1
|
Chen C, Zheng X, Wang C, Zhou H, Zhang Y, Ye T, Yang Y. CTHRC1 Attenuates Tendinopathy via Enhancing EGFR/MAPK Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2406611. [PMID: 39540237 DOI: 10.1002/advs.202406611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Tendinopathy poses a formidable challenge due to the inherent limitations of tendon regenerative capabilities post-injury. At present, effective curative approaches for tendinopathy are still lacking. Collagen triple helix repeat-containing 1 (CTHRC1) is an extracellular matrix protein with significant roles in both physiological and pathological processes. The present study aims to investigate the function and underlying mechanism of CTHRC1 in tendinopathy. In this study, CTHRC1 is identified as a potential effector in promoting tendon regeneration through multi-proteomic analysis of Achilles tendon tissues in mice. In vitro, CTHRC1 enhances the proliferation, migration, and tenogenic differentiation of tendon stem/progenitor cell (TSPC). In vivo, CTHRC1 deletion impairs tendon healing, while its overexpression reverses the detrimental effects caused by CTHRC1 deficiency. Mechanistically, proteomics on TSPC stimulated with recombinant CTHRC1 reveal that CTHRC1 activates the mitogen-activated protein kinase (MAPK) signaling pathway via binding to epidermal growth factor receptor (EGFR), which in turn promotes the proliferative, migrative, and tenogenic capacities of TSPC to attenuate Achilles tendinopathy. Conversely, inhibiting EGFR reverses the tendon-healing effect of CRHRC1. The study demonstrates that CTHRC1 can promote the proliferative, migrative, and tenogenic capacities of TSPC, ultimately facilitating tendon healing through activating the EGFR/MAPK signaling pathway. CTHRC1 holds promise as a potential intervention for tendinopathy.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Xu Zheng
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai, 200001, China
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Cheng Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - HaiChao Zhou
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Yi Zhang
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - TianBao Ye
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361008, China
- Shanghai Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - YunFeng Yang
- School of Medicine, Tongji University, Shanghai, 200092, China
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
2
|
Gnanagurusamy J, Krishnamoorthy S, Muthusami S. Transforming growth factor-β micro-environment mediated immune cell functions in cervical cancer. Int Immunopharmacol 2024; 140:112837. [PMID: 39111147 DOI: 10.1016/j.intimp.2024.112837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/02/2024] [Accepted: 07/28/2024] [Indexed: 09/01/2024]
Abstract
Propensity to develop cervical cancer (CC) in human papilloma virus (HPV) infected individual could potentially involve the impaired immune functioning. Several stages of HPV surveillance by immune cells in tumor micro-environment (TME) is regulated mainly by transforming growth factor-beta (TGF-β) and is crucial for the establishment of CC. The role of TGF-β in the initiation and progression of CC is very complex and involve different suppressor of mothers against decapentaplegic homolog (SMAD) dependent and SMAD independent signaling mechanism(s). This review summarizes the handling of HPV by immune cells such as T lymphocytes, B lymphocytes, natural killer cells (NK), dendritic cells (DC), monocytes, macrophages, myeloid derived suppressor cells (MDSC) and their regulation by TGF-β. The hijack mechanisms adapted by HPV to evade this surveillance process is discussed. Biomarkers indicating the stages of CC and immune checkpoints that can be targeted for improved outcome are included for immune-based theragnostics. This review also addresses the direct actions of TGF-β on CC cells and tumor/immune cell interactions. Therapies focused on targeting TGF-β using small molecule inhibitors, monoclonal antibodies and TGF-β chimeric antigen receptor (CAR)T cells are collated to understand the current strategies related to TGF-β in the management of CC.
Collapse
Affiliation(s)
- Jayapradha Gnanagurusamy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India
| | - Sneha Krishnamoorthy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India
| | - Sridhar Muthusami
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India; Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India.
| |
Collapse
|
3
|
Shantaraman A, Dammer EB, Ugochukwu O, Duong DM, Yin L, Carter EK, Gearing M, Chen-Plotkin A, Lee EB, Trojanowski JQ, Bennett DA, Lah JJ, Levey AI, Seyfried NT, Higginbotham L. Network proteomics of the Lewy body dementia brain reveals presynaptic signatures distinct from Alzheimer's disease. Mol Neurodegener 2024; 19:60. [PMID: 39107789 PMCID: PMC11302177 DOI: 10.1186/s13024-024-00749-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Lewy body dementia (LBD), a class of disorders comprising Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB), features substantial clinical and pathological overlap with Alzheimer's disease (AD). The identification of biomarkers unique to LBD pathophysiology could meaningfully advance its diagnosis, monitoring, and treatment. Using quantitative mass spectrometry (MS), we measured over 9,000 proteins across 138 dorsolateral prefrontal cortex (DLPFC) tissues from a University of Pennsylvania autopsy collection comprising control, Parkinson's disease (PD), PDD, and DLB diagnoses. We then analyzed co-expression network protein alterations in those with LBD, validated these disease signatures in two independent LBD datasets, and compared these findings to those observed in network analyses of AD cases. The LBD network revealed numerous groups or "modules" of co-expressed proteins significantly altered in PDD and DLB, representing synaptic, metabolic, and inflammatory pathophysiology. A comparison of validated LBD signatures to those of AD identified distinct differences between the two diseases. Notably, synuclein-associated presynaptic modules were elevated in LBD but decreased in AD relative to controls. We also found that glial-associated matrisome signatures consistently elevated in AD were more variably altered in LBD, ultimately stratifying those LBD cases with low versus high burdens of concurrent beta-amyloid deposition. In conclusion, unbiased network proteomic analysis revealed diverse pathophysiological changes in the LBD frontal cortex distinct from alterations in AD. These results highlight the LBD brain network proteome as a promising source of biomarkers that could enhance clinical recognition and management.
Collapse
Affiliation(s)
- Anantharaman Shantaraman
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B Dammer
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Obiadada Ugochukwu
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc M Duong
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Luming Yin
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - E Kathleen Carter
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Marla Gearing
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - James J Lah
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas T Seyfried
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Lenora Higginbotham
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
4
|
Ding Z, Zhang R, Zhu W, Lu Y, Zhu Z, Xie H, Tang W. CTHRC1 serves as an indicator in biliary atresia for evaluating the stage of liver fibrosis and predicting prognosis. Dig Liver Dis 2024:S1590-8658(24)00869-7. [PMID: 39043537 DOI: 10.1016/j.dld.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Liver fibrosis is a pathological feature of biliary atresia (BA). However, both histological fibrosis stage and existing biomarkers fail to predict prognosis at the time of hepatoportonterostomy (HPE). AIMS To explore the role of collagen triple- helix repeat containing-1 (CTHRC1) in BA. METHODS CTHRC1 expression levels were detected and its association with liver fibrosis stage was analyzed in patients with BA. Immunohistochemistry and immunofluorescent analyses were performed to detect the expression and localization of CTHRC1. Epithelial-mesenchymal transition (EMT) and proliferation were analyzed in cholangiocytes treated with recombinant human CTHRC1 protein. Survival analyses were performed to assess the prognostic value of CTHRC1 in patients with BA. RESULTS CTHRC1 was upregulated in BA, and its expression level was positively correlated with fibrosis-related markers and the severity of liver fibrosis. In liver tissue CTHRC1 was co-localized with CK19 and highly expressed in patients with severe liver fibrosis. Further experiments revealed that CTHRC1 promoted cholangiocyte EMT and proliferation. Additionally, CTHRC1 expression levels at HPE could predict the 2-year native liver survival (NLS). CONCLUSIONS CTHRC1 promotes the EMT and proliferation of cholangiocytes and indicate the stage of liver fibrosis. The CTHRC1 expression levels can predict outcomes of BA.
Collapse
Affiliation(s)
- Zequan Ding
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210000, Jiangsu Province, China
| | - Ruyi Zhang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210000, Jiangsu Province, China
| | - Wei Zhu
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210000, Jiangsu Province, China
| | - Yao Lu
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210000, Jiangsu Province, China
| | - Zhongxian Zhu
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210000, Jiangsu Province, China
| | - Hua Xie
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210000, Jiangsu Province, China.
| | - Weibing Tang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210000, Jiangsu Province, China.
| |
Collapse
|
5
|
Potus F, Bonnet S. Collagen Triple Helix Repeat-Containing Protein 1: More Than Just a Novel Biomarker for Right Ventricular Involvement in Pulmonary Hypertension. Can J Cardiol 2024:S0828-282X(24)00514-2. [PMID: 38960126 DOI: 10.1016/j.cjca.2024.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024] Open
Affiliation(s)
- Francois Potus
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, Québec, Canada.
| | - Sebastien Bonnet
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, Québec, Canada
| |
Collapse
|
6
|
Mukhatayev Z, Adilbayeva A, Kunz J. CTHRC1: An Emerging Hallmark of Pathogenic Fibroblasts in Lung Fibrosis. Cells 2024; 13:946. [PMID: 38891078 PMCID: PMC11171484 DOI: 10.3390/cells13110946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Pulmonary fibrosis is a chronic, progressive, irreversible lung disease characterized by fibrotic scarring in the lung parenchyma. This condition involves the excessive accumulation of extracellular matrix (ECM) due to the aberrant activation of myofibroblasts in the alveolar environment. Transforming growth factor beta (TGF-β) signaling is a crucial driver of fibrogenesis because it promotes excessive ECM deposition, thereby leading to scar formation and lung damage. A primary target of TGF-β signaling in fibrosis is Collagen Triple Helix Repeat Containing 1 (CTHRC1), a secreted glycoprotein that plays a pivotal role in ECM deposition and wound repair. TGF-β transcriptionally regulates CTHRC1 in response to tissue injury and controls the wound healing response through functional activity. CTHRC1 may also play an essential role in re-establishing and maintaining tissue homeostasis after wound closure by modulating both the TGF-β and canonical Wnt signaling pathways. This dual function suggests that CTHRC1 regulates tissue remodeling and homeostasis. However, deregulated CTHRC1 expression in pathogenic fibroblasts has recently emerged as a hallmark of fibrosis in multiple organs and tissues. This review highlights recent studies suggesting that CTHRC1 can serve as a diagnostic and prognostic biomarker for fibrosis in idiopathic pulmonary fibrosis, systemic sclerosis, and post-COVID-19 lung fibrosis. Notably, CTHRC1 expression is responsive to antifibrotic drugs that target the TGF-β pathway, such as pirfenidone and bexotegrast, indicating its potential as a biomarker of treatment success. These findings suggest that CTHRC1 may present new opportunities for diagnosing and treating patients with lung fibrosis.
Collapse
Affiliation(s)
| | | | - Jeannette Kunz
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, 5/1 Kerey and Zhanibek Khans St., 020000 Astana, Kazakhstan; (Z.M.); (A.A.)
| |
Collapse
|
7
|
Yang J, Xiao L, Zhang L, Luo G, Ma Y, Wang X, Zhang Y. Platelets: A Potential Factor that Offers Strategies for Promoting Bone Regeneration. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 38482796 DOI: 10.1089/ten.teb.2024.0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Bone defects represent a prevalent category of clinical injuries, causing significant pain and escalating health care burdens. Effectively addressing bone defects is thus of paramount importance. Platelets, formed from megakaryocyte lysis, have emerged as pivotal players in bone tissue repair, inflammatory responses, and angiogenesis. Their intracellular storage of various growth factors, cytokines, and membrane protein receptors contributes to these crucial functions. This article provides a comprehensive overview of platelets' roles in hematoma structure, inflammatory responses, and angiogenesis throughout the process of fracture healing. Beyond their application in conjunction with artificial bone substitute materials for treating bone defects, we propose the potential future use of anticoagulants such as heparin in combination with these materials to regulate platelet number and function, thereby promoting bone healing. Ultimately, we contemplate whether manipulating platelet function to modulate bone healing could offer innovative ideas and directions for the clinical treatment of bone defects.
Collapse
Affiliation(s)
- Jingjing Yang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, China
| | - Lan Xiao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- School of Medicine and Dentistry, Griffith University, Queensland, Australia
| | - Lijia Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| | - Guochen Luo
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology in Colleges and Universities, Zunyi Medical University, Zunyi, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi Medical University, Zunyi, China
| |
Collapse
|
8
|
Sağraç D, Aydın S, Kırbaş OK, Öztürkoğlu D, Şahin F. Extracellular vesicles derived from human foreskin cells (hFS-Exo) accelerate cell migration and angiogenesis through MAPK pathway: an in vitro study. Mol Biol Rep 2024; 51:471. [PMID: 38551706 DOI: 10.1007/s11033-024-09378-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/26/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Wound healing is one of the important processes in the body. Attempts to create new drugs are of interest due to the side effects of natural and chemical wound healing compounds. To overcome this obstacle, stem cells have been used as healing agents. However, both difficulties in collection and risks such as rejection and teratoma in the recipient body have limited the use of stem cells, directly. Since the potential content of the stem cells can be transferred to the recipient cells by vesicles, small extracellular vesicles have recently become prominent agents. METHODS AND RESULTS The wound-healing effect of extracellular vesicles derived from foreskin cells was investigated in both keratinocyte and endothelial cells. Migration assay, RT-PCR, Col1a1 ELISA and Western Blot experiments were utilized to reveal healing effect of EVs and its possible molecular pathways. EV-treated groups exhibited more proliferative, invasive, and migrative characteristics. When comparing to the control group, new vessel formation was induced in EV groups. An increase in gene levels of growth factors related to wound healing and change in the mitogen-activated protein kinase (MAPK) signaling pathway proteins in EV-treated groups were determined. Possible molecular mechanisms underlying cell movements were associated with the MAPK pathway. It was found that human foreskin cell EVs (hFS-Exo) may have a potential to heal wounds in a short period of time by triggering the MAPK pathway. CONCLUSIONS hFS-Exo could be a new promising wound healing agent in the future.
Collapse
Affiliation(s)
- Derya Sağraç
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Safa Aydın
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Oğuz Kaan Kırbaş
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Dilek Öztürkoğlu
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Turkey.
| |
Collapse
|
9
|
He Y, Cen Y, Tian M. Immunomodulatory hydrogels for skin wound healing: cellular targets and design strategy. J Mater Chem B 2024; 12:2435-2458. [PMID: 38284157 DOI: 10.1039/d3tb02626d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Skin wounds significantly impact the global health care system and represent a significant burden on the economy and society due to their complicated dynamic healing processes, wherein a series of immune events are required to coordinate normal and sequential healing phases, involving multiple immunoregulatory cells such as neutrophils, macrophages, keratinocytes, and fibroblasts, since dysfunction of these cells may impede skin wound healing presenting persisting inflammation, impaired vascularization, and excessive collagen deposition. Therefore, cellular target-based immunomodulation is promising to promote wound healing as cells are the smallest unit of life in immune response. Recently, immunomodulatory hydrogels have become an attractive avenue to promote skin wound healing. However, a detailed and comprehensive review of cellular targets and related hydrogel design strategies remains lacking. In this review, the roles of the main immunoregulatory cells participating in skin wound healing are first discussed, and then we highlight the cellular targets and state-of-the-art design strategies for immunomodulatory hydrogels based on immunoregulatory cells that cover defect, infected, diabetic, burn and tumor wounds and related scar healing. Finally, we discuss the barriers that need to be addressed and future prospects to boost the development and prosperity of immunomodulatory hydrogels.
Collapse
Affiliation(s)
- Yinhai He
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Cen
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meng Tian
- Department of Neurosurgery and Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Wei R, Song J, Liu X, Huo S, Liu C, Liu X. Immunosuppressive MFAP2 + cancer associated fibroblasts conferred unfavorable prognosis and therapeutic resistance in gastric cancer. Cell Oncol (Dordr) 2024; 47:55-68. [PMID: 37540308 DOI: 10.1007/s13402-023-00849-y] [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] [Accepted: 07/24/2023] [Indexed: 08/05/2023] Open
Abstract
PURPOSE To explore the predictive merit of MFAP2+ cancer associated fibroblasts (CAFs) infiltration for clinical outcomes and adjuvant chemotherapy or immunotherapy responsiveness in gastric cancer (GC). METHODS In this study, several independent cohorts were included respectively to dissect the relationship of clinical outcomes, therapeutic responses and tumor microenvironment with different MFAP2+ CAFs infiltration. Drug sensitivity analysis was conducted to predict the relationship between MFAP2+ CAFs infiltration and targeted drug response. Kaplan-Meier curves and the log-rank test were used to compare clinical outcomes of patients with different MFAP2+ CAFs infiltration. RESULTS High MFAP2+ CAFs infiltration yielded inferior prognosis in terms of overall survival, progress free survival and recurrence free survival in GC. Patients with low MFAP2+ CAFs infiltration were more likely to gain benefit from adjuvant therapy. Moreover, low MFAP2+ CAFs infiltration could predict a promising response to immunotherapy in GC patients. MFAP2+ CAFs with immunosuppressive features were highly relevant to immune evasive contexture characterized by the dysfunction of CD8+ T cells. We found that MFAP2+ CAFs communicated with T cells, B cells and Macrophages through releasing macrophage migration inhibitor factor (MIF), which further suggested that MFAP2+ CAFs might promote therapeutic resistance through regulating T cells dysfunction and M2 macrophages polarization. CONCLUSION Immunosuppressive MFAP2+ CAFs constructed an immune evasive tumor microenvironment characterized by incapacitated immune effector cells, consequently predicting inferior clinical outcomes and response on adjuvant therapy and immunotherapy in patients with GC. The potential of immunosuppressive MFAP2+ CAFs as a therapeutic target for GC deserved thoroughly exploration.
Collapse
Affiliation(s)
- Rongyuan Wei
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Junquan Song
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xuanjun Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Shiying Huo
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Chenchen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
| | - Xiaowen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Shantaraman A, Dammer EB, Ugochukwu O, Duong DM, Yin L, Carter EK, Gearing M, Chen-Plotkin A, Lee EB, Trojanowski JQ, Bennett DA, Lah JJ, Levey AI, Seyfried NT, Higginbotham L. Network Proteomics of the Lewy Body Dementia Brain Reveals Presynaptic Signatures Distinct from Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.23.576728. [PMID: 38328211 PMCID: PMC10849701 DOI: 10.1101/2024.01.23.576728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Lewy body dementia (LBD), a class of disorders comprising Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB), features substantial clinical and pathological overlap with Alzheimer's disease (AD). The identification of biomarkers unique to LBD pathophysiology could meaningfully advance its diagnosis, monitoring, and treatment. Using quantitative mass spectrometry (MS), we measured over 9,000 proteins across 138 dorsolateral prefrontal cortex (DLPFC) tissues from a University of Pennsylvania autopsy collection comprising control, Parkinson's disease (PD), PDD, and DLB diagnoses. We then analyzed co-expression network protein alterations in those with LBD, validated these disease signatures in two independent LBD datasets, and compared these findings to those observed in network analyses of AD cases. The LBD network revealed numerous groups or "modules" of co-expressed proteins significantly altered in PDD and DLB, representing synaptic, metabolic, and inflammatory pathophysiology. A comparison of validated LBD signatures to those of AD identified distinct differences between the two diseases. Notably, synuclein-associated presynaptic modules were elevated in LBD but decreased in AD relative to controls. We also found that glial-associated matrisome signatures consistently elevated in AD were more variably altered in LBD, ultimately stratifying those LBD cases with low versus high burdens of concurrent beta-amyloid deposition. In conclusion, unbiased network proteomic analysis revealed diverse pathophysiological changes in the LBD frontal cortex distinct from alterations in AD. These results highlight the LBD brain network proteome as a promising source of biomarkers that could enhance clinical recognition and management.
Collapse
Affiliation(s)
- Anantharaman Shantaraman
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B. Dammer
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Obiadada Ugochukwu
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc M. Duong
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Luming Yin
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - E. Kathleen Carter
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Marla Gearing
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B. Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - James J. Lah
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I. Levey
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas T. Seyfried
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lenora Higginbotham
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
12
|
Lv X, He Z, Yang M, Wang L, Fu S. Analysis of subsets and localization of macrophages in skin lesions and peripheral blood of patients with keloids. Heliyon 2024; 10:e24034. [PMID: 38283250 PMCID: PMC10818209 DOI: 10.1016/j.heliyon.2024.e24034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Keloids are a type of abnormal fibrous proliferation disease of the skin, characterized by local inflammation that lacks clear pathogenesis and satisfactory treatment. The phenomenon of distinct phenotypes, including M1 and M2 macrophages, is called macrophage polarization. Recently, macrophage polarization has been suggested to play a role in keloid formation. This study aimed to evaluate the relation between macrophage polarization and keloids and identify novel effective treatments for keloids. Differentially expressed genes were identified via RNA sequencing of the skin tissue of healthy controls and patients with keloids and validated using quantitative PCR. Multiplex immunofluorescence microscopy was used to detect different phenotypes of macrophages in keloid tissues. Finally, quantitative PCR validation of differentially expressed genes and flow cytometry were used to analyze macrophages in the peripheral blood of healthy controls and patients with keloids. Total and M2 macrophages were significantly increased in the local skin tissue and peripheral blood of patients with keloids compared with healthy controls. In addition, inflammation- and macrophage polarization-related differentially expressed genes in keloid tissue showed similar expression patterns in the peripheral blood. This study highlighted an increased frequency of total macrophages and M2 polarization in the local skin tissue and peripheral blood of patients with keloids. This systematic macrophage polarization tendency also indicates a potential genetic predisposition to keloids. These findings suggest the possibility of developing new diagnostic and therapeutic indicators for keloids focusing on macrophages.
Collapse
Affiliation(s)
- Xinyi Lv
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Zhenghao He
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan 410011, China
- Department of Plastic Surgery, Zhongshan City People's Hospital, Zhongshan, Guangdong, China
| | - Ming Yang
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Lu Wang
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Siqi Fu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| |
Collapse
|
13
|
Liu Y, Chen X, Xu Y, Yang T, Wang H, Wang Z, Hu Z, Chen L, Zhang Z, Wu Y. CTHRC1 promotes colorectal cancer progression by recruiting tumor-associated macrophages via up-regulation of CCL15. J Mol Med (Berl) 2024; 102:81-94. [PMID: 37987774 DOI: 10.1007/s00109-023-02399-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
Tumor-associated macrophages (TAMs) represent a key factor in the tumor immune microenvironment (TME), exerting significant influence over tumor migration, invasion, immunosuppressive features, and drug resistance. Collagen triple helix repeat containing 1 (CTHRC1), a 30 KDa protein which was secreted during the tissue-repair process, is highly expressed in several malignant tumors, including colorectal cancer (CRC). Previous studies demonstrated that CTHRC1 expression in TAMs was positively correlated to M2 macrophage polarization and liver metastasis, while our discovery suggesting a novel mechanism that CTHRC1 secreted from cancer cell could indirectly interplay with TAMs. In this study, the high expression level of CTHRC1 was evaluated in CRC based on GEO and TCGA databases. Further, CTHRC1 was detected high in all stages of CRC patients by ELISA and was correlated to poor prognosis. Multispectral imaging of IHC demonstrated that M2 macrophage infiltration was increased accompanied with CTHRC1 enrichment, suggesting that CTHRC1 may have chemotactic effect on macrophages. In vitro, CTHRC1 could have chemotactic ability of macrophage in the presence of HT-29 cell line. Cytokine microarray revealed that CTHRC1 could up-regulate the CCL15 level of HT-29, pathway analysis demonstrated that CTHRC1 could regulate CCL15 by controlling the TGFβ activation and Smad phosphorylation level. In vivo, knocking down of CTHRC1 from CT-26 also inhibits tumor formation. In conclusion, CTHRC1 could promote the chemotactic ability of macrophages by up-regulating CCL15 via TGFβ/Smad pathway; additionally, a high level of CTHRC1 could promote macrophage's M2 polarization. This discovery may be related to tumor immune tolerance and tumor immunotherapy resistance in CRC. KEY MESSAGES: CTHRC1 promotes CRC progression by up-regulating CCL15 via TGF-β/Smad pathways to further recruit tumor-associated macrophages. By the means of autocrine or paracrine, CTHRC1 can indeed promote macrophage chemotaxis and enhance the infiltration of macrophages in tumor tissues but in the presence of tumor cells. CAFs were another source of CTHRC1, indicating CTHRC1 can infiltrate tumor islet as well as the stomal and be secreted from both tumor cells and CAFs. This study validated CTHRC1 as a potential immune therapy target CRC.
Collapse
Affiliation(s)
- Yixin Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangzheng Chen
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Ying Xu
- Targeted Tracer Research and Development Laboratory, Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tinghan Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Haichuan Wang
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Ziqiang Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhangyong Hu
- Department of Infectious Diseases, the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zheng Zhang
- Department of Liver Surgery & Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yangping Wu
- Targeted Tracer Research and Development Laboratory, Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
14
|
Bonnici L, Suleiman S, Schembri-Wismayer P, Cassar A. Targeting Signalling Pathways in Chronic Wound Healing. Int J Mol Sci 2023; 25:50. [PMID: 38203220 PMCID: PMC10779022 DOI: 10.3390/ijms25010050] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Chronic wounds fail to achieve complete closure and are an economic burden to healthcare systems due to the limited treatment options and constant medical attention. Chronic wounds are characterised by dysregulated signalling pathways. Research has focused on naturally derived compounds, stem-cell-based therapy, small molecule drugs, oligonucleotide delivery nanoparticles, exosomes and peptide-based platforms. The phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT), Wingless-related integration (Wnt)/β-catenin, transforming growth factor-β (TGF-β), nuclear factor erythroid 2-related factor 2 (Nrf2), Notch and hypoxia-inducible factor 1 (HIF-1) signalling pathways have critical roles in wound healing by modulating the inflammatory, proliferative and remodelling phases. Moreover, several regulators of the signalling pathways were demonstrated to be potential treatment targets. In this review, the current research on targeting signalling pathways under chronic wound conditions will be discussed together with implications for future studies.
Collapse
Affiliation(s)
| | | | | | - Analisse Cassar
- Department of Anatomy, University of Malta, MSD 2080 Msida, Malta; (L.B.); (S.S.); (P.S.-W.)
| |
Collapse
|
15
|
Liu YJ, Du J, Li J, Tan XP, Zhang Q. CTHRC1, a novel gene with multiple functions in physiology, disease and solid tumors (Review). Oncol Lett 2023; 25:266. [PMID: 37216164 PMCID: PMC10193374 DOI: 10.3892/ol.2023.13852] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 03/01/2023] [Indexed: 05/24/2023] Open
Abstract
Collagen triple helix repeat containing 1 (CTHRC1) is a gene discovered in 2005; it is highly conserved, and no homologous proteins have been disclosed thus far. A number of studies have shown that CTHRC1 is present in normal tissues and organs, and it has vital functions in physiological processes, including participating in the regulation of metabolism, arterial remodeling, bone formation and myelination of the peripheral nervous system. It has been reported that abnormal expression of CTHRC1 is involved in the carcinogenesis of various human organs, such as the breast, colon, pancreas, lung, stomach and liver. Therefore, the present review aims to collate all known findings and results on the regulation of CTHRC1 expression and related signaling pathways. To conclude, this review also provides a hypothesis of the functional mechanism of this gene.
Collapse
Affiliation(s)
- Ya-Juan Liu
- Department of Gastroenterology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
- Medical College of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jing Du
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Jie Li
- Department of Gastroenterology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Xiao-Ping Tan
- Department of Gastroenterology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Qing Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
- Medical College of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
16
|
Baiken Y, Markhametova Z, Ashimova A, Zhulamanova A, Nogaibayeva A, Kozina L, Matkarimov B, Aituov B, Gaipov A, Myngbay A. Elevated Levels of Plasma Collagen Triple Helix Repeat Containing 1 (CTHRC1) Is Strongly Associated with eGFR and Albuminuria in Chronic Kidney Disease. Medicina (B Aires) 2023; 59:medicina59040651. [PMID: 37109608 PMCID: PMC10146339 DOI: 10.3390/medicina59040651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Background: Chronic kidney disease (CKD) has various etiologies, making it impossible to fully understand its complex pathophysiology. Elevated levels of plasma creatinine, proteinuria, and albuminuria and declined eGFR are traits observed in CKD patients. The current study attempts to highlight the collagen triple helix repeat containing 1 (CTHRC1) protein as a putative blood biomarker for CKD in addition to existing recognized indicators of CKD progression. Methods: A total of 26 CKD patients and 18 healthy controls were enrolled in this study. Clinical characteristics and complete blood and biochemical analyses were collected, and human ELISA kits were used to detect possible CKD biomarkers. Results: The study’s findings showed that CTHRC1 correlates with key clinical markers of kidney function such as 24 h urine total protein, creatinine, urea, and uric acid. In addition, CTHRC1 demonstrated a strong significant difference (p ≤ 0.0001) between the CKD and control group. Conclusions: Our research demonstrates that the plasma level of CTHRC1 can distinguish between those with CKD and healthy patients. Plasma CTHRC1 levels may aid in the diagnosis of CKD given the current state of knowledge, and these results call for further investigation in a wider, more diverse patient group.
Collapse
|
17
|
Cai S, Zhao M, Zhou B, Yoshii A, Bugg D, Villet O, Sahu A, Olson GS, Davis J, Tian R. Mitochondrial dysfunction in macrophages promotes inflammation and suppresses repair after myocardial infarction. J Clin Invest 2023; 133:e159498. [PMID: 36480284 PMCID: PMC9927948 DOI: 10.1172/jci159498] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
Innate immune cells play important roles in tissue injury and repair following acute myocardial infarction (MI). Although reprogramming of macrophage metabolism has been observed during inflammation and resolution phases, the mechanistic link to macrophage phenotype is not fully understood. In this study, we found that myeloid-specific deletion (mKO) of mitochondrial complex I protein, encoded by Ndufs4, reproduced the proinflammatory metabolic profile in macrophages and exaggerated the response to LPS. Moreover, mKO mice showed increased mortality, poor scar formation, and worsened cardiac function 30 days after MI. We observed a greater inflammatory response in mKO mice on day 1 followed by increased cell death of infiltrating macrophages and blunted transition to the reparative phase during post-MI days 3-7. Efferocytosis was impaired in mKO macrophages, leading to lower expression of antiinflammatory cytokines and tissue repair factors, which suppressed the proliferation and activation of myofibroblasts in the infarcted area. Mitochondria-targeted ROS scavenging rescued these impairments, improved myofibroblast function in vivo, and reduced post-MI mortality in mKO mice. Together these results reveal a critical role of mitochondria in inflammation resolution and tissue repair via modulation of efferocytosis and crosstalk with fibroblasts. These findings have potential significance for post-MI recovery as well as for other inflammatory conditions.
Collapse
Affiliation(s)
- Shanshan Cai
- Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and
| | - Mingyue Zhao
- Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and
| | - Bo Zhou
- Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and
| | - Akira Yoshii
- Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and
| | - Darrian Bugg
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Outi Villet
- Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and
| | - Anita Sahu
- Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and
| | - Gregory S. Olson
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Medical Scientist Training Program, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jennifer Davis
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Rong Tian
- Center for Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine and
| |
Collapse
|
18
|
Bohn B, Lutsey PL, Tang W, Pankow JS, Norby FL, Yu B, Ballantyne CM, Whitsel EA, Matsushita K, Demmer RT. A proteomic approach for investigating the pleiotropic effects of statins in the atherosclerosis risk in communities (ARIC) study. J Proteomics 2023; 272:104788. [PMID: 36470581 PMCID: PMC9819193 DOI: 10.1016/j.jprot.2022.104788] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Statins are prescribed to reduce LDL-c and risk of CVD. Statins have pleiotropic effects, affecting pathophysiological functions beyond LDL-c reduction. We compared the proteome of statin users and nonusers (controls). We hypothesized that statin use is associated with proteins unrelated to lipid metabolism. METHODS Among 10,902 participants attending ARIC visit 3 (1993-95), plasma concentrations of 4955 proteins were determined using SOMAlogic's DNA aptamer-based capture array. 379 participants initiated statins within the 2 years prior. Propensity scores (PS) were calculated based on visit 2 (1990-92) LDL-c levels and visit 3 demographic/clinical characteristics. 360 statin users were PS matched to controls. Log2-transformed and standardized protein levels were compared using t-tests, with false discovery rate (FDR) adjustment for multiple comparisons. Analyses were replicated in visit 2. RESULTS Covariates were balanced after PS matching, except for higher visit 3 LDL-c levels among controls (125.70 vs 147.65 mg/dL; p < 0.0001). Statin users had 11 enriched and 11 depleted protein levels after FDR adjustment (q < 0.05). Proteins related and unrelated to lipid metabolism differed between groups. Results were largely replicated in visit 2. CONCLUSION Proteins unrelated to lipid metabolism differed by statin use. Pending external validation, exploring their biological functions could elucidate pleiotropic effects of statins. SIGNIFICANCE Statins are the primary pharmacotherapy for lowering low-density lipoprotein (LDL) cholesterol and preventing cardiovascular disease. Their primary mechanism of action is through inhibiting the protein 3hydroxy-3-methylglutaryl CoA reductase (HMGCR) in the mevalonate pathway of LDL cholesterol synthesis. However, statins have pleiotropic effects and may affect other biological processes directly or indirectly, with hypothesized negative and positive effects. The present study contributes to identifying these pathways by comparing the proteome of stain users and nonusers with propensity score matching. Our findings highlight potential biological mechanisms underlying statin pleiotropy, informing future efforts to identify statin users at risk of rare nonatherosclerotic outcomes and identify health benefits of statin use independent of LDL-C reduction.
Collapse
Affiliation(s)
- Bruno Bohn
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, United States of America
| | - Pamela L Lutsey
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, United States of America
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, United States of America
| | - James S Pankow
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, United States of America
| | - Faye L Norby
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, CA, United States of America
| | - Bing Yu
- Baylor College of Medicine, United States of America
| | | | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Public Health and Department of Medicine, University of North Carolina - Chapel Hill, NC, United States of America
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, United States of America
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, United States of America.
| |
Collapse
|
19
|
Gharbia FZ, Abouhashem AS, Moqidem YA, Elbaz AA, Abdellatif A, Singh K, Sen CK, Azzazy HME. Adult skin fibroblast state change in murine wound healing. Sci Rep 2023; 13:886. [PMID: 36650180 PMCID: PMC9845335 DOI: 10.1038/s41598-022-27152-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/27/2022] [Indexed: 01/18/2023] Open
Abstract
Wound healing is a well-organized dynamic process involving coordinated consecutive phases: homeostasis, inflammation, proliferation and resolution. Fibroblasts play major roles in skin wound healing such as in wound contraction and release of growth factors which are of importance in angiogenesis and tissue remodeling. Abnormal fibroblast phenotypes have been identified in patients with chronic wounds. In this work, we analyzed scRNA-seq datasets of normal and wounded skin from mice at day 4 post-wound to investigate fibroblast heterogeneity during the proliferative phase of wound healing. Compositional analysis revealed a specific subset of fibroblast (cluster 3) that primarily increased in wounded skin (14%) compared to normal skin (3.9%). This subset was characterized by a gene signature marked by the plasma membrane proteins Sfrp2 + Sfrp4 + Sfrp1 + and the transcription factors Ebf1 + Prrx1 + Maged1 + . Differential gene expression and enrichment analysis identified epithelial to mesenchymal transition (EMT) and angiogenesis to be upregulated in the emerging subset of fibroblasts of the wounded skin. Using two other datasets for murine wounded skin confirmed the increase in cluster 3-like fibroblasts at days 2, 7 and 14 post-wounding with a peak at day 7. By performing a similarity check between the differential gene expression profile between wounded and normal skin for this emerging fibroblast subset with drug signature from the ConnectivityMap database, we identified drugs capable of mimicking the observed gene expression change in fibroblasts during wound healing. TTNPB, verteprofin and nicotinic acid were identified as candidate drugs capable of inducing fibroblast gene expression profile necessary for wound healing. On the other hand, methocarbamol, ifosfamide and penbutolol were recognized to antagonize the identified fibroblast differential expression profile during wound healing which might cause delay in wound healing. Taken together, analysis of murine transcriptomic skin wound healing datasets suggested a subset of fibroblasts capable of inducing EMT and further inferred drugs that might be tested as potential candidates to induce wound closure.
Collapse
Affiliation(s)
- Fatma Z Gharbia
- Graduate Nanotechnology Program, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Ahmed S Abouhashem
- Indiana Center for Regenerative Medicine & Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
- Sharkia Clinical Research Department, Ministry of Health & Population, Zagazig, 44511, Sharkia, Egypt
- CytoTalk LLC, Cheyenne, WY, 82001, USA
| | - Yomna A Moqidem
- Department of Biology, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
| | - Ahmed A Elbaz
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
- CytoTalk LLC, Cheyenne, WY, 82001, USA
| | - Ahmed Abdellatif
- Department of Biology, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
| | - Kanhaiya Singh
- Indiana Center for Regenerative Medicine & Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Chandan K Sen
- Indiana Center for Regenerative Medicine & Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Hassan M E Azzazy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt.
- Department of Nanobiophotonics, Leibniz Institute for Photonic Technology, Albert Einstein Str. 9, 07745, Jena, Germany.
| |
Collapse
|
20
|
Li Y, Wang H, Zhao Z, Yang Y, Meng Z, Qin L. Effects of the interactions between platelets with other cells in tumor growth and progression. Front Immunol 2023; 14:1165989. [PMID: 37153586 PMCID: PMC10158495 DOI: 10.3389/fimmu.2023.1165989] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/31/2023] [Indexed: 05/09/2023] Open
Abstract
It has been confirmed that platelets play a key role in tumorigenesis. Tumor-activated platelets can recruit blood cells and immune cells to migrate, establish an inflammatory tumor microenvironment at the sites of primary and metastatic tumors. On the other hand, they can also promote the differentiation of mesenchymal cells, which can accelerate the proliferation, genesis and migration of blood vessels. The role of platelets in tumors has been well studied. However, a growing number of studies suggest that interactions between platelets and immune cells (e.g., dendritic cells, natural killer cells, monocytes, and red blood cells) also play an important role in tumorigenesis and tumor development. In this review, we summarize the major cells that are closely associated with platelets and discuss the essential role of the interaction between platelets with these cells in tumorigenesis and tumor development.
Collapse
|
21
|
Chen Q, Gu Y, Tan C, Sundararajan B, Li Z, Wang D, Zhou Z. Comparative effects of five polymethoxyflavones purified from Citrus tangerina on inflammation and cancer. Front Nutr 2022; 9:963662. [PMID: 36159482 PMCID: PMC9493082 DOI: 10.3389/fnut.2022.963662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/11/2022] [Indexed: 12/24/2022] Open
Abstract
Although the Citrus tangerina cultivar “Dahongpao” (CTD) has been established as a rich source of polymethoxyflavones (PMFs) with anti-inflammatory and anti-cancer properties, their individual effects on cellular signaling remain to be elucidated. In this study, five major PMFs from the peel of CTD were isolated, including sinensetin, tetramethyl-O-scutellarin (5,6,7,4′-tetramethoxyflavone), nobiletin (5,6,7,8,3′, 4′-hexamethoxyflavone), tangeretin (5,6,7,8,4′-pentamethoxyflavone), and 5-demethylnobiletin (5-OH-6,7,8,3′,4′-pentamethoxyflavone). These PMFs were found to significantly (p < 0.05) inhibit the production of NO and biomarkers of chronic inflammation (TNF-α and IL-6). Additionally, they effectively suppressed mRNA biomarkers of acute inflammation (Cox-2 and iNOS), and to varying degrees promoted the activation of anti-inflammatory cytokines (IL-4, IL-13, TNF-β, and IL-10). Among the five PMFs, tangeretin was found to have a considerable anti-proliferative effect on tumor cell lines (PC-3 and DU145) and synergistically enhanced the cytotoxicity of mitoxantrone, partially via activation of the PTEN/AKT pathway. The findings of this study provide valuable insights into the activity of different PMF monomers and advance the understanding of the roles of PMFs in promoting apoptotic and anti-cancer effects.
Collapse
Affiliation(s)
- Qiyang Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Yue Gu
- Key Laboratory of Horticulture Science for Southern Mountainous Regions of Ministry of Education, College of Horticulture and Landscape Architecture, Southwest University, Chongqing, China
| | - Chun Tan
- Key Laboratory of Horticulture Science for Southern Mountainous Regions of Ministry of Education, College of Horticulture and Landscape Architecture, Southwest University, Chongqing, China
| | - Balasubramani Sundararajan
- Key Laboratory of Horticulture Science for Southern Mountainous Regions of Ministry of Education, College of Horticulture and Landscape Architecture, Southwest University, Chongqing, China
| | - Zhenqing Li
- Key Laboratory of Horticulture Science for Southern Mountainous Regions of Ministry of Education, College of Horticulture and Landscape Architecture, Southwest University, Chongqing, China
| | - Dan Wang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, China
- *Correspondence: Dan Wang
| | - Zhiqin Zhou
- Key Laboratory of Horticulture Science for Southern Mountainous Regions of Ministry of Education, College of Horticulture and Landscape Architecture, Southwest University, Chongqing, China
- The Southwest Institute of Fruits Nutrition, Chongqing, China
- Zhiqin Zhou
| |
Collapse
|
22
|
Tong KP, Intine R, Wu S. Vitamin C and the management of diabetic foot ulcers: a literature review. J Wound Care 2022; 31:S33-S44. [PMID: 36113854 DOI: 10.12968/jowc.2022.31.sup9.s33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The lifetime risk of developing a diabetic foot ulcer (DFU) in people with diabetes is as high as 25%. A trio of factors constitute the diabetic foot syndrome that characterises DFUs, including neuropathy, vascular disease and infections. Vitamin C has important functions in the nervous, cardiovascular, and immune systems that are implicated in DFU development. Furthermore, vitamin C deficiency has been observed in individuals with DFUs, suggesting an important function of vitamin C in DFU management and treatment. Therefore, this literature review evaluates the role of vitamin C in the nervous, cardiovascular and immune systems in relation to wound healing and DFUs, as well as discussing vitamin C's lesser known role in depression, a condition that affects many individuals with a DFU. METHOD A literature search was done using PubMed, Cochrane Library, Embase, Ovid, Computer Retrieval of Information on Scientific Projects, and NIH Clinical Center. Search terms included 'diabetic foot ulcer,' 'diabetic foot,' 'vitamin C,' and 'ascorbic acid.' RESULTS Of the 71 studies initially identified, seven studies met the inclusion criteria, and only three were human clinical trials. Overall, the literature on this subject is limited, with mainly observational and animal studies, and few human clinical trials. CONCLUSION There is a need for additional human clinical trials on vitamin C supplementation in individuals with a DFU to fill the knowledge gap and guide clinical practice.
Collapse
Affiliation(s)
- Khanh Phuong Tong
- Dr William M Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, Illinois, US
| | - Robert Intine
- School of Graduate and Postdoctoral Studies, College of Health Professions, Rosalind Franklin University of Medicine and Science, Illinois, US
| | - Stephanie Wu
- Dr William M Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, Illinois, US
| |
Collapse
|
23
|
Duan X, Yuan X, Yao B, Song W, Li Z, Enhejirigala, Kong Y, Wang Y, Fu X, Huang S. The role of CTHRC1 in promotion of cutaneous wound healing. Signal Transduct Target Ther 2022; 7:183. [PMID: 35701414 PMCID: PMC9197944 DOI: 10.1038/s41392-022-01008-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 04/07/2022] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Xianlan Duan
- School of Medicine, NanKai University, 94 Wei Jin Road, Tianjin, 300071, People's Republic of China.,Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital, 51 Fu Cheng Road, Beijing, 100048, People's Republic of China
| | - Xingyu Yuan
- School of Medicine, NanKai University, 94 Wei Jin Road, Tianjin, 300071, People's Republic of China.,Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital, 51 Fu Cheng Road, Beijing, 100048, People's Republic of China
| | - Bin Yao
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital, 51 Fu Cheng Road, Beijing, 100048, People's Republic of China.,Academy of Medical Engineering and Translational Medicine, Tianjin University, 92 Weijin Road, Tianjin, 300072, People's Republic of China
| | - Wei Song
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital, 51 Fu Cheng Road, Beijing, 100048, People's Republic of China
| | - Zhao Li
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital, 51 Fu Cheng Road, Beijing, 100048, People's Republic of China
| | - Enhejirigala
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital, 51 Fu Cheng Road, Beijing, 100048, People's Republic of China
| | - Yi Kong
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital, 51 Fu Cheng Road, Beijing, 100048, People's Republic of China
| | - Yuzhen Wang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital, 51 Fu Cheng Road, Beijing, 100048, People's Republic of China
| | - Xiaobing Fu
- School of Medicine, NanKai University, 94 Wei Jin Road, Tianjin, 300071, People's Republic of China. .,Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital, 51 Fu Cheng Road, Beijing, 100048, People's Republic of China.
| | - Sha Huang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital, 51 Fu Cheng Road, Beijing, 100048, People's Republic of China.
| |
Collapse
|
24
|
Ye Z, Wei J, Zhan C, Hou J. Role of Transforming Growth Factor Beta in Peripheral Nerve Regeneration: Cellular and Molecular Mechanisms. Front Neurosci 2022; 16:917587. [PMID: 35769702 PMCID: PMC9234557 DOI: 10.3389/fnins.2022.917587] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/11/2022] [Indexed: 11/24/2022] Open
Abstract
Peripheral nerve injury (PNI) is one of the most common concerns in trauma patients. Despite significant advances in repair surgeries, the outcome can still be unsatisfactory, resulting in morbidities such as loss of sensory or motor function and reduced quality of life. This highlights the need for more supportive strategies for nerve regrowth and adequate recovery. Multifunctional cytokine transforming growth factor-β (TGF-β) is essential for the development of the nervous system and is known for its neuroprotective functions. Accumulating evidence indicates its involvement in multiple cellular and molecular responses that are critical to peripheral nerve repair. Following PNI, TGF-β is released at the site of injury where it can initiate a series of phenotypic changes in Schwann cells (SCs), modulate immune cells, activate neuronal intrinsic growth capacity, and regulate blood nerve barrier (BNB) permeability, thus enhancing the regeneration of the nerves. Notably, TGF-β has already been applied experimentally in the treatment of PNI. These treatments with encouraging outcomes further demonstrate its regeneration-promoting capacity. Herein, we review the possible roles of TGF-β in peripheral nerve regeneration and discuss the underlying mechanisms, thus providing new cues for better treatment of PNI.
Collapse
Affiliation(s)
- Zhiqian Ye
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junbin Wei
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chaoning Zhan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Hou
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Jin Hou,
| |
Collapse
|
25
|
LUO X, YIN J, CAI Y, LIN S, TONG C, SUI H, YE M, Long Y, LIN P, LAN T. Cytoplasm or supernatant—where is the treasury of the bioactive antiaging factor from mesenchymal stem cells? Stem Cells Dev 2022; 31:529-540. [PMID: 35491559 DOI: 10.1089/scd.2021.0245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Xuewei LUO
- Xiamen Medical College, 519884, Xiamen, Fujian, China
- Guangxi University, 12664, Medicinal College, Nanning, Guangxi, China
| | - Jingwen YIN
- Xiamen Medical College, 519884, Xiamen, Fujian, China
| | - Yiwen CAI
- Xiamen Medical College, 519884, Xiamen, Fujian, China
| | | | | | - Huaxiu SUI
- Xiamen Medical College, 519884, Xiamen, Fujian, China
| | - Mingzhu YE
- Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Yufei Long
- Xiamen Medical College, 519884, Xiamen, Fujian, China
- Guangxi University, 12664, Medicinal College, Nanning, Guangxi, China
| | - Pingli LIN
- Xiamen Fifth Hospital, Department of Obstetrics, Xiamen, Fujian, China
| | - Tianshu LAN
- Xiamen Medical College, 519884, Xiamen, China
- Xiamen Medical College, 519884, Key laboratory of functional and clinical translational medicine, Fujian province university, Xiamen, Fujian, China
| |
Collapse
|
26
|
Yuan X, Duan X, Li Z, Yao B, Enhejirigala, Song W, Kong Y, Wang Y, Zhang F, Liang L, Zhu S, Zhang M, Zhang C, Huang S, Fu X. Collagen triple helix repeat containing-1 promotes functional recovery of sweat glands by inducing adjacent microvascular network reconstruction in vivo. BURNS & TRAUMA 2022; 10:tkac035. [PMID: 35937591 PMCID: PMC9346565 DOI: 10.1093/burnst/tkac035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/30/2022] [Accepted: 06/14/2022] [Indexed: 11/23/2022]
Abstract
Background Sweat glands (SGs) have low regenerative potential after severe burns or trauma and their regeneration or functional recovery still faces many obstacles. In practice, restoring SG function requires not only the structural integrity of the gland itself, but also its neighboring tissues, especially blood vessels. Collagen triple helix repeat containing-1 (CTHRC1) was first identified in vascular repair, and increasing reports showed a close correlation between cutaneous appendage specification, patterning and regeneration. The purpose of the present study was to clarify the role of CTHRC1 in SGs and their adjacent microvessels and find therapeutic strategies to restore SG function. Methods The SGs and their adjacent microvascular network of Cthrc1−/− mice were first investigated using sweat test, laser Doppler imaging, tissue clearing technique and transcriptome analysis. The effects of CTHRC1 on dermal microvascular endothelial cells (DMECs) were further explored with cell proliferation, DiI-labeled acetylated low-density lipoprotein uptake, tube formation and intercellular junction establishment assays. The effects of CTHRC1 on SG function restoration were finally confirmed by replenishing the protein into the paws of Cthrc1−/− mice. Results CTHRC1 is a key regulator of SG function in mice. At the tissue level, Cthrc1 deletion resulted in the disorder and reduction of the microvascular network around SGs. At the molecular level, the knockout of Cthrc1 reduced the expression of vascular development genes and functional proteins in the dermal tissues. Furthermore, CTHRC1 administration considerably enhanced SG function by inducing adjacent vascular network reconstruction. Conclusions CTHRC1 promotes the development, morphogenesis and function execution of SGs and their neighboring vasculature. Our study provides a novel target for the restoration or regeneration of SG function in vivo.
Collapse
Affiliation(s)
- Xingyu Yuan
- School of Medicine , Nankai University, 94 Wei Jin Road, Tianjin 300071, PR China
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
| | - Xianlan Duan
- School of Medicine , Nankai University, 94 Wei Jin Road, Tianjin 300071, PR China
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
| | - Zhao Li
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Chinese PLA General Hospital and PLA Medical College , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
| | - Bin Yao
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Chinese PLA General Hospital and PLA Medical College , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Academy of Medical Engineering and Translational Medicine, Tianjin University , 92 Weijin Road, Tianjin, 300072, PR China
| | - Enhejirigala
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
- College of Graduate, Tianjin Medical University , Tianjin 300070, PR China
- Institute of Basic Medical Research, Inner Mongolia Medical University , Hohhot 010110, Inner Mongolia, PR China
| | - Wei Song
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Chinese PLA General Hospital and PLA Medical College , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
| | - Yi Kong
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Chinese PLA General Hospital and PLA Medical College , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
| | - Yuzhen Wang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Chinese PLA General Hospital and PLA Medical College , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Department of Burn and Plastic Surgery, Air Force Hospital of Chinese PLA Central Theater Command , Datong 037000, Shanxi, PR China
| | - Fanliang Zhang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Chinese PLA General Hospital and PLA Medical College , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
| | - Liting Liang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
| | - Shijun Zhu
- School of Medicine , Nankai University, 94 Wei Jin Road, Tianjin 300071, PR China
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
| | - Mengde Zhang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Chinese PLA General Hospital and PLA Medical College , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
| | - Chao Zhang
- School of Medicine , Nankai University, 94 Wei Jin Road, Tianjin 300071, PR China
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
| | - Sha Huang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
| | - Xiaobing Fu
- School of Medicine , Nankai University, 94 Wei Jin Road, Tianjin 300071, PR China
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital , 28 Fu Xing Road, Beijing 100853, PR China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Chinese PLA General Hospital and PLA Medical College , Repair and Regeneration, , 51 Fu Cheng Road, Beijing 100048, PR China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051 , Beijing 100048, PR China
| |
Collapse
|
27
|
Song N, Pan K, Chen L, Jin K. Platelet Derived Vesicles Enhance the TGF-beta Signaling Pathway of M1 Macrophage. Front Endocrinol (Lausanne) 2022; 13:868893. [PMID: 35370988 PMCID: PMC8972998 DOI: 10.3389/fendo.2022.868893] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/17/2022] [Indexed: 01/13/2023] Open
Abstract
Macrophages, mainly divided into M1 pro-inflammatory and M2 anti-inflammatory types, play a key role in the transition from inflammation to repair after trauma. In chronic inflammation, such as diabetes and complex bone injury, or the process of certain inflammatory specific emergencies, the ratio of M1/M2 cell populations is imbalanced so that M1-macrophages cannot be converted into M2 macrophages in time, resulting in delayed trauma repair. Early and timely transformation of macrophages from the pro-inflammatory M1-type into the pro-reparative M2-type is an effective strategy to guide trauma repair and establish the original homeostasis. We prepared purified nano-platelet vesicles (NPVs) and assessed their effects on macrophage phenotype switching through transcriptome analysis. The results elucidate that NPVs promote pathways related to angiogenesis, collagen synthesis, cell adhesion, and migration in macrophages, and we speculate that these advantages may promote healing in traumatic diseases.
Collapse
Affiliation(s)
- Nan Song
- Department of Pathophysiology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Intelligent Critical Care and Life Support Research of Zhejiang Province, Wenzhou, China
- Zhejiang Decell Biotechnology Co. LTD, Hangzhou, China
| | - Kaifeng Pan
- Department of Pathophysiology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Intelligent Critical Care and Life Support Research of Zhejiang Province, Wenzhou, China
| | - Lei Chen
- Key Laboratory of Intelligent Critical Care and Life Support Research of Zhejiang Province, Wenzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Lei Chen, ; Keke Jin,
| | - Keke Jin
- Department of Pathophysiology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Intelligent Critical Care and Life Support Research of Zhejiang Province, Wenzhou, China
- *Correspondence: Lei Chen, ; Keke Jin,
| |
Collapse
|
28
|
Nordquist EM, Dutta P, Kodigepalli KM, Mattern C, McDermott MR, Trask AJ, LaHaye S, Lindner V, Lincoln J. Tgfβ1-Cthrc1 Signaling Plays an Important Role in the Short-Term Reparative Response to Heart Valve Endothelial Injury. Arterioscler Thromb Vasc Biol 2021; 41:2923-2942. [PMID: 34645278 PMCID: PMC8612994 DOI: 10.1161/atvbaha.121.316450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/23/2021] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Aortic valve disease is a common worldwide health burden with limited treatment options. Studies have shown that the valve endothelium is critical for structure-function relationships, and disease is associated with its dysfunction, damage, or injury. Therefore, therapeutic targets to maintain a healthy endothelium or repair damaged endothelial cells could hold promise. In this current study, we utilize a surgical mouse model of heart valve endothelial cell injury to study the short-term response at molecular and cellular levels. The goal is to determine if the native heart valve exhibits a reparative response to injury and identify the mechanisms underlying this process. Approach and Results: Mild aortic valve endothelial injury and abrogated function was evoked by inserting a guidewire down the carotid artery of young (3 months) and aging (16-18 months) wild-type mice. Short-term cellular responses were examined at 6 hours, 48 hours, and 4 weeks following injury, whereas molecular profiles were determined after 48 hours by RNA-sequencing. Within 48 hours following endothelial injury, young wild-type mice restore endothelial barrier function in association with increased cell proliferation, and upregulation of transforming growth factor beta 1 (Tgfβ1) and the glycoprotein, collagen triple helix repeat containing 1 (Cthrc1). Interestingly, this beneficial response to injury was not observed in aging mice with known underlying endothelial dysfunction. CONCLUSIONS Data from this study suggests that the healthy valve has the capacity to respond to mild endothelial injury, which in short term has beneficial effects on restoring endothelial barrier function through acute activation of the Tgfβ1-Cthrc1 signaling axis and cell proliferation.
Collapse
Affiliation(s)
- Emily M. Nordquist
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
- Department of Pediatrics, Section of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Punashi Dutta
- Department of Pediatrics, Section of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Karthik M. Kodigepalli
- Department of Pediatrics, Section of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Carol Mattern
- Department of Pediatrics, Section of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| | - Michael R. McDermott
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Aaron J. Trask
- Center for Cardiovascular Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Stephanie LaHaye
- The Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Volkhard Lindner
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Joy Lincoln
- Department of Pediatrics, Section of Pediatric Cardiology, Medical College of Wisconsin, Milwaukee, WI, USA
- The Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
29
|
The bright side of fibroblasts: molecular signature and regenerative cues in major organs. NPJ Regen Med 2021; 6:43. [PMID: 34376677 PMCID: PMC8355260 DOI: 10.1038/s41536-021-00153-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
Fibrosis is a pathologic process characterized by the replacement of parenchymal tissue by large amounts of extracellular matrix, which may lead to organ dysfunction and even death. Fibroblasts are classically associated to fibrosis and tissue repair, and seldom to regeneration. However, accumulating evidence supports a pro-regenerative role of fibroblasts in different organs. While some organs rely on fibroblasts for maintaining stem cell niches, others depend on fibroblast activity, particularly on secreted molecules that promote cell adhesion, migration, and proliferation, to guide the regenerative process. Herein we provide an up-to-date overview of fibroblast-derived regenerative signaling across different organs and discuss how this capacity may become compromised with aging. We further introduce a new paradigm for regenerative therapies based on reverting adult fibroblasts to a fetal/neonatal-like phenotype.
Collapse
|
30
|
Xia Z, Wang J, Yang S, Liu C, Qin S, Li W, Cheng Y, Hu H, Qian J, Liu Y, Deng C. Emodin alleviates hypertrophic scar formation by suppressing macrophage polarization and inhibiting the Notch and TGF-β pathways in macrophages. ACTA ACUST UNITED AC 2021; 54:e11184. [PMID: 34320121 PMCID: PMC8302142 DOI: 10.1590/1414-431x2021e11184] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/01/2021] [Indexed: 01/10/2023]
Abstract
Hypertrophic scar (HS) formation is a common complication that develops after skin injury; however, there are few effective and specific therapeutic approaches for HS. Emodin has previously been reported to inhibit mechanical stress-induced HS inflammation. Here, we investigated the molecular mechanisms underlying the inhibitory effects of emodin on HS formation. First, we conducted in vitro assays that revealed that emodin inhibited M1 and M2 polarization in rat macrophages. We subsequently established a combined rat model of tail HS and dorsal subcutaneous polyvinyl alcohol (PVA) sponge-induced wounds. Rats were treated with emodin or vehicle (DMEM). Tail scar specimens were harvested at 14, 28, and 42 days post-incision and subjected to H&E staining and Masson's trichrome staining. Histopathological analyses confirmed that emodin attenuated HS formation and fibrosis. Macrophages were separated from wound cells collected from the PVA sponge at 3 and 7 days after implantation. Flow cytometry analysis demonstrated that emodin suppressed in vivo macrophage recruitment and polarization at the wound site. Finally, we explored the molecular mechanisms of emodin in modulating macrophage polarization by evaluating the expression levels of selected effectors of the Notch and TGF-β pathways in macrophages isolated from PVA sponges. Western blot and qPCR assays showed that Notch1, Notch4, Hes1, TGF-β, and Smad3 were downregulated in response to emodin treatment. Taken together, our findings suggested that emodin attenuated HS formation and fibrosis by suppressing macrophage polarization, which is associated with the inhibition of the Notch and TGF-β pathways in macrophages.
Collapse
Affiliation(s)
- Zihuan Xia
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiancheng Wang
- Department of General Surgery, Rujin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Songlin Yang
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cheng Liu
- Department of Plastic Surgery, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Shu Qin
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wenbo Li
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yulong Cheng
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Huan Hu
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jin Qian
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yi Liu
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chenliang Deng
- Department of Plastic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
31
|
Zhang XL, Hu LP, Yang Q, Qin WT, Wang X, Xu CJ, Tian GA, Yang XM, Yao LL, Zhu L, Nie HZ, Li Q, Xu Q, Zhang ZG, Zhang YL, Li J, Wang YH, Jiang SH. CTHRC1 promotes liver metastasis by reshaping infiltrated macrophages through physical interactions with TGF-β receptors in colorectal cancer. Oncogene 2021; 40:3959-3973. [PMID: 33986509 DOI: 10.1038/s41388-021-01827-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/13/2021] [Accepted: 04/27/2021] [Indexed: 02/03/2023]
Abstract
Metastasis is a major cause of cancer-related deaths. Tumor-intrinsic properties can determine whether tumor metastasis occurs or not. Here, by comparing the gene expression patterns in primary colorectal cancer (CRC) patients with or without metastasis, we found that Collagen Triple Helix Repeat Containing 1 (CTHRC1) in primary CRC served as a metastasis-associated gene. Animal experiments verified that CTHRC1 secreted by CRC cells promoted hepatic metastasis, which was closely correlated with macrophage infiltration. Depletion of macrophages by liposomal clodronate largely abolished the promoting effect of CTHRC1 on CRC liver metastasis. Furthermore, we demonstrated that CTHRC1 modulated macrophage polarization to M2 phenotypes through TGF-β signaling. A mechanistic study revealed that CTHRC1 bound directly to TGF-β receptor II and TGF-β receptor III, stabilized the TGF-β receptor complex, and activated TGF-β signaling. The combination treatment of CTHRC1 monoclonal antibody and anti-PD-1 blocking antibody effectively suppressed CRC hepatic metastasis. Taken together, our data demonstrated that CTHRC1 is an intrinsic marker of CRC metastasis and further revealed that CTHRC1 promoted CRC liver metastasis by reshaping infiltrated macrophages through TGF-β signaling, suggesting that CTHRC1 could be a potential biomarker for the early prediction of and a therapeutic target of CRC hepatic metastasis.
Collapse
Affiliation(s)
- Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Ting Qin
- Department of Radiation Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chun-Jie Xu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guang-Ang Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Mei Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin-Li Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Zhen Nie
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Xu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ya-Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
32
|
Shen P, Chen Y, Luo S, Fan Z, Wang J, Chang J, Deng J. Applications of biomaterials for immunosuppression in tissue repair and regeneration. Acta Biomater 2021; 126:31-44. [PMID: 33722787 DOI: 10.1016/j.actbio.2021.03.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022]
Abstract
The immune system plays an essential role in tissue repair and regeneration. Regardless of innate or adaptive immune responses, immunosuppressive strategies such as macrophage polarization and regulatory T (Treg) cell induction can be used to modulate the immune system to promote tissue repair and regeneration. Biomaterials can improve the production of anti-inflammatory macrophages and Treg cells by providing physiochemical cues or delivering therapeutics such as cytokines, small molecules, microRNA, growth factors, or stem cells in the damaged tissues. Herein, we present an overview of immunosuppressive modulation by biomaterials in tissue regeneration and highlight the mechanisms of macrophage polarization and Treg cell induction. Overall, we foresee that future biomaterials for regenerative strategies will entail more interactions between biomaterials and the immune cells, and more mechanisms of immunosuppression related to T cell subsets remain to be discovered and applied to develop novel biomaterials for tissue repair and regeneration. STATEMENT OF SIGNIFICANCE: Immunosuppression plays a key role in tissue repair and regeneration, and biomaterials can interact with the immune system through their biological properties and by providing physiochemical cues. Here, we summarize the studies on biomaterials that have been used for immunosuppression to facilitate tissue regeneration. In the first part of this review, we demonstrate the crucial role of macrophage polarization and induction of T regulatory (Treg) cells in immunosuppression. In the second part, distinct approaches used by biomaterials to induce immunosuppression are introduced, which show excellent performance in terms of promoting tissue regeneration.
Collapse
Affiliation(s)
- Peng Shen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Yanxin Chen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Shuai Luo
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Zhiyuan Fan
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA 19104, USA
| | - Jilong Wang
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Jiang Chang
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China; State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.
| | - Junjie Deng
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
| |
Collapse
|
33
|
CTHRC1 Knockdown Promotes Inflammatory Responses Partially by p38 MAPK Activation in Human Periodontal Ligament Cells. Inflammation 2021; 44:1831-1842. [PMID: 33846931 DOI: 10.1007/s10753-021-01461-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/18/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022]
Abstract
Collagen triple helix repeat containing 1 (CTHRC1), a secreted glycoprotein, is widely expressed in many tissues. It has been recently defined as a novel marker for rheumatoid arthritis (RA), a systemic inflammatory disorder. However, the precise role of CTHRC1 in other chronic inflammatory diseases, like periodontal disease, remains unclear. This research aimed to explore the presence of CTHRC1 in periodontal inflammation, determine the precise role in inflammatory response modulation in periodontal ligament cells (PDLCs), and explore its underlying mechanisms. In vivo gingival crevicular fluid (GCF) and gingivae were obtained from healthy people and chronic periodontitis patients. Maxillary tissues of mice with or without ligature-induced periodontitis were immunostained for CTHRC1. In vitro human PDLCs were treated with tumor necrosis factor alpha (TNF-α) to mimic the inflammatory environment. Small interfering RNA (siRNA) was used to silence CTHRC1. SB203580 was used to inhibit the p38 mitogen-activated protein kinase (MAPK) pathway. CTHRC1 was highly expressed in GCF and gingival tissues of periodontitis patients. Animal models also revealed the same tendency. CTHRC1 knockdown promoted inflammatory cytokine production and activated the p38 MAPK signaling pathway in PDLCs. Inhibiting the p38 MAPK signaling pathway partially attenuated the inflammatory responses. This study revealed that CTHRC1 was highly expressed in periodontitis and suggested that CTHRC1 might play an important role in modulating periodontal inflammation.
Collapse
|
34
|
Skin Immunomodulation during Regeneration: Emerging New Targets. J Pers Med 2021; 11:jpm11020085. [PMID: 33573342 PMCID: PMC7911085 DOI: 10.3390/jpm11020085] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/25/2020] [Accepted: 01/07/2021] [Indexed: 02/08/2023] Open
Abstract
Adipose-Derived Stem Cells (ADSC) are present within the hypodermis and are also expected to play a pivotal role in wound healing, immunomodulation, and rejuvenation activities. They orchestrate, through their exosome, the mechanisms associated to cell differentiation, proliferation, and cell migration by upregulating genes implicated in different functions including skin barrier, immunomodulation, cell proliferation, and epidermal regeneration. ADSCs directly interact with their microenvironment and specifically the immune cells, including macrophages and T and B cells, resulting in differential inflammatory and anti-inflammatory mechanisms impacting, in return, ADSCs microenvironment and thus skin function. These useful features of ADSCs are involved in tissue repair, where the required cell proliferation, angiogenesis, and anti-inflammatory responses should occur rapidly in damaged sites. Different pathways involved have been reported such as Growth Differentiation Factor-11 (GDF11), Tumor Growth Factor (TGF)-β, Metalloproteinase (MMP), microRNA, and inflammatory cytokines that might serve as specific biomarkers of their immunomodulating capacity. In this review, we try to highlight ADSCs’ network and explore the potential indicators of their immunomodulatory effect in skin regeneration and aging. Assessment of these biomarkers might be useful and should be considered when designing new clinical therapies using ADSCs or their specific exosomes focusing on their immunomodulation activity.
Collapse
|
35
|
Doykov I, Hällqvist J, Gilmour KC, Grandjean L, Mills K, Heywood WE. 'The long tail of Covid-19' - The detection of a prolonged inflammatory response after a SARS-CoV-2 infection in asymptomatic and mildly affected patients. F1000Res 2021; 9:1349. [PMID: 33391730 PMCID: PMC7745182 DOI: 10.12688/f1000research.27287.2] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
'Long Covid', or medical complications associated with post SARS-CoV-2 infection, is a significant post-viral complication that is being more and more commonly reported in patients. Therefore, there is an increasing need to understand the disease mechanisms, identify drug targets and inflammatory processes associated with a SARS-CoV-2 infection. To address this need, we created a targeted mass spectrometry based multiplexed panel of 96 immune response associated proteins. We applied the multiplex assay to a cohort of serum samples from asymptomatic and moderately affected patients. All patients had tested positive for a SARS-CoV-2 infection by PCR and were determined to be subsequently positive for antibodies. Even 40-60 days post-viral infection, we observed a significant remaining inflammatory response in all patients. Proteins that were still affected were associated with the anti-inflammatory response and mitochondrial stress. This indicates that biochemical and inflammatory pathways within the body can remain perturbed long after SARS-CoV-2 infections have subsided even in asymptomatic and moderately affected patients.
Collapse
Affiliation(s)
- Ivan Doykov
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, UK
| | - Jenny Hällqvist
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, UK,Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, UK
| | - Kimberly C. Gilmour
- Great Ormond Street Children's Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Louis Grandjean
- Great Ormond Street Children's Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Kevin Mills
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, UK
| | - Wendy E. Heywood
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, UK,
| |
Collapse
|
36
|
Doykov I, Hällqvist J, Gilmour KC, Grandjean L, Mills K, Heywood WE. 'The long tail of Covid-19' - The detection of a prolonged inflammatory response after a SARS-CoV-2 infection in asymptomatic and mildly affected patients. F1000Res 2021; 9:1349. [PMID: 33391730 PMCID: PMC7745182 DOI: 10.12688/f1000research.27287.1] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/12/2020] [Indexed: 12/14/2022] Open
Abstract
'Long Covid', or medical complications associated with post SARS-CoV-2 infection, is a significant post-viral complication that is being more and more commonly reported in patients. Therefore, there is an increasing need to understand the disease mechanisms, identify drug targets and inflammatory processes associated with a SARS-CoV-2 infection. To address this need, we created a targeted mass spectrometry based multiplexed panel of 96 immune response associated proteins. We applied the multiplex assay to a cohort of serum samples from asymptomatic and moderately affected patients. All patients had tested positive for a SARS-CoV-2 infection by PCR and were determined to be subsequently positive for antibodies. Even 40-60 days post-viral infection, we observed a significant remaining inflammatory response in all patients. Proteins that were still affected were associated with the anti-inflammatory response and mitochondrial stress. This indicates that biochemical and inflammatory pathways within the body can remain perturbed long after SARS-CoV-2 infections have subsided even in asymptomatic and moderately affected patients.
Collapse
Affiliation(s)
- Ivan Doykov
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, UK
| | - Jenny Hällqvist
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, UK,Clinical and Movement Neurosciences, University College London Queen Square Institute of Neurology, London, UK
| | - Kimberly C. Gilmour
- Great Ormond Street Children's Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Louis Grandjean
- Great Ormond Street Children's Hospital NHS Foundation Trust, Great Ormond Street, London, WC1N 3JH, UK
| | - Kevin Mills
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, UK
| | - Wendy E. Heywood
- Translational Mass Spectrometry Research Group, University College London Institute of Child Health, London, UK,
| |
Collapse
|
37
|
Mazini L, Rochette L, Malka G. Adipose-Derived Stem Cells (ADSCs) and Growth Differentiation Factor 11 (GDF11): Regenerative and Antiaging Capacity for the Skin. Regen Med 2020. [DOI: 10.5772/intechopen.91233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
38
|
The Role of CTHRC1 in Regulation of Multiple Signaling and Tumor Progression and Metastasis. Mediators Inflamm 2020; 2020:9578701. [PMID: 32848510 PMCID: PMC7441421 DOI: 10.1155/2020/9578701] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
Collagen triple helix repeat containing-1 (CTHRC1) has been identified as cancer-related protein. CTHRC1 expresses mainly in adventitial fibroblasts and neointimal smooth muscle cells of balloon-injured vessels and promotes cell migration and tissue repair in response to injury. CTHRC1 plays a pivotal role in some pathophysiological processes, including increasing bone mass, preventing myelination, and reversing collagen synthesis in many tumor cells. The ascended expression of CTHRC1 is related to tumorigenesis, proliferation, invasion, and metastasis in various human malignancies, including gastric cancer, pancreatic cancer, hepatocellular carcinoma, keloid, breast cancer, colorectal cancer, epithelial ovarian cancer, esophageal squamous cell carcinoma, cervical cancer, non-small-cell lung carcinoma, and melanoma. And molecules that regulate the expression of CTHRC1 include miRNAs, lncRNAs, WAIF1, and DPAGT1. Many reports have pointed that CTHRC1 could exert different effects through several signaling pathways such as TGF-β, Wnt, integrin β/FAK, Src/FAK, MEK/ERK, PI3K/AKT/ERK, HIF-1α, and PKC-δ/ERK signaling pathways. As a participant in tissue remodeling or immune response, CTHRC1 may promote early-stage cancer. Several recent studies have identified CTHRC1 as an effectual prognostic biomarker for predicting tumor recurrence or metastasis. It is worth noting that CTHRC1 has different cellular localization and mechanisms of action in different cells and different microenvironments. In this article, we focus on the advances in the signaling pathways mediated by CTHRC1 in tumors.
Collapse
|
39
|
He D, Zhao F, Jiang H, Kang Y, Song Y, Lin X, Shi P, Zhang T, Pang X. LOXL2 from human amniotic mesenchymal stem cells accelerates wound epithelialization by promoting differentiation and migration of keratinocytes. Aging (Albany NY) 2020; 12:12960-12986. [PMID: 32621591 PMCID: PMC7377892 DOI: 10.18632/aging.103384] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 05/01/2020] [Indexed: 12/11/2022]
Abstract
In this study, we identified wound healing-related proteins secreted by human amniotic epithelial cells (hAECs) and human amniotic mesenchymal stem cells (hAMSCs). We observed increased migration and reduced proliferation and differentiation when keratinocytes were co-cultured in media conditioned by hAECs (hAECs-CM) and hAMSCs (hAMSCs-CM). Label-free mass spectrometry and bioinformatic analyses of the hAECs-CM and hAMSCs-CM proteome revealed several proteins associated with wound healing, angiogenesis, cellular differentiation, immune response and cell motility. The levels of the proteins related to wound healing, including CTHRC1, LOXL2 and LGALS1, were significantly higher in hAMSCs-CM than hAECs-CM. LOXL2 significantly enhanced in vitro keratinocyte migration and differentiation compared to CTHRC1 and LGALS1. Moreover, LOXL2 enhanced keratinocyte migration and differentiation by activating the JNK signaling pathway. We observed significant reduction in the in vitro migration and differentiation of keratinocytes when co-cultured with medium conditioned by LOXL2-silenced hAMSCs and when treated with 10 μM SP600125, a specific JNK inhibitor. Treatment with hAMSCs-CM and LOXL2 significantly accelerated wound healing in the murine skin wound model. These findings show that LOXL2 promotes wound healing by inducing keratinocyte migration and differentiation via a JNK signaling pathway.
Collapse
Affiliation(s)
- Dan He
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang 110013, Liaoning, China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang 110013, Liaoning, China
| | - Han Jiang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Yue Kang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Yang Song
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang 110013, Liaoning, China.,Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Xuewen Lin
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang 110013, Liaoning, China
| | - Ping Shi
- Shenyang Amnion Bioengineering and Technology R & D Center, Shenyang Liaoning Amnion Stem Cell and Regenerative Medicine Professional Technology Innovation Platform, Liaoning Human Amniotic Membrane Biological Dressing Stem Cell and Regenerative Medicine Engineering Research Center, Shenyang 110015, Liaoning, China
| | - Tao Zhang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang 110013, Liaoning, China
| | - Xining Pang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, Shenyang 110013, Liaoning, China.,Shenyang Amnion Bioengineering and Technology R & D Center, Shenyang Liaoning Amnion Stem Cell and Regenerative Medicine Professional Technology Innovation Platform, Liaoning Human Amniotic Membrane Biological Dressing Stem Cell and Regenerative Medicine Engineering Research Center, Shenyang 110015, Liaoning, China
| |
Collapse
|
40
|
Arenas Gómez CM, Sabin KZ, Echeverri K. Wound healing across the animal kingdom: Crosstalk between the immune system and the extracellular matrix. Dev Dyn 2020; 249:834-846. [PMID: 32314465 PMCID: PMC7383677 DOI: 10.1002/dvdy.178] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/10/2020] [Accepted: 04/11/2020] [Indexed: 12/11/2022] Open
Abstract
Tissue regeneration is widespread in the animal kingdom. To date, key roles for different molecular and cellular programs in regeneration have been described, but the ultimate blueprint for this talent remains elusive. In animals capable of tissue regeneration, one of the most crucial stages is wound healing, whose main goal is to close the wound and prevent infection. In this stage, it is necessary to avoid scar formation to facilitate the activation of the immune system and remodeling of the extracellular matrix, key factors in promoting tissue regeneration. In this review, we will discuss the current state of knowledge regarding the role of the immune system and the interplay with the extracellular matrix to trigger a regenerative response.
Collapse
Affiliation(s)
- Claudia M. Arenas Gómez
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological LaboratoryWoods HoleMassachusettsUSA
| | - Keith Z. Sabin
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological LaboratoryWoods HoleMassachusettsUSA
| | - Karen Echeverri
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological LaboratoryWoods HoleMassachusettsUSA
| |
Collapse
|
41
|
Kong X, Fu J, Shao K, Wang L, Lan X, Shi J. Biomimetic hydrogel for rapid and scar-free healing of skin wounds inspired by the healing process of oral mucosa. Acta Biomater 2019; 100:255-269. [PMID: 31606531 DOI: 10.1016/j.actbio.2019.10.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/26/2019] [Accepted: 10/04/2019] [Indexed: 12/18/2022]
Abstract
Inspired by the wound healing characteristics of the oral mucosa, a biomimetic hydrogel was prepared to realize the rapid and scar-free healing of skin wounds. Through monitoring the healing process of injured oral mucosa, we find out that the combination of high, rapid and sequential expression of some growth factors and the sterile-moist microenvironment are crucial for re-epithelialization and precise control of the inflammation process. On the base of our findings, a hydrogel loaded with several functional compounds was prepared to achieve a comprehensive simulation of the oral mucosal trauma microenvironment for skin wound healing. After 7 days treatment, the skin wound area of the treated group was only about 20% of that of the untreated group, and the proportion of collagen type III and type I in the treated group was much higher than that of the untreated group, suggesting lighter scar hyperplasia. The comprehensive treatment strategy of sequential expression of growth factors in combination with maintaining of a sterile and humid environment is expected to have great application prospect in the field of chronic trauma repair and cosmetic surgery. STATEMENT OF SIGNIFICANCE: Long healing time and scar hyperplasia during wound healing have been a serious problem in the past decades of wound healing research. Oral cavity wound healing occurs in an environment that sustains ongoing physical trauma and is rich in bacteria. Despite this, injuries to the mucosal surface often heal faster than cutaneous wounds and leave less noticeable scars. Therefore, in recent years, many scholars have begun to study the healing mechanism of oral mucosa, which supports a new inspiration for the study of skin wound repair: whether the injured skin can achieve a rapid scar-free healing effect similar to oral mucosa? Imitating the biological process of oral mucosa wound healing would be a promising therapeutic strategy in wound healing. Therefore, inspired by the wound healing characteristics of the oral mucosa, a biomimetic gel was prepared to realize the rapid and scar-free healing of skin wounds. Through monitoring the healing process of injured oral mucosa, the combination of high, rapid and sequential expression of some growth factors and sterile-moist microenvironment was crucial for re-epithelialization and precise control of the inflammation process. The comprehensive treatment strategy of sequential expression of growth factors in combination with maintance of a sterile and humid environment implies its potential use in the field of chronic trauma repair and cosmetic surgery.
Collapse
Affiliation(s)
- Xiaoying Kong
- College of Chemistry and Pharmacy, Qingdao Agricultural University, 700 Changcheng Road, Qingdao 266109, China
| | - Jun Fu
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 Zhongguan West Road, Ningbo 315200, China
| | - Kai Shao
- Medical Experimental Center, Qilu Hospital of Shandong University (Qingdao), 758 Hefei Road, Qingdao 266035, China
| | - Lili Wang
- College of Chemistry and Pharmacy, Qingdao Agricultural University, 700 Changcheng Road, Qingdao 266109, China
| | - Xuefang Lan
- College of Chemistry and Pharmacy, Qingdao Agricultural University, 700 Changcheng Road, Qingdao 266109, China
| | - Jinsheng Shi
- College of Chemistry and Pharmacy, Qingdao Agricultural University, 700 Changcheng Road, Qingdao 266109, China.
| |
Collapse
|