.
Collapse
Affiliation(s)
- Shulin Li
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Fei Wang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China. .,Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
11
|
León-Moreno LC, Castañeda-Arellano R, Aguilar-García IG, Desentis-Desentis MF, Torres-Anguiano E, Gutiérrez-Almeida CE, Najar-Acosta LJ, Mendizabal-Ruiz G, Ascencio-Piña CR, Dueñas-Jiménez JM, Rivas-Carrillo JD, Dueñas-Jiménez SH. Kinematic Changes in a Mouse Model of Penetrating Hippocampal Injury and Their Recovery After Intranasal Administration of Endometrial Mesenchymal Stem Cell-Derived Extracellular Vesicles. Front Cell Neurosci 2020; 14:579162. [PMID: 33192324 PMCID: PMC7533596 DOI: 10.3389/fncel.2020.579162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/14/2020] [Indexed: 12/20/2022] Open
Abstract
Locomotion speed changes appear following hippocampal injury. We used a hippocampal penetrating brain injury mouse model to analyze other kinematic changes. We found a significant decrease in locomotion speed in both open-field and tunnel walk tests. We described a new quantitative method that allows us to analyze and compare the displacement curves between mice steps. In the tunnel walk, we marked mice with indelible ink on the knee, ankle, and metatarsus of the left and right hindlimbs to evaluate both in every step. Animals with hippocampal damage exhibit slower locomotion speed in both hindlimbs. In contrast, in the cortical injured group, we observed significant speed decrease only in the right hindlimb. We found changes in the displacement patterns after hippocampal injury. Mesenchymal stem cell-derived extracellular vesicles had been used for the treatment of several diseases in animal models. Here, we evaluated the effects of intranasal administration of endometrial mesenchymal stem cell-derived extracellular vesicles on the outcome after the hippocampal injury. We report the presence of vascular endothelial growth factor, granulocyte–macrophage colony-stimulating factor, and interleukin 6 in these vesicles. We observed locomotion speed and displacement pattern preservation in mice after vesicle treatment. These mice had lower pyknotic cells percentage and a smaller damaged area in comparison with the nontreated group, probably due to angiogenesis, wound repair, and inflammation decrease. Our results build up on the evidence of the hippocampal role in walk control and suggest that the extracellular vesicles could confer neuroprotection to the damaged hippocampus.
Collapse
Affiliation(s)
- Lilia Carolina León-Moreno
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico.,Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Rolando Castañeda-Arellano
- Laboratory of Tissue Engineering and Transplant, Department of Physiology, cGMP Cell Processing Facility, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Irene Guadalupe Aguilar-García
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | | | - Elizabeth Torres-Anguiano
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Coral Estefanía Gutiérrez-Almeida
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Luis Jesús Najar-Acosta
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Gerardo Mendizabal-Ruiz
- Department of Computer Sciences, University Center of Exact Sciences and Engineering, University of Guadalajara, Guadalajara, Mexico
| | - César Rodolfo Ascencio-Piña
- Department of Computer Sciences, University Center of Exact Sciences and Engineering, University of Guadalajara, Guadalajara, Mexico
| | - Judith Marcela Dueñas-Jiménez
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Jorge David Rivas-Carrillo
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Sergio Horacio Dueñas-Jiménez
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
12
|
Wu Q, Yuan X, Li B, Han R, Zhang H, Xiu R. Salvianolic Acid Alleviated Blood-Brain Barrier Permeability in Spontaneously Hypertensive Rats by Inhibiting Apoptosis in Pericytes via P53 and the Ras/Raf/MEK/ERK Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1523-1534. [PMID: 32368011 PMCID: PMC7170553 DOI: 10.2147/dddt.s245959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/15/2020] [Indexed: 01/02/2023]
Abstract
Objective To investigate the effect of salvianolic acid A (SA) on the permeability of blood–brain barrier (BBB) and brain microvascular pericyte apoptosis in spontaneously hypertensive rats (SHR). Methods Evans Blue was used to determine the BBB permeability in control rats and SHR. Western blotting was used to evaluate the expression levels of relevant proteins in the pericytes isolated from the differentially treated animals. An in vitro model of hypertension was established by stimulating pericytes with angiopoietin-2 (Ang2). MTT assay was used to assess cell viability, and apoptosis and cell cycle distribution were analyzed by flow cytometry. Results SA attenuated BBB permeability in SHR in a dose-dependent manner. It downregulated pro-apoptotic proteins including p53, p21, Fas, FasL, cleaved-caspase 3/caspase 3 and Bax in the pericytes of SHR and upregulated CDK6, cyclin D1, CDK2, cyclin E and Bcl2. In addition, SA activated the Ras/Raf/MEK/ERK pathway in a dose-dependent manner by increasing the levels of Ras, Raf, p-MEK1, p-MEK2, p-ERK1 and p-ERK2. Finally, SA reduced Ang2-induced apoptosis of cerebral microvessels pericytes and decreased the proportion of cells in the G0/G1 phase of the cell cycle by inhibiting the p53 pathway and activating the Ras/Raf/MEK/ERK pathway. Conclusion SA reduced BBB permeability in spontaneously hypertensive rats, possibly by inhibiting Ang2-induced apoptosis of pericytes by activating the Ras/Raf/MEK/ERK pathway.
Collapse
Affiliation(s)
- Qingbin Wu
- Institute of Microcirculation, Chinese Academy Medical Sciences & Pecking Union Medical College
| | - Xiaochen Yuan
- Institute of Microcirculation, Chinese Academy Medical Sciences & Pecking Union Medical College
| | - Bingwei Li
- Institute of Microcirculation, Chinese Academy Medical Sciences & Pecking Union Medical College
| | - Ruiqin Han
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, People's Republic of China
| | - Honggang Zhang
- Institute of Microcirculation, Chinese Academy Medical Sciences & Pecking Union Medical College
| | - Ruijuan Xiu
- Institute of Microcirculation, Chinese Academy Medical Sciences & Pecking Union Medical College
| |
Collapse
|
13
|
Pang M, Lei X, Yao Z, Chen C, Cheng B. Nonselective alpha-/beta- AR antagonists can inhibit pericyte proliferation, migration, and secretion in vitro. Clin Hemorheol Microcirc 2020; 75:313-323. [PMID: 32224529 DOI: 10.3233/ch-190780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
It has been reported that the beta-adrenergic receptor blocker (propranolol) and the a-adrenergic receptor (AR) blocker (phentolamine) both can inhibit human endothelial cell (EC) angiogenesis in vitro. However, it is unknown whether this inhibition also acts on pericytes. The present study aimed to determine how pericytes react to treatment with an a-/β- AR blocker. In the study, cell proliferation assays and scratch assay were performed to assess the effect of phentolamine or propranolol on cell proliferation and migration. Western blot and ELISA were employed to determine changes in VEGF-A and Ang-1 expression levels. The results indicated that the nonselective a-/β- AR blocker inhibited the proliferation, migration, and secretion of pericytes. The use of the nonselective a-/β- AR blocker might have an impact on vascularization and vascular maturation. Our research suggests the rational use of nonselective a-/β- AR blockers to treat angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Mengru Pang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, PLA, Guangzhou, China
| | - Xiaoxuan Lei
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, PLA, Guangzhou, China
| | - Zexin Yao
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, PLA, Guangzhou, China.,Guangdong Pharmaceutical University, Guangzhou, China
| | - Caihong Chen
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, PLA, Guangzhou, China.,Guangdong Pharmaceutical University, Guangzhou, China
| | - Biao Cheng
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, PLA, Guangzhou, China.,The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Center of Wound Treatment, General Hospital of Southern Theater Command, PLA, Guangzhou, China.,The Key Laboratory of Trauma Treatment and Tissue Repair of Tropical Area, PLA, Guangzhou, China
| |
Collapse
|
14
|
Evdokiou A, Kanisicak O, Gierek S, Barry A, Ivey MJ, Zhang X, Bodnar RJ, Satish L. Characterization of Burn Eschar Pericytes. J Clin Med 2020; 9:jcm9020606. [PMID: 32102389 PMCID: PMC7074206 DOI: 10.3390/jcm9020606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 02/19/2020] [Indexed: 12/31/2022] Open
Abstract
Pericytes are cells that reside adjacent to microvasculature and regulate vascular function. Pericytes gained great interest in the field of wound healing and regenerative medicine due to their multipotential fate and ability to enhance angiogenesis. In burn wounds, scarring and scar contractures are the major pathologic feature and cause loss of mobility. The present study investigated the influence of burn wound environment on pericytes during wound healing. Pericytes isolated from normal skin and tangentially excised burn eschar tissues were analyzed for differences in gene and protein expression using RNA-seq., immunocytochemistry, and ELISA analyses. RNA-seq identified 443 differentially expressed genes between normal- and burn eschar-derived pericytes. Whereas, comparing normal skin pericytes to normal skin fibroblasts identified 1021 distinct genes and comparing burn eschar pericytes to normal skin fibroblasts identified 2449 differential genes. Altogether, forkhead box E1 (FOXE1), a transcription factor, was identified as a unique marker for skin pericytes. Interestingly, FOXE1 levels were significantly elevated in burn eschar pericytes compared to normal. Additionally, burn wound pericytes showed increased expression of profibrotic genes periostin, fibronectin, and endosialin and a gain in contractile function, suggesting a contribution to scarring and fibrosis. Our findings suggest that the burn wound environment promotes pericytes to differentiate into a myofibroblast-like phenotype promoting scar formation and fibrosis.
Collapse
Affiliation(s)
- Alexander Evdokiou
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA; (O.K.); (M.J.I.)
| | - Stephanie Gierek
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
| | - Amanda Barry
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
| | - Malina J. Ivey
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA; (O.K.); (M.J.I.)
| | - Xiang Zhang
- Genomics, Epigenomics and Sequencing Core, University of Cincinnati, Cincinnati, OH 45267, USA;
| | - Richard J. Bodnar
- Veterans Affairs Medical Center, University Dr. C, Pittsburgh, PA 15240, USA;
| | - Latha Satish
- Shriners Hospitals for Children, Research Department, Cincinnati, OH 45229, USA; (A.E.); (S.G.); (A.B.)
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA; (O.K.); (M.J.I.)
- Correspondence: or ; Tel.: +1-513-872-6278
| |
Collapse
|
15
|
Spampinato SF, Bortolotto V, Canonico PL, Sortino MA, Grilli M. Astrocyte-Derived Paracrine Signals: Relevance for Neurogenic Niche Regulation and Blood-Brain Barrier Integrity. Front Pharmacol 2019; 10:1346. [PMID: 31824311 PMCID: PMC6881379 DOI: 10.3389/fphar.2019.01346] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 10/24/2019] [Indexed: 01/12/2023] Open
Abstract
Astrocytes are essential for proper regulation of the central nervous system (CNS). Importantly, these cells are highly secretory in nature. Indeed they can release hundreds of molecules which play pivotal physiological roles in nervous tissues and whose abnormal regulation has been associated with several CNS disorders. In agreement with these findings, recent studies have provided exciting insights into the key contribution of astrocyte-derived signals in the pleiotropic functions of these cells in brain health and diseases. In the future, deeper analysis of the astrocyte secretome is likely to further increase our current knowledge on the full potential of these cells and their secreted molecules not only as active participants in pathophysiological events, but as pharmacological targets or even as therapeutics for neurological and psychiatric diseases. Herein we will highlight recent findings in our and other laboratories on selected molecules that are actively secreted by astrocytes and contribute in two distinct functions with pathophysiological relevance for the astroglial population: i) regulation of neural stem cells (NSCs) and their progeny within adult neurogenic niches; ii) modulation of the blood–brain barrier (BBB) integrity and function.
Collapse
Affiliation(s)
- Simona Federica Spampinato
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Valeria Bortolotto
- Laboratory of Neuroplasticity, Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Pier Luigi Canonico
- Laboratory of Neuroplasticity, Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Maria Angela Sortino
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Mariagrazia Grilli
- Laboratory of Neuroplasticity, Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
16
|
Angiopoietin-1 Promotes the Integrity of Neovascularization in the Subcutaneous Matrigel of Type 1 Diabetic Rats. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2016972. [PMID: 30729120 PMCID: PMC6343146 DOI: 10.1155/2019/2016972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/30/2018] [Indexed: 12/21/2022]
Abstract
Objective This study aimed to investigate the effects of Ang-1 on neovascularization of diabetic organs by subcutaneous Matrigel angiogenesis model, established in type 1 diabetic rats. Methods Ang-1 adenoviral vector was constructed. The rat model was established by STZ and divided into four group. The Matrigel was inserted subcutaneously into the abdominal cavity of rats at 8 weeks, the treatment group was injected with Ang-1 adenovirus vector via tail vein, and the rats were sacrificed at 10 weeks. Neovascularization of Matrigel was observed with transmission electron microscopy. The marker of vascular endothelial cell and pericyte were detected by immunofluorescence. Immunohistochemical detection of the neovascular endothelial junction protein was performed. RT-PCR was used to determine protein expression of neovascular in Matrigel. Results Vascular cavity-like structure could be seen in subcutaneous Matrigel of diabetic rats, and the cavity was filled with a lot of red blood cells. Transmission electron microscopy showed that neovascular endothelial structure of the Matrigel was incomplete, while the Ang-1 treatment group had more vascular cavity-like structures, intact vascular endothelial structure, and reduced inflammatory cell infiltration in Matrigel. Additionally, the integrity of vascularization improved, and the marker of pericyte and the cell tight junctions protein was upregulated in Ang-1 treatment group. Conclusion Hyperglycemia could induce pathological angiogenesis in subcutaneous Matrigel of diabetic rats, and Ang-1 could upregulate the expression of intercellular junction protein in subcutaneous Matrigel of diabetic rats and promote the integrity of neovascularization in the subcutaneous Matrigel of diabetic rats.
Collapse
|
17
|
Zhong X, Zheng Y, Li H, Huang S, Ge J. Identification of Myocardial Telocytes and Bone Marrow Mesenchymal Stem Cells in Mice. Cell Transplant 2018; 27:1515-1522. [PMID: 30203685 PMCID: PMC6180723 DOI: 10.1177/0963689718796773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES The aim of this study was to compare the morphology, immune phenotype, and cytokine profiles between myocardial telocytes (TCs) and bone marrow mesenchymal stem cells (MSCs), and explore the difference between those two types of interstitial cells. METHODS TCs and MSCs were cultured in vitro and cell morphology was observed with a light microscope. The expression levels of CD34, c-kit, and vimentin were detected by immunofluorescence, RT-qPCR, and Western blotting in both TCs and MSCs. The related supernatant was collected and total of 49 cytokine profiles were detected by RayBio Mice Cytokine Antibody Array. Significantly different cytokines were further confirmed by ELISA. RESULTS TCs have small cellular body and very long prolongations and they were CD34+/c-kit+/vimentin+, whereas MSCs have no telopodes and they were CD34-/c-kit- /vimentin+. Cytokine profile analysis and ELISA showed that 19 of 49 cytokines were increased dramatically in the supernatant of TCs compared with those of MSCs. Moreover, 9 of 19 cytokines were increased 2-fold at least in the supernatant of TCs compared with those of MSCs. Of 49 cytokines, 30 exhibited no significant changes in the supernatant of TCs compared with those of MSCs. CONCLUSIONS Using various technologies, we identified that myocardial TCs and MSCs are significantly different in terms of cell structure and cytokine profiles.
Collapse
Affiliation(s)
- Xin Zhong
- 1 Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Xin Zhong and Yonghua Zheng contributed equally to this work
| | - Yonghua Zheng
- 2 Department of Respiratory Medicine, Shanghai Jinshan Tinglin Hospital, Shanghai, China.,Xin Zhong and Yonghua Zheng contributed equally to this work
| | - Hua Li
- 1 Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,3 Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Steve Huang
- 1 Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junbo Ge
- 1 Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,3 Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Haubruck P, Heller R, Tanner MC, Daniel V, Schmidmaier G, Bolourchi F, Moghaddam A, Fischer C. A Preliminary Study of Contrast-Enhanced Ultrasound (CEUS) and Cytokine Expression Analysis (CEA) as Early Predictors for the Outcome of Tibial Non-Union Therapy. Diagnostics (Basel) 2018; 8:diagnostics8030055. [PMID: 30149501 PMCID: PMC6164610 DOI: 10.3390/diagnostics8030055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/19/2018] [Accepted: 08/22/2018] [Indexed: 01/09/2023] Open
Abstract
The current study investigates if contrast-enhanced ultrasound (CEUS) or cytokine expression analysis (CEA) evaluating vascularization are capable of predicting the outcome of non-union therapy. Patients with tibial non-unions were surgically treated and participated in our follow-up program including perioperative collection of blood as well as CEUS analysis. Two groups were formed: Responders in group 1 (G1, N = 8) and Non-Responders in group 2 (G2, N = 5). Serum cytokine expression and local microperfusion were compared and correlated to the radiologic outcome. Evaluation of TNF-α expression revealed significantly lower values prior to first surgery in G1 (G1: 9.66 ± 0.96 pg/mL versus G2: 12.63 ± 1.2 pg/mL; p = 0.045); whereas after treatment both CEA and CEUS indicated a higher potential for angiogenesis in Responders. Logistic regression modelling revealed the highest predictive power regarding eventual osseous consolidation for the combination of both CEUS and serum CEA. The results provide first evidence regarding a link between changes in the serum expression of distinct pro-angiogenic cytokines and alterations in the local microperfusion assessed via both non-invasive and radiation-free diagnostic modalities. In addition, a combination of CEUS and CEA is a promising novel tool in early prediction of the outcome of non-union therapy.
Collapse
Affiliation(s)
- Patrick Haubruck
- HTRG-Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Institute of Bone and Joint Research, University of Sydney, St. Leonards, New South Wales 2065, Australia.
| | - Raban Heller
- HTRG-Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
| | - Michael C Tanner
- HTRG-Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
| | - Volker Daniel
- Department of Transplantation Immunology, Institute of Immunology, University of Heidelberg, Im Neuenheimer Feld 305, 69120 Heidelberg, Germany.
| | - Gerhard Schmidmaier
- HTRG-Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
| | - Farhoud Bolourchi
- HTRG-Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
| | - Arash Moghaddam
- HTRG-Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
- ATORG-Aschaffenburg Trauma and Orthopedic Research Group, Center for Trauma Surgery, Orthopedics and Sports Medicine, Am Hasenkopf 1, 63739 Aschaffenburg, Germany.
| | - Christian Fischer
- HTRG-Heidelberg Trauma Research Group, Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
| |
Collapse
|