1
|
Wu J, Qian Y, Yang K, Zhang S, Zeng E, Luo D. Innate immune cells in vascular lesions: mechanism and significance of diversified immune regulation. Ann Med 2025; 57:2453826. [PMID: 39847394 PMCID: PMC11758805 DOI: 10.1080/07853890.2025.2453826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/18/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
Angiogenesis is a complex physiological process. In recent years, the immune regulation of angiogenesis has received increasing attention, and innate immune cells, which are centred on macrophages, are thought to play important roles in vascular neogenesis and development. Various innate immune cells can act on the vasculature through a variety of mechanisms, with commonalities as well as differences and synergistic effects, which are crucial for the progression of vascular lesions. In recent years, monotherapy with antiangiogenic drugs has encountered therapeutic bottlenecks because of the short-term effect of 'vascular normalization'. The combination treatment of antiangiogenic therapy and immunotherapy breaks the traditional treatment pattern. While it has a remarkable curative effect and survival benefits, it also faces many challenges. This review focuses on innate immune cells and mainly introduces the regulatory mechanisms of monocytes, macrophages, natural killer (NK) cells, dendritic cells (DCs) and neutrophils in vascular lesions. The purpose of this paper was to elucidate the underlying mechanisms of angiogenesis and development and the current research status of innate immune cells in regulating vascular lesions in different states. This review provides a theoretical basis for addressing aberrant angiogenesis in disease processes or finding new antiangiogenic immune targets in inflammation and tumor.
Collapse
Affiliation(s)
- Jinjing Wu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yulu Qian
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Kuang Yang
- Queen Mary University of London, Nanchang University, Nanchang, China
| | - Shuhua Zhang
- Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Jiangxi Cardiovascular Research Institute, Nanchang, Jiangxi, China
| | - Erming Zeng
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Daya Luo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
2
|
Zhang YW, Sun L, Wang YN, Zhan SY. Role of macrophage polarization in diabetic foot ulcer healing: A bibliometric study. World J Diabetes 2025; 16:99755. [PMID: 39817209 PMCID: PMC11718451 DOI: 10.4239/wjd.v16.i1.99755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Diabetic foot ulcers (DFUs) are a significant contributor to disability and mortality in diabetic patients. Macrophage polarization and functional regulation are promising areas of research and show therapeutic potential in the field of DFU healing. However, the complex mechanism, the difficulty in clinical translation, and the large heterogeneity present significant challenges. Hence, this study was to comprehensively analyze the publication status and trends of studies on macrophage polarization and DFU healing. AIM To examine the relevant literature on macrophage polarization in DFU healing. METHODS A bibliometric analysis was conducted using the Web of Science database. Relevant literature was retrieved from the Web of Science Core Collection database between 2013 to 2023 using literature visualization and analysis software (VOSviewer and CiteSpace) and bibliometric online platforms. The obtained literature was then subjected to visualization and analysis of different countries/regions, institutions, journals, authors, and keywords to reveal the research's major trends and focus. RESULTS The number of publications on the role of macrophage polarization in DFU healing increased rapidly from 2013 to 2023, especially in the latter period. Chinese researchers were the most prolific in this field, with 217 publications, while American researchers had been engaged in this field for a longer period. Qian Tan of Nanjing Drum Tower Hospital and Qian Ding of Nanjing University were the first to publish in this field. Shanghai Jiao Tong University was the institution with the most publications (27). The keywords "bone marrow", "adjustment, replacement, response, tissue repair", and "activation, repair, differentiation" appeared more frequently. The study of macrophage polarization in DFU healing focused on the regulatory mechanism, gene expression, and other aspects. CONCLUSION This study through the bibliometric method reveals the research trends and development trends in this field of macrophage polarization in DFU healing from 2013 to 2023 in the Web of Science Core Collection database. The key hotspots in this field mainly include the regulation of macrophage activation, gene expression, wound tissue repair, and new wound materials. This study provides references for future research directions.
Collapse
Affiliation(s)
- You-Wen Zhang
- Department of Peripheral Vascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Lei Sun
- Department of Surgery, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, Shandong Province, China
| | - Yan-Nan Wang
- Department of Peripheral Vascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Shi-Yu Zhan
- Department of Surgery, Longkou Traditional Chinese Medicine Hospital, Yantai 265701, Shandong Province, China
| |
Collapse
|
3
|
Song J, Wu Y, Chen Y, Sun X, Zhang Z. Epigenetic regulatory mechanism of macrophage polarization in diabetic wound healing (Review). Mol Med Rep 2025; 31:2. [PMID: 39422035 PMCID: PMC11551531 DOI: 10.3892/mmr.2024.13367] [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: 07/27/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Diabetic wounds represent a significant complication of diabetes and present a substantial challenge to global public health. Macrophages are crucial effector cells that play a pivotal role in the pathogenesis of diabetic wounds, through their polarization into distinct functional phenotypes. The field of epigenetics has emerged as a rapidly advancing research area, as this phenomenon has the potential to markedly affect gene expression, cellular differentiation, tissue development and susceptibility to disease. Understanding epigenetic mechanisms is crucial to further exploring disease pathogenesis. A growing body of scientific evidence has highlighted the pivotal role of epigenetics in the regulation of macrophage phenotypes. Various epigenetic mechanisms, such as DNA methylation, histone modification and non‑coding RNAs, are involved in the modulation of macrophage phenotype differentiation in response to the various environmental stimuli present in diabetic wounds. The present review provided an overview of the various changes that take place in macrophage phenotypes and functions within diabetic wounds and discussed the emerging role of epigenetic modifications in terms of regulating macrophage plasticity in diabetic wounds. It is hoped that this synthesis of information will facilitate the elucidation of diabetic wound pathogenesis and the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Jielin Song
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| | - Yuqing Wu
- The First Clinical Medical College, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yunli Chen
- The First Clinical Medical College, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xu Sun
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| | - Zhaohui Zhang
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| |
Collapse
|
4
|
Varady SRS, Greiner D, Roh-Johnson M. Macrophage subtypes inhibit breast cancer proliferation in culture. Mol Biol Cell 2025; 36:br2. [PMID: 39602294 PMCID: PMC11742110 DOI: 10.1091/mbc.e24-06-0241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/11/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
Macrophages are a highly plastic cell type that adopt distinct subtypes and functional states depending on environmental cues. These functional states can vary widely, with distinct macrophages capable of displaying opposing functions. We sought to understand how macrophage subtypes that exist on two ends of a spectrum influence the function of other cells. We used a coculture system with primary human macrophages to probe the effects of macrophage subtypes on breast cancer cell proliferation. Our studies revealed a surprising phenotype in which both macrophage subtypes inhibited cancer cell proliferation compared with cancer cells alone. Of particular interest, using two different proliferation assays with two different breast cancer cell lines, we showed that differentiating macrophages into a "protumor" subtype inhibited breast cancer cell proliferation. These findings are inconsistent with the prevailing interpretation that "protumor" macrophages promote cancer cell proliferation and suggest a re-evaluation of how these interpretations are made.
Collapse
Affiliation(s)
- Sophia R. S. Varady
- Department of Biochemistry, University of Utah School of Medicine; Salt Lake City, UT 84112
| | - Daniel Greiner
- Department of Biochemistry, University of Utah School of Medicine; Salt Lake City, UT 84112
| | - Minna Roh-Johnson
- Department of Biochemistry, University of Utah School of Medicine; Salt Lake City, UT 84112
| |
Collapse
|
5
|
Zhang Z, Yang D, Shen F, Xue TT, Jiang JS, Luo Y, Zhang Y, Song JK, Kuai L, Wang MX, Li B, Ru Y. Epidermal keratinocytes-specific PD-L1 knockout causes delayed healing of diabetic wounds. Int Immunopharmacol 2024; 143:113540. [PMID: 39510031 DOI: 10.1016/j.intimp.2024.113540] [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: 03/25/2024] [Revised: 09/14/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Diabetic ulcers (DUs) is a common complication of diabetes, for which the morbidity and mortality increasing annually worldwide. The deficiency of PD-L1 in keratinocytes (KCs) may be linked to the diabetic wound healing impediments. OBJECTIVE Our study utilized transgenic mice to assess the functions of epidermal KCs-specific PD-L1 in DUs treatment. METHODS AND RESULTS Epidermal KCs-specific PD-L1 knockouted mice demonstrate deteriorated healing rates, concomitant with exacerbated inflammatory infiltration and excessive angiogenesis. The streptozotocin-induced murine diabetes model was used to imitate DUs in-vivo context, and the delayed healing was found under diabetic conditions. We then generated transgenic mice overexpressing PD-L1 in the epidermis. PD-L1 overexpression accelerate the DUs healing process accompanied by a reduction in inflammatory infiltration and a corresponding decreasion of angiogenesis. Therefore, overexpression of PD-L1 accelerates the healing process of DUs. CONCLUSION In sum, epidermal KCs-specific PD-L1 plays vital roles in epidermis regeneration, inflammatory infiltration, and angiogenesis during DUs restoration, and would not be easily to format fibrous scar. Our study elucidated a new therapeutic idea for slow-healing wound care.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Dan Yang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Fang Shen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Ting-Ting Xue
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jing-Si Jiang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Ying Zhang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Jian-Kun Song
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Ming-Xia Wang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China.
| | - Bin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China.
| | - Yi Ru
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China.
| |
Collapse
|
6
|
Radermacher C, Rohde A, Kucikas V, Buhl EM, Wein S, Jonigk D, Jahnen-Dechent W, Neuss S. Various Hydrogel Types as a Potential In Vitro Angiogenesis Model. Gels 2024; 10:820. [PMID: 39727578 DOI: 10.3390/gels10120820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/28/2024] Open
Abstract
Angiogenesis, the formation of new blood vessels, is a fundamental process in both physiological repair mechanisms and pathological conditions, including cancer and chronic inflammation. Hydrogels are commonly used as in vitro models to mimic the extracellular matrix (ECM) and support endothelial cell behavior during angiogenesis. Mesenchymal stem cells further augment cell and tissue growth and are therefore widely used in regenerative medicine. Here we examined the combination of distinct hydrogel types-fibrin, collagen, and human platelet lysate (HPL)-on the formation of capillaries in a co-culture system containing human umbilical vein endothelial cells (HUVECs) and bone marrow-derived mesenchymal stem cells (BM-MSCs). The mechanical properties and structural changes of the hydrogels were characterized through scanning electron microscopy (SEM) and nanoindentation over 10 days. Fibrin and HPL gels sustained complex network formations, with HPL gels promoting even vascular tube formation of up to 10-fold capillary caliber. Collagen gels supported negligible angiogenesis. Our results suggest that HPL gels in combination with MSC-EC co-culture may be employed to obtain robust vascularization in tissue engineering. This study provides a comparative analysis of fibrin, collagen, and HPL hydrogels, focusing on their ability to support angiogenesis under identical conditions. Our findings demonstrate the superior performance of HPL gels in promoting robust vascular structures, highlighting their potential as a versatile tool for in vitro angiogenesis modeling.
Collapse
Affiliation(s)
- Chloé Radermacher
- Biointerface Laboratory, Helmholtz-Institut for Biomedical Engineering, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Annika Rohde
- Biointerface Laboratory, Helmholtz-Institut for Biomedical Engineering, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Vytautas Kucikas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Eva Miriam Buhl
- Electron Microscopy Facility, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Svenja Wein
- Biointerface Laboratory, Helmholtz-Institut for Biomedical Engineering, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Danny Jonigk
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
- German Center for Lung Research (DZL), Biomedical Research in Endstage & Obstructive Lung Disease (BREATH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Willi Jahnen-Dechent
- Biointerface Laboratory, Helmholtz-Institut for Biomedical Engineering, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Sabine Neuss
- Biointerface Laboratory, Helmholtz-Institut for Biomedical Engineering, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
- Institute of Pathology, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| |
Collapse
|
7
|
Martin P, Pardo-Pastor C, Jenkins RG, Rosenblatt J. Imperfect wound healing sets the stage for chronic diseases. Science 2024; 386:eadp2974. [PMID: 39636982 PMCID: PMC7617408 DOI: 10.1126/science.adp2974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Although the age of the genome gave us much insight about how our organs fail with disease, it also suggested that diseases do not arise from mutations alone; rather, they develop as we age. In this Review, we examine how wound healing might act to ignite disease. Wound healing works well when we are younger, repairing damage from accidents, environmental assaults, and battles with pathogens. Yet, with age and accumulation of mutations and tissue damage, the repair process can devolve, leading to inflammation, fibrosis, and neoplastic signaling. We discuss healthy wound responses and how our bodies might misappropriate these pathways in disease. Although we focus predominantly on epithelial-based (lung and skin) diseases, similar pathways might operate in cardiac, muscle, and neuronal diseases.
Collapse
Affiliation(s)
- Paul Martin
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Carlos Pardo-Pastor
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - R Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart & Lung Institute, NIHR Imperial Biomedical Research Centre, Imperial College London, London, UK
| | - Jody Rosenblatt
- The Randall and Cancer Centres King's College London, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|
8
|
Cortada E, Yao J, Xia Y, Dündar F, Zumbo P, Yang B, Rubio-Navarro A, Perder B, Qiu M, Pettinato AM, Homan EA, Stoll L, Betel D, Cao J, Lo JC. Cross-species single-cell RNA-seq analysis reveals disparate and conserved cardiac and extracardiac inflammatory responses upon heart injury. Commun Biol 2024; 7:1611. [PMID: 39627536 PMCID: PMC11615278 DOI: 10.1038/s42003-024-07315-x] [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: 08/12/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
The immune system coordinates the response to cardiac injury and controls regenerative and fibrotic scar outcomes in the heart and subsequent chronic low-grade inflammation associated with heart failure. Adult mice and humans lack the ability to fully recover while adult zebrafish spontaneously regenerate after heart injury. Here we profile the inflammatory response to heart cryoinjury in zebrafish and coronary artery ligation in mouse using single cell transcriptomics. We interrogate the extracardiac reaction to cardiomyocyte necrosis to assess the specific peripheral tissue and immune cell reaction to chronic stress. Cardiac macrophages play a critical role in determining tissue homeostasis by healing versus scarring. We identify distinct transcriptional clusters of monocytes/macrophages (mono/Mϕ) in each species and find analogous pairs in zebrafish and mice. However, the reaction to myocardial injury is largely disparate between mice and zebrafish. The dichotomous response to heart damage between the murine and zebrafish mono/Mϕ and/or the presence of distinct zebrafish mono/Mϕ subtypes may underlie the impaired regenerative process in adult mammals and humans. Our study furnishes a direct cross-species comparison of immune responses between regenerative and profibrotic myocardial injury models, providing a useful resource to the fields of regenerative biology and cardiovascular research.
Collapse
Affiliation(s)
- Eric Cortada
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Jun Yao
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Yu Xia
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Friederike Dündar
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Paul Zumbo
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Boris Yang
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Björn Perder
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Miaoyan Qiu
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Anthony M Pettinato
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
| | - Edwin A Homan
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Lisa Stoll
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA.
- Institute for Computational Biomedicine, Division of Hematology and Medical, Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Jingli Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA.
| | - James C Lo
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA.
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
9
|
Wei W, Qu ZL, Lei L, Zhang P. TREM2-mediated Macrophage Glycolysis Promotes Skin Wound Angiogenesis via the Akt/mTOR/HIF-1α Signaling Axis. Curr Med Sci 2024; 44:1280-1292. [PMID: 39672999 DOI: 10.1007/s11596-024-2946-3] [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: 08/20/2024] [Accepted: 09/30/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE The trigger receptor expressed on myeloid cells-2 (TREM2) pathway in myeloid cells is a key disease-inducing immune signaling hub that is essential for detecting tissue damage and limiting its pathological spread. However, the role and potential mechanisms of TREM2 in wound repair remain unclear. The purpose of this study was to determine the role and mechanism of TREM2 in skin wound healing in mice. METHODS Immunofluorescence staining was used to determine the expression and cellular localization of TREM2 and test the effects of TREM2 knockout on angiogenesis, glycolysis, and lactylation in skin tissue. Western blotting was used to analyze the expression of the Akt/mTOR/HIF-1α signaling pathway in the wounded skin tissues of wild-type (WT) and TREM2 knockout mice. A coimmunoprecipitation assay was used to determine whether HIF-1α, which mediates angiogenesis, is modified by lactylation. RESULTS The number of TREM2+ macrophages was increased, and TREM2+ macrophages mediated angiogenesis after skin injury. TREM2 promoted glycolysis and lactylation in macrophages during wound healing. Mechanistically, TREM2 promoted macrophage glycolysis and angiogenesis in wounded skin tissues by activating the Akt/mTOR/HIF-1α signaling pathway. HIF-1α colocalized with Klac to mediate lactylation in macrophages, and lactate could stabilize the expression of the HIF-1α protein through lactylation. Lactate treatment ameliorated the impaired angiogenesis and delayed wound healing in wounded skin in TREM2 knockout mice. CONCLUSION TREM2+ macrophage-mediated glycolysis can promote angiogenesis and wound healing. Our findings provide an effective strategy and target for promoting skin wound healing.
Collapse
Affiliation(s)
- Wei Wei
- Department of Dermatology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Key Laboratory of Skin Infection and Immunity, Wuhan No. 1 Hospital, Wuhan, 430022, China
| | - Zi-Lu Qu
- Department of Dermatology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Key Laboratory of Skin Infection and Immunity, Wuhan No. 1 Hospital, Wuhan, 430022, China
| | - Li Lei
- Department of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210000, China
| | - Ping Zhang
- Department of Dermatology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Provincial Key Laboratory of Skin Infection and Immunity, Wuhan No. 1 Hospital, Wuhan, 430022, China.
| |
Collapse
|
10
|
Rehak L, Giurato L, Monami M, Meloni M, Scatena A, Panunzi A, Manti GM, Caravaggi CMF, Uccioli L. The Immune-Centric Revolution Translated into Clinical Application: Peripheral Blood Mononuclear Cell (PBMNC) Therapy in Diabetic Patients with No-Option Critical Limb-Threatening Ischemia (NO-CLTI)-Rationale and Meta-Analysis of Observational Studies. J Clin Med 2024; 13:7230. [PMID: 39685690 DOI: 10.3390/jcm13237230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Chronic limb-threatening ischemia (CLTI), the most advanced form of peripheral arterial disease (PAD), is the comorbidity primarily responsible for major lower-limb amputations, particularly for diabetic patients. Autologous cell therapy has been the focus of efforts over the past 20 years to create non-interventional therapeutic options for no-option CLTI to improve limb perfusion and wound healing. Among the different available techniques, peripheral blood mononuclear cells (PBMNC) appear to be the most promising autologous cell therapy due to physio-pathological considerations and clinical evidence, which will be discussed in this review. A meta-analysis of six clinical studies, including 256 diabetic patients treated with naive, fresh PBMNC produced via a selective filtration point-of-care device, was conducted. PBMNC was associated with a mean yearly amputation rate of 15.7%, a mean healing rate of 62%, and a time to healing of 208.6 ± 136.5 days. Moreover, an increase in TcPO2 and a reduction in pain were observed. All-cause mortality, with a mean rate of 22.2% and a yearly mortality rate of 18.8%, was reported. No serious adverse events were reported. Finally, some practical and financial considerations are provided, which point to the therapy's recommendation as the first line of treatment for this particular and crucial patient group.
Collapse
Affiliation(s)
- Laura Rehak
- Athena Cell Therapy Technologies, 50126 Florence, Italy
| | - Laura Giurato
- Department of Biomedicine and Prevention, Diabetes-Endocrine Section CTO Hospital, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Matteo Monami
- Department of Diabetology Azienda Ospedaliera Universitaria Careggi, University of Florence, 50134 Florence, Italy
| | - Marco Meloni
- Diabetic Foot Unit, Department of Systems Medicine, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Alessia Scatena
- Diabetology Unit, San Donato Hospital Arezzo, Local Health Authorities Southeast Tuscany, 52100 Arezzo, Italy
| | - Andrea Panunzi
- Department of Biomedicine and Prevention, Diabetes-Endocrine Section CTO Hospital, Tor Vergata University of Rome, 00133 Rome, Italy
- PhD School of Applied Medical and Surgical Sciences, University of Rome Tor Vergata Italy, 00133 Rome, Italy
| | | | | | - Luigi Uccioli
- Department of Biomedicine and Prevention, Diabetes-Endocrine Section CTO Hospital, Tor Vergata University of Rome, 00133 Rome, Italy
| |
Collapse
|
11
|
Nazari M, Taremi S, Elahi R, Mostanadi P, Esmeilzadeh A. Therapeutic Properties of M2 Macrophages in Chronic Wounds: An Innovative Area of Biomaterial-Assisted M2 Macrophage Targeted Therapy. Stem Cell Rev Rep 2024:10.1007/s12015-024-10806-3. [PMID: 39556244 DOI: 10.1007/s12015-024-10806-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Wound healing is a dynamic, multi-stage process essential for restoring skin integrity. Dysregulated wound healing is often linked to impaired macrophage function, particularly in individuals with chronic underlying conditions. Macrophages, as key regulators of wound healing, exhibit significant phenotypic diversity, ranging from the pro-healing M2 phenotype to the pro-inflammatory M1 phenotype. Imbalances in the M1/M2 ratio or hyperactivation of the M1 phenotype can delay the normal healing. Consequently, strategies aimed at suppressing the M1 phenotype or promoting the shift of local skin macrophages toward the M2 phenotype can potentially treat chronic non-healing wounds. This manuscript provides an overview of macrophages' role in normal and pathological wound-healing processes. It examines various therapeutic approaches targeting M2 macrophages, such as ex vivo-activated macrophage therapy, immunopharmacological strategies, and biomaterial-directed macrophage polarization. However, it also highlights that M2 macrophage therapies and immunopharmacological interventions may have drawbacks, including rapid phenotypic changes, adverse effects on other skin cells, biotoxicity, and concerns related to biocompatibility, stability, and drug degradation. Therefore, there is a need for more targeted macrophage-based therapies that ensure optimal biosafety, allowing for effective reprogramming of dysregulated macrophages and improved therapeutic outcomes. Recent advances in nano-biomaterials have demonstrated promising regenerative potential compared to traditional treatments. This review discusses the progress of biomaterial-assisted macrophage targeting in chronic wound repair and addresses the challenges faced in its clinical application. Additionally, it explores novel design concepts for combinational therapies, such as incorporating regenerative particles like exosomes into dressing materials or encapsulating them in microneedling systems to enhance wound healing rates.
Collapse
Affiliation(s)
- Mahdis Nazari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Siavash Taremi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Elahi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Parsa Mostanadi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran.
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
12
|
Blaise O, Duchesne C, Capuzzo E, Nahori MA, Fernandes J, Connor MG, Hamon MA, Pizarro-Cerda J, Lataillade JJ, McGuckin C, Rousseau A, Banzet S, Dussurget O, Frescaline N. Infected wound repair correlates with collagen I induction and NOX2 activation by cold atmospheric plasma. NPJ Regen Med 2024; 9:28. [PMID: 39358383 PMCID: PMC11447178 DOI: 10.1038/s41536-024-00372-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Cold atmospheric plasma (CAP) is a promising complement to tissue repair and regenerative medicine approaches. CAP has therapeutic potential in infected cutaneous wounds by mechanisms which remain enigmatic. Here, CAP is shown to activate phagocyte NADPH oxidase complex NOX2. CAP induced increased intracellular reactive oxygen species, alleviated by NOX2 inhibitors. Genetic and pharmacological inhibitions of NOX2 in macrophages and bioengineered skin infected with Staphylococcus aureus and treated with CAP reduced intracellular oxidants and increased bacterial survival. CAP triggered Rac activation and phosphorylation of p40phox and p47phox required for NOX2 assembly and activity. Furthermore, CAP induced collagen I expression by fibroblasts. Infection and healing kinetics showed that murine skin wounds infected with S. aureus and treated with CAP are characterized by decreased bacterial burden, increased length of neoepidermis and extracellular matrix formation. Collectively, our findings identify mechanisms triggered by CAP that subdue infection and result in enhanced repair following skin injury.
Collapse
Affiliation(s)
- Océane Blaise
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France
- École Polytechnique, Sorbonne Université, CNRS UMR7648, Laboratoire de Physique des Plasmas, Palaiseau, France
| | - Constance Duchesne
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France
- École Polytechnique, Sorbonne Université, CNRS UMR7648, Laboratoire de Physique des Plasmas, Palaiseau, France
| | - Elena Capuzzo
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France
| | - Marie-Anne Nahori
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité des Toxines Bactériennes, Paris, France
| | | | - Michael G Connor
- Institut Pasteur, Université Paris Cité, Unité Chromatine et Infection, Paris, France
| | - Mélanie A Hamon
- Institut Pasteur, Université Paris Cité, Unité Chromatine et Infection, Paris, France
| | - Javier Pizarro-Cerda
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France
| | | | | | - Antoine Rousseau
- École Polytechnique, Sorbonne Université, CNRS UMR7648, Laboratoire de Physique des Plasmas, Palaiseau, France
| | - Sébastien Banzet
- Centre de Transfusion Sanguine des Armées, Clamart, France
- Institut de Recherche Biomédicale des Armées, INSERM UMRS-MD 1197, Brétigny-sur-Orge, France
| | - Olivier Dussurget
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France.
| | - Nadira Frescaline
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France.
- Centre de Transfusion Sanguine des Armées, Clamart, France.
| |
Collapse
|
13
|
Furgiuele S, Cappello E, Ruggeri M, Camilli D, Palasciano G, Guerrieri MW, Michelagnoli S, Dorrucci V, Pompeo F. One-Year Analysis of Autologous Peripheral Blood Mononuclear Cells as Adjuvant Therapy in Treatment of Diabetic Revascularizable Patients Affected by Chronic Limb-Threatening Ischemia: Real-World Data from Italian Registry ROTARI. J Clin Med 2024; 13:5275. [PMID: 39274487 PMCID: PMC11396002 DOI: 10.3390/jcm13175275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024] Open
Abstract
Wounds in diabetic patients with peripheral arterial disease (PAD) may be poorly responsive to revascularization and conventional therapies. Background/Objective: This study's objective is to analyze the results of regenerative cell therapy with peripheral blood mononuclear cells (PBMNCs) as an adjuvant to revascularization. Methods: This study is based on 168 patients treated with endovascular revascularization below the knee plus three PBMNC implants. The follow-up included clinical outcomes at 1-2-3-6 and 12 months based on amputations, wound healing, pain, and TcPO2. Results: The results at 1 year for 122 cases showed a limb rescue rate of 94.26%, a complete wound healing in 65.59% of patients, and an improvement in the wound area, significant pain relief, and increased peripheral oxygenation. In total, 64.51% of patients completely healed at 6 months, compared to the longer wound healing time reported in the literature in the same cohort of patients, suggesting that PBMNCs have an adjuvant effect in wound healing after revascularization. Conclusions: PBMNC regenerative therapy is a safe and promising treatment for diabetic PAD. In line with previous experiences, this registry shows improved healing in diabetic patients with below-the-knee arteriopathy. The findings support the use of this cell therapy and advocate for further research.
Collapse
Affiliation(s)
- Sergio Furgiuele
- Unit of Vascular and Endovascular Surgery, High Specialty Hospital "Mediterranea", 80122 Napoli, Italy
| | - Enrico Cappello
- Second Unit of Vascular and Endovascular Surgery, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Massimo Ruggeri
- Unit of Vascular Surgery, San Camillo de Lellis Hospital, 02100 Rieti, Italy
| | - Daniele Camilli
- Casa di Cura Santa Caterina della Rosa Asl RM 2, 00176 Roma, Italy
| | - Giancarlo Palasciano
- Vascular Surgery Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Massimiliano Walter Guerrieri
- Vascular Surgery Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
- UOC Vascular Surgery, San Donato Hospital, 52100 Arezzo, Italy
| | - Stefano Michelagnoli
- Vascular and Endovascular Surgery Unit, San Giovanni di Dio Hospital, 50143 Florence, Italy
| | - Vittorio Dorrucci
- Department of Vascular Surgery, Umberto I Hospital, 96100 Venice, Italy
| | - Francesco Pompeo
- Second Unit of Vascular and Endovascular Surgery, IRCCS Neuromed, 86077 Pozzilli, Italy
| |
Collapse
|
14
|
Zhang L, Huang J, Li L, Zhang H, Li S, Chai W, Chen X, Liu C, Honiball JR, Li B, Ren Y, Chu L, Luo X, Pan H, Cui X. Exploratory Investigation of Zinc-Modified Borosilicate Bioactive Glass: A New Methodology for Its Biocompatibility, Immunoregulation, and Pro-Angiogenic Property Evaluation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:46016-46034. [PMID: 39167416 DOI: 10.1021/acsami.4c08487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
The assessment of biodegradable materials, such as bioactive glass, under the existing ISO 10993 standard test methods poses a significant challenge due to potential cell viability impairment caused by the accumulation of degraded products in a static environment. Therefore, innovative methodologies are urgently needed to tailor the unique biodegradation characteristics of these materials, providing more precise and scientific insights into biosafety and efficacy verification. Motivation by its bidirectional regulation of angiogenesis and immunity, zinc (Zn) was incorporated into sol-gel-derived borosilicate bioactive glasses (SBSGs) to fabricate Zn-incorporated borosilicate bioactive glasses (SBSG-Zn) to complement the tissue repair capabilities of bioactive glasses. Both SBSG and SBSG-Zn glasses consist of nanosized particles, slit mesoporous pores, high specific surface areas, and bioreactivity. In vitro comparative analysis, conducted according to ISO 10993 standards, demonstrates that only at suitable dilution rates─such as the 8-fold dilution employed in this study─do extracts of SBSG and SBSG-Zn glasses exhibit low cytotoxicity when cultured with human umbilical vein endothelial cells (HUVECs). Notably, SBSG-Zn glasses show optimal promotion of angiogenic gene expression in HUVECs. Furthermore, within an appropriate concentration range of released ions, SBSG-Zn glass extracts not only promote cell survival but also modulate the expression of anti-inflammatory genes while simultaneously inhibiting pro-inflammatory genes concurrently. After being implanted in rat subcutaneous defect models, both SBSG and SBSG-Zn glasses demonstrated the local immunoregulation and angiogenic effects. SBSG-Zn stands out by demonstrating superior modulation of M1/M2 polarization in macrophages as validated by altered secretion of key factors in macrophages and expression of relevant growth factors in HUVECs. These findings underscore the potential for convenient manipulation of localized angiogenic and immunoregulation through the incorporation of zinc into bioactive glass, emphasizing the importance of ensuring the appropriate ion doses are applied for achieving optimal therapeutic efficiency.
Collapse
Affiliation(s)
- Liyan Zhang
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Jing Huang
- Department of Orthopedics, Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker's Hospital, Liuzhou 545000, Guangxi, P. R. China
| | - Li Li
- Department of Orthopedics, Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker's Hospital, Liuzhou 545000, Guangxi, P. R. China
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR 999077, P. R. China
| | - Hao Zhang
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - Shuaijie Li
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - Wenwen Chai
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - Xiaochen Chen
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - Chunyu Liu
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - John Robert Honiball
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR 999077, P. R. China
| | - Bing Li
- Department of Orthopedics, Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Worker's Hospital, Liuzhou 545000, Guangxi, P. R. China
| | - Youliang Ren
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing 400010, P. R. China
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Lei Chu
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing 400010, P. R. China
| | - Xuegang Luo
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P. R. China
| | - Haobo Pan
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| | - Xu Cui
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen 518055, Guangdong, P. R. China
| |
Collapse
|
15
|
Yang W, Cao M, Wang W, Diao N, Liu X, Hu Y, Wang X, Sun T, Guo C, Chen D. Multifunctional composite soluble microneedle patch based on "one stone, three birds" strategy for promoting the healing of infectious wounds. Colloids Surf B Biointerfaces 2024; 241:114049. [PMID: 38908043 DOI: 10.1016/j.colsurfb.2024.114049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
The colonisation of microorganisms such as bacteria forms a biofilm barrier on the wound's surface, preventing or delaying the penetration of antibacterial drugs. At the same time, continuous bacterial infection can cause oxidative stress and an inflammatory response and hinder angiogenesis, resulting in difficult wound healing. Based on the "one stone, three birds" strategy, a multi-functional nanoparticle composite soluble microneedle was designed and developed to solve this dilemma better. Ginsenoside-liposomes(R-Lipo) were prepared by ginsenoside Rg3, which had the effect of promoting repair, instead of cholesterol, and loaded with berberine (Ber), the antibacterial component of Coptis, together with polydopamine (PDA), which had anti-inflammatory and antioxidant properties, into microneedles based on hyaluronic acid (PDA/R-Lipo@BerMN). PDA/R-Lipo@BerMN tip can penetrate and destroy the integrity of the biofilm, dissolve under the action of hyaluronidase in the skin, and gradually release the drug to achieve rapid antibacterial, anti-inflammatory, antioxidant, and proliferation. As expected, the PDA/R-Lipo@BerMN patch effectively cleared ROS during wound closure, further promoted M2 macrophage polarisation, eradicated bacterial infection, and regulated the immune microenvironment, promoting inflammation suppression, collagen deposition, angiogenesis, and tissue regeneration.
Collapse
Affiliation(s)
- Weili Yang
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Min Cao
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Wenxin Wang
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Ningning Diao
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Xiaowei Liu
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Yue Hu
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Xinxin Wang
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Tianying Sun
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China
| | - Chunjing Guo
- College of Marine Life Science, Ocean University of China, 5# Yushan 10 Road, Qingdao 266003, PR China.
| | - Daquan Chen
- School of Pharmacy, Yantai University, 30# Qingquan Road, Yantai 264005, PR China.
| |
Collapse
|
16
|
Zuo S, Wang Y, Bao H, Zhang Z, Yang N, Jia M, Zhang Q, Jian A, Ji R, Zhang L, Lu Y, Huang Y, Shen P. Lipid synthesis, triggered by PPARγ T166 dephosphorylation, sustains reparative function of macrophages during tissue repair. Nat Commun 2024; 15:7269. [PMID: 39179603 PMCID: PMC11343878 DOI: 10.1038/s41467-024-51736-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
Macrophages may acquire a reparative phenotype that supports tissue repair and remodeling in response to tissue injury. However, the metabolic requirements underpinning this process are incompletely understood. Here, we show that posttranslational modification (PTM) of PPARγ regulates lipid synthesis in response to wound microenvironmental cues and that metabolic rewiring orchestrates function of reparative macrophages. In injured tissues, repair signaling leads to decreased macrophage PPARγ threonine 166 (T166) phosphorylation, which results in a partially active PPARγ transcriptional program comprised of increased binding activity to the regulator regions of lipid synthesis-associated genes, thereby increased lipogenesis. The accumulated lipids serve as signaling molecules, triggering STAT3-mediated growth factor expression, and supporting the synthesis of phospholipids for the expansion of the endoplasmic reticulum (ER), which is required for protein secretion. Genetic or pharmacological inhibition of PPARγ T166 phosphorylation promotes the reparative function of macrophages and facilitates tissue regeneration. In summary, our work identifies PPARγ T166-regulated lipid biosynthesis as an essential pathway for meeting the anabolic demands of the activation and function of macrophages and provides a rationale for potential therapeutic targeting of tissue repair.
Collapse
Affiliation(s)
- Shiman Zuo
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Yuxin Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Hanjing Bao
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Zehui Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Nanfei Yang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Meng Jia
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Qing Zhang
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Ani Jian
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Rong Ji
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China
| | - Lidan Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China
| | - Yan Lu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Yahong Huang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, 210023, China.
- Shenzhen Research Institute of Nanjing University, Shenzhen, 518000, China.
| |
Collapse
|
17
|
Zhu A, Baur C, Götz P, Elbs K, Lasch M, Faro A, Preissner KT, Deindl E. The Complement System Is Essential for Arteriogenesis by Enhancing Sterile Inflammation as a Relevant Step in Collateral Artery Growth. Cells 2024; 13:1405. [PMID: 39272977 PMCID: PMC11394660 DOI: 10.3390/cells13171405] [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: 07/25/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Arteriogenesis is an inflammatory driven mechanism, describing the growth of a natural bypass from pre-existing collateral arteries to compensate for an occluded artery. The complement system component C3 is a potent natural inflammatory activator. Here, we investigated its impact on the process of collateral artery growth using C3-deficient (C3 -/-) and wildtype control mice in a murine hindlimb model of arteriogenesis. Induction of arteriogenesis by unilateral femoral artery ligation resulted in decreased perfusion recovery in C3 -/- mice on day 7 as shown by Laser Doppler imaging. Immunofluorescence staining revealed a reduced vascular cell proliferation in C3 -/- mice. Gene expression analysis displayed a significant reduction in monocyte chemoattractant protein-1 (MCP-1) expression in C3 -/- mice. Interestingly, 3 days after induction of arteriogenesis, the number of macrophages (CD68+) recruited to growing collaterals was not affected by C3 deficiency. However, a significant reduction in inflammatory M1-like polarized macrophages (CD68+/MRC1-) was noted. Forced mast cell activation by Compound 48/80 as well as exogenous MCP-1 application rescued the number of M1-like polarized macrophages along with perfusion recovery in C3 -/- mice. In summary, this study demonstrates that complement C3 influences arteriogenesis by mediating MCP-1 expression, which is essential for the induction and enhancement of sterile inflammation.
Collapse
Affiliation(s)
- Amanda Zhu
- Institute of Surgical Research at the Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (A.Z.); (C.B.); (P.G.); (K.E.); (M.L.); (A.F.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Carolin Baur
- Institute of Surgical Research at the Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (A.Z.); (C.B.); (P.G.); (K.E.); (M.L.); (A.F.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Philipp Götz
- Institute of Surgical Research at the Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (A.Z.); (C.B.); (P.G.); (K.E.); (M.L.); (A.F.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Katharina Elbs
- Institute of Surgical Research at the Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (A.Z.); (C.B.); (P.G.); (K.E.); (M.L.); (A.F.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Manuel Lasch
- Institute of Surgical Research at the Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (A.Z.); (C.B.); (P.G.); (K.E.); (M.L.); (A.F.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Anna Faro
- Institute of Surgical Research at the Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (A.Z.); (C.B.); (P.G.); (K.E.); (M.L.); (A.F.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Klaus T. Preissner
- Department of Cardiology, Kerckhoff-Heart Research Institute, Faculty of Medicine, Justus-Liebig-University, 35392 Giessen, Germany;
| | - Elisabeth Deindl
- Institute of Surgical Research at the Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; (A.Z.); (C.B.); (P.G.); (K.E.); (M.L.); (A.F.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
18
|
Williamson AE, Liyanage S, Hassanshahi M, Dona MSI, Toledo-Flores D, Tran DXA, Dimasi C, Schwarz N, Fernando S, Salagaras T, Long A, Kazenwadel J, Harvey NL, Drummond GR, Vinh A, Chandrakanthan V, Misra A, Neufeld Z, Tan JTM, Martelotto L, Polo JM, Bonder CS, Pinto AR, Sharma S, Nicholls SJ, Bursill CA, Psaltis PJ. Discovery of an embryonically derived bipotent population of endothelial-macrophage progenitor cells in postnatal aorta. Nat Commun 2024; 15:7097. [PMID: 39154007 PMCID: PMC11330468 DOI: 10.1038/s41467-024-51637-7] [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: 12/20/2023] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
Converging evidence indicates that extra-embryonic yolk sac is the source of both macrophages and endothelial cells in adult mouse tissues. Prevailing views are that these embryonically derived cells are maintained after birth by proliferative self-renewal in their differentiated states. Here we identify clonogenic endothelial-macrophage (EndoMac) progenitor cells in the adventitia of embryonic and postnatal mouse aorta, that are independent of Flt3-mediated bone marrow hematopoiesis and derive from an early embryonic CX3CR1+ and CSF1R+ source. These bipotent progenitors are proliferative and vasculogenic, contributing to adventitial neovascularization and formation of perfused blood vessels after transfer into ischemic tissue. We establish a regulatory role for angiotensin II, which enhances their clonogenic and differentiation properties and rapidly stimulates their proliferative expansion in vivo. Our findings demonstrate that embryonically derived EndoMac progenitors participate in local vasculogenic responses in the aortic wall by contributing to the expansion of endothelial cells and macrophages postnatally.
Collapse
Affiliation(s)
- Anna E Williamson
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Sanuri Liyanage
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Mohammadhossein Hassanshahi
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Malathi S I Dona
- Cardiac Cellular Systems Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Deborah Toledo-Flores
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Dang X A Tran
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Catherine Dimasi
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Nisha Schwarz
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Sanuja Fernando
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Thalia Salagaras
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Aaron Long
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Department of Cardiology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Jan Kazenwadel
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Natasha L Harvey
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Grant R Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology and Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Antony Vinh
- Department of Microbiology, Anatomy, Physiology and Pharmacology and Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Vashe Chandrakanthan
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Ashish Misra
- Faculty of Medicine and Health, University of Sydney and Heart Research Institute, Newtown, NSW, Australia
| | - Zoltan Neufeld
- School of Mathematics and Physics, The University of Queensland, Brisbane, QLD, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Luciano Martelotto
- Adelaide Centre for Epigenetics and the South Australian Immunogenomics Cancer Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Jose M Polo
- Adelaide Centre for Epigenetics and the South Australian Immunogenomics Cancer Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Claudine S Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Alexander R Pinto
- Cardiac Cellular Systems Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology and Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Shiwani Sharma
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Flinders Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Stephen J Nicholls
- Monash Cardiovascular Research Centre, Monash University, Melbourne, VIC, Australia
| | - Christina A Bursill
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Peter J Psaltis
- Vascular Research Centre, Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.
- Department of Cardiology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia.
| |
Collapse
|
19
|
López-Cuevas P, Oates TCL, Tong Q, McGowan LM, Cross SJ, Xu C, Zhao Y, Yin Z, Toye AM, Boussahel A, Hammond CL, Mann S, Martin P. Reprogramming macrophages with R848-loaded artificial protocells to modulate skin and skeletal wound healing. J Cell Sci 2024; 137:jcs262202. [PMID: 39078119 PMCID: PMC11385641 DOI: 10.1242/jcs.262202] [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: 04/16/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
After tissue injury, inflammatory cells are rapidly recruited to the wound where they clear microbes and other debris, and coordinate the behaviour of other cell lineages at the repair site in both positive and negative ways. In this study, we take advantage of the translucency and genetic tractability of zebrafish to evaluate the feasibility of reprogramming innate immune cells in vivo with cargo-loaded protocells and investigate how this alters the inflammatory response in the context of skin and skeletal repair. Using live imaging, we show that protocells loaded with R848 cargo (which targets TLR7 and TLR8 signalling), are engulfed by macrophages resulting in their switching to a pro-inflammatory phenotype and altering their regulation of angiogenesis, collagen deposition and re-epithelialization during skin wound healing, as well as dampening osteoblast and osteoclast recruitment and bone mineralization during fracture repair. For infected skin wounds, R848-reprogrammed macrophages exhibited enhanced bactericidal activities leading to improved healing. We replicated our zebrafish studies in cultured human macrophages, and showed that R848-loaded protocells similarly reprogramme human cells, indicating how this strategy might be used to modulate wound inflammation in the clinic.
Collapse
Affiliation(s)
- Paco López-Cuevas
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Tiah C L Oates
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Qiao Tong
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Lucy M McGowan
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Stephen J Cross
- Wolfson Bioimaging Facility, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Can Xu
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Yu Zhao
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Zhuping Yin
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell Products, University of Bristol, Bristol BS34 7QH, UK
| | - Asme Boussahel
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Stephen Mann
- Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- Max Planck Bristol Centre for Minimal Biology, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
20
|
Peña OA, Martin P. Cellular and molecular mechanisms of skin wound healing. Nat Rev Mol Cell Biol 2024; 25:599-616. [PMID: 38528155 DOI: 10.1038/s41580-024-00715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/27/2024]
Abstract
Wound healing is a complex process that involves the coordinated actions of many different tissues and cell lineages. It requires tight orchestration of cell migration, proliferation, matrix deposition and remodelling, alongside inflammation and angiogenesis. Whereas small skin wounds heal in days, larger injuries resulting from trauma, acute illness or major surgery can take several weeks to heal, generally leaving behind a fibrotic scar that can impact tissue function. Development of therapeutics to prevent scarring and successfully repair chronic wounds requires a fuller knowledge of the cellular and molecular mechanisms driving wound healing. In this Review, we discuss the current understanding of the different phases of wound healing, from clot formation through re-epithelialization, angiogenesis and subsequent scar deposition. We highlight the contribution of different cell types to skin repair, with emphasis on how both innate and adaptive immune cells in the wound inflammatory response influence classically studied wound cell lineages, including keratinocytes, fibroblasts and endothelial cells, but also some of the less-studied cell lineages such as adipocytes, melanocytes and cutaneous nerves. Finally, we discuss newer approaches and research directions that have the potential to further our understanding of the mechanisms underpinning tissue repair.
Collapse
Affiliation(s)
- Oscar A Peña
- School of Biochemistry, University of Bristol, Bristol, UK.
| | - Paul Martin
- School of Biochemistry, University of Bristol, Bristol, UK.
| |
Collapse
|
21
|
Yan L, Wang J, Cai X, Liou Y, Shen H, Hao J, Huang C, Luo G, He W. Macrophage plasticity: signaling pathways, tissue repair, and regeneration. MedComm (Beijing) 2024; 5:e658. [PMID: 39092292 PMCID: PMC11292402 DOI: 10.1002/mco2.658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Macrophages are versatile immune cells with remarkable plasticity, enabling them to adapt to diverse tissue microenvironments and perform various functions. Traditionally categorized into classically activated (M1) and alternatively activated (M2) phenotypes, recent advances have revealed a spectrum of macrophage activation states that extend beyond this dichotomy. The complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications orchestrates macrophage polarization, allowing them to respond to various stimuli dynamically. Here, we provide a comprehensive overview of the signaling cascades governing macrophage plasticity, focusing on the roles of Toll-like receptors, signal transducer and activator of transcription proteins, nuclear receptors, and microRNAs. We also discuss the emerging concepts of macrophage metabolic reprogramming and trained immunity, contributing to their functional adaptability. Macrophage plasticity plays a pivotal role in tissue repair and regeneration, with macrophages coordinating inflammation, angiogenesis, and matrix remodeling to restore tissue homeostasis. By harnessing the potential of macrophage plasticity, novel therapeutic strategies targeting macrophage polarization could be developed for various diseases, including chronic wounds, fibrotic disorders, and inflammatory conditions. Ultimately, a deeper understanding of the molecular mechanisms underpinning macrophage plasticity will pave the way for innovative regenerative medicine and tissue engineering approaches.
Collapse
Affiliation(s)
- Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Jue Wang
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Xin Cai
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Han‐Ming Shen
- Faculty of Health SciencesUniversity of MacauMacauChina
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospitaland West China School of Basic Medical Sciences and Forensic MedicineSichuan University, and Collaborative Innovation Center for BiotherapyChengduChina
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| | - Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Damage Repair and RegenerationChongqingChina
| |
Collapse
|
22
|
Senk A, Fazzari J, Djonov V. Vascular mimicry in zebrafish fin regeneration: how macrophages build new blood vessels. Angiogenesis 2024; 27:397-410. [PMID: 38546923 PMCID: PMC11303510 DOI: 10.1007/s10456-024-09914-y] [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: 01/15/2024] [Accepted: 03/10/2024] [Indexed: 08/07/2024]
Abstract
Vascular mimicry has been thoroughly investigated in tumor angiogenesis. In this study, we demonstrate for the first time that a process closely resembling tumor vascular mimicry is present during physiological blood vessel formation in tissue regeneration using the zebrafish fin regeneration assay. At the fin-regenerating front, vasculature is formed by mosaic blood vessels with endothelial-like cells possessing the morphological phenotype of a macrophage and co-expressing both endothelial and macrophage markers within single cells. Our data demonstrate that the vascular segments of the regenerating tissue expand, in part, through the transformation of adjacent macrophages into endothelial-like cells, forming functional, perfused channels and contributing to the de novo formation of microvasculature. Inhibiting the formation of tubular vascular-like structures by CVM-1118 prevents vascular mimicry and network formation resulting in a 70% shorter regeneration area with 60% reduced vessel growth and a complete absence of any signs of regeneration in half of the fin area. Additionally, this is associated with a significant reduction in macrophages. Furthermore, depleting macrophages using macrophage inhibitor PLX-3397, results in impaired tissue regeneration and blood vessel formation, namely a reduction in the regeneration area and vessel network by 75% in comparison to controls.
Collapse
Affiliation(s)
- Anita Senk
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | | | - Valentin Djonov
- Institute of Anatomy, University of Bern, Bern, Switzerland.
| |
Collapse
|
23
|
Shah FH, Lee HW. Endothelial and macrophage interactions in the angiogenic niche. Cytokine Growth Factor Rev 2024; 78:64-76. [PMID: 39019663 DOI: 10.1016/j.cytogfr.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
The interactions between vascular cells, especially endothelial cells, and macrophages play a pivotal role in maintaining the subtle balance of vascular biology, which is crucial for angiogenesis in both healthy and diseased states. These cells are central to ensuring a harmonious balance between tissue repair and preventing excessive angiogenic activity, which could lead to pathological conditions. Recent advances in sophisticated genetic engineering vivo models and novel sequencing approaches, such as single-cell RNA-sequencing, in immunobiology have significantly enhanced our understanding of the gene expression and behavior of macrophages. These insights offer new perspectives on the role macrophages play not only in development but also across various health conditions. In this review, we explore the complex interactions between multiple types of macrophages and endothelium, focusing on their impact on new blood vessel formation. By understanding these intricate interactions, we aim to provide insights into new methods for managing angiogenesis in various diseases, thereby offering hope for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Fahad Hassan Shah
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| | - Heon-Woo Lee
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea.
| |
Collapse
|
24
|
Das A, Smith RJ, Andreadis ST. Harnessing the potential of monocytes/macrophages to regenerate tissue-engineered vascular grafts. Cardiovasc Res 2024; 120:839-854. [PMID: 38742656 PMCID: PMC11218695 DOI: 10.1093/cvr/cvae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024] Open
Abstract
Cell-free tissue-engineered vascular grafts provide a promising alternative to treat cardiovascular disease, but timely endothelialization is essential for ensuring patency and proper functioning post-implantation. Recent studies from our lab showed that blood cells like monocytes (MCs) and macrophages (Mϕ) may contribute directly to cellularization and regeneration of bioengineered arteries in small and large animal models. While MCs and Mϕ are leucocytes that are part of the innate immune response, they share common developmental origins with endothelial cells (ECs) and are known to play crucial roles during vessel formation (angiogenesis) and vessel repair after inflammation/injury. They are highly plastic cells that polarize into pro-inflammatory and anti-inflammatory phenotypes upon exposure to cytokines and differentiate into other cell types, including EC-like cells, in the presence of appropriate chemical and mechanical stimuli. This review focuses on the developmental origins of MCs and ECs; the role of MCs and Mϕ in vessel repair/regeneration during inflammation/injury; and the role of chemical signalling and mechanical forces in Mϕ inflammation that mediates vascular graft regeneration. We postulate that comprehensive understanding of these mechanisms will better inform the development of strategies to coax MCs/Mϕ into endothelializing the lumen and regenerate the smooth muscle layers of cell-free bioengineered arteries and veins that are designed to treat cardiovascular diseases and perhaps the native vasculature as well.
Collapse
Affiliation(s)
- Arundhati Das
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
| | - Randall J Smith
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
- Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, 701 Ellicott St, Buffalo, NY 14203, USA
- Cell, Gene and Tissue Engineering (CGTE) Center, University at Buffalo, The State University of New York, 813 Furnas Hall, Buffalo, NY 14260-4200, USA
| |
Collapse
|
25
|
Li C, Song Y, Meng X. The Role of Macrophages in Nerve Regeneration: Polarization and Combination with Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 38832865 DOI: 10.1089/ten.teb.2024.0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Peripheral nerve regeneration after trauma poses a substantial clinical challenge that has already been investigated for many years. Infiltration of immune cells is a critical step in the response to nerve damage that creates a supportive microenvironment for regeneration. In this work, we focus on a special type of immune cell, macrophage, in addressing the problem of neuronal regeneration. We discuss the complex endogenous mechanisms of peripheral nerve injury and regrowth vis-à-vis macrophages, including their recruitment, polarization, and interplay with Schwann cells post-trauma. Furthermore, we elucidate the underlying mechanisms by which exogenous stimuli govern the above events. Finally, we summarize the necessary roles of macrophages in peripheral nerve lesions and reconstruction. There are many challenges in controlling macrophage functions to achieve complete neuronal regeneration, even though considerable progress has been made in understanding the connection between these cells and peripheral nerve damage.
Collapse
Affiliation(s)
- Changqing Li
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuanyu Song
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xianyu Meng
- Department of Orthopedics, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
26
|
Wang Y, Gao Y, Shi H, Gao R, Yang J, Qu Y, Hu S, Zhang J, Wang J, Cao J, Zhang F, Ge J. CCL11 released by GSDMD-mediated macrophage pyroptosis regulates angiogenesis after hindlimb ischemia. Cell Death Discov 2024; 10:294. [PMID: 38906863 PMCID: PMC11192718 DOI: 10.1038/s41420-023-01764-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/02/2023] [Accepted: 12/06/2023] [Indexed: 06/23/2024] Open
Abstract
Peripheral vascular disease (PVD) is an emerging public health burden with a high rate of disability and mortality. Gasdermin D (GSDMD) has been reported to exert pyroptosis and play a critical role in the pathophysiology of many cardiovascular diseases. We ought to determine the role of GSDMD in the regulation of perfusion recovery after hindlimb ischemia (HLI). Our study revealed that GSDMD-mediated pyroptosis occurred in HLI. GSDMD deletion aggravated perfusion recovery and angiogenesis in vitro and in vivo. However, how GSDMD regulates angiogenesis after ischemic injury remains unclear. We then found that GSDMD-mediated pyroptosis exerted the angiogenic capacity in macrophages rather than endothelial cells after HLI. GSDMD deletion led to a lower level of CCL11 in mice serum. GSDMD knockdown in macrophages downregulated the expression and decreased the releasing level of CCL11. Furthermore, recombinant CCL11 improved endothelial functions and angiogenesis, which was attenuated by CCL11 antibody. Taken together, these results demonstrate that GSDMD promotes angiogenesis by releasing CCL11, thereby improving blood flow perfusion recovery after hindlimb ischemic injury. Therefore, CCL11 may be a novel target for prevention and treatment of vascular ischemic diseases.
Collapse
Affiliation(s)
- Yiwen Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Yang Gao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Huairui Shi
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Rifeng Gao
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, 200240, Shanghai, China
| | - Ji'e Yang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Ya'nan Qu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Shiyu Hu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Jian Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Jingpu Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Jiatian Cao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Feng Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, 200032, Shanghai, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, 200032, Shanghai, China.
- National Clinical Research Center for Interventional Medicine, 200032, Shanghai, China.
- Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
27
|
Varady SR, Greiner D, Roh-Johnson M. Macrophage subtypes inhibit breast cancer proliferation in culture. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.01.596963. [PMID: 38853881 PMCID: PMC11160732 DOI: 10.1101/2024.06.01.596963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Macrophages are a highly plastic cell type that adopt distinct subtypes and functional states depending on environmental cues. These functional states can vary wildly, with distinct macrophages capable of displaying opposing functions. We sought to understand how macrophage subtypes that exist on two ends of a spectrum influence the function of other cells. We used a co-culture system with primary human macrophages to probe the effects of macrophage subtypes on breast cancer cell proliferation. Our studies revealed a surprising phenotype in which both macrophage subtypes inhibited cancer cell proliferation compared to cancer cells alone. Of particular interest, using two different proliferation assays with two different breast cancer cell lines, we showed that differentiating macrophages into a "pro-tumor" subtype inhibited breast cancer cell proliferation. These findings are inconsistent with the prevailing interpretation that "pro-tumor" macrophages promote cancer cell proliferation and suggest a re-evaluation of how these interpretations are made.
Collapse
Affiliation(s)
- Sophia R.S. Varady
- Department of Biochemistry, University of Utah School of Medicine; Salt Lake City, UT, 84112, USA
| | - Daniel Greiner
- Department of Biochemistry, University of Utah School of Medicine; Salt Lake City, UT, 84112, USA
| | - Minna Roh-Johnson
- Department of Biochemistry, University of Utah School of Medicine; Salt Lake City, UT, 84112, USA
| |
Collapse
|
28
|
Strobel HA, Moss SM, Hoying JB. Isolated Fragments of Intact Microvessels: Tissue Vascularization, Modeling, and Therapeutics. Microcirculation 2024; 31:e12852. [PMID: 38619428 DOI: 10.1111/micc.12852] [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: 01/31/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/16/2024]
Abstract
The microvasculature is integral to nearly every tissue in the body, providing not only perfusion to and from the tissue, but also homing sites for immune cells, cellular niches for tissue dynamics, and cooperative interactions with other tissue elements. As a microtissue itself, the microvasculature is a composite of multiple cell types exquisitely organized into structures (individual vessel segments and extensive vessel networks) capable of considerable dynamics and plasticity. Consequently, it has been challenging to include a functional microvasculature in assembled or fabricated tissues. Isolated fragments of intact microvessels, which retain the cellular composition and structures of native microvessels, are proving effective in a variety of vascularization applications including tissue in vitro disease modeling, vascular biology, mechanistic discovery, and tissue prevascularization in regenerative therapeutics and grafting. In this review, we will discuss the importance of recapitulating native tissue biology and the successful vascularization applications of isolated microvessels.
Collapse
Affiliation(s)
| | - Sarah M Moss
- Advanced Solutions Life Sciences, Manchester, USA
| | | |
Collapse
|
29
|
Peterson EA, Sun J, Chen X, Wang J. Neutrophils facilitate the epicardial regenerative response after zebrafish heart injury. Dev Biol 2024; 508:93-106. [PMID: 38286185 PMCID: PMC10923159 DOI: 10.1016/j.ydbio.2024.01.011] [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: 07/29/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
Despite extensive studies on endogenous heart regeneration within the past 20 years, the players involved in initiating early regeneration events are far from clear. Here, we assessed the function of neutrophils, the first-responder cells to tissue damage, during zebrafish heart regeneration. We detected rapid neutrophil mobilization to the injury site after ventricular amputation, peaking at 1-day post-amputation (dpa) and resolving by 3 dpa. Further analyses indicated neutrophil mobilization coincides with peak epicardial cell proliferation, and recruited neutrophils associated with activated, expanding epicardial cells at 1 dpa. Neutrophil depletion inhibited myocardial regeneration and significantly reduced epicardial cell expansion, proliferation, and activation. To explore the molecular mechanism of neutrophils on the epicardial regenerative response, we performed scRNA-seq analysis of 1 dpa neutrophils and identified enrichment of the FGF and MAPK/ERK signaling pathways. Pharmacological inhibition of FGF signaling indicated its' requirement for epicardial expansion, while neutrophil depletion blocked MAPK/ERK signaling activation in epicardial cells. Ligand-receptor analysis indicated the EGF ligand, hbegfa, is released from neutrophils and synergizes with other FGF and MAPK/ERK factors for induction of epicardial regeneration. Altogether, our studies revealed that neutrophils quickly motivate epicardial cells, which later accumulate at the injury site and contribute to heart regeneration.
Collapse
Affiliation(s)
- Elizabeth A Peterson
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Jisheng Sun
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Xin Chen
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Jinhu Wang
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
30
|
Wang M, Chen S, He X, Yuan Y, Wei X. Targeting inflammation as cancer therapy. J Hematol Oncol 2024; 17:13. [PMID: 38520006 PMCID: PMC10960486 DOI: 10.1186/s13045-024-01528-7] [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: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 03/25/2024] Open
Abstract
Inflammation has accompanied human beings since the emergence of wounds and infections. In the past decades, numerous efforts have been undertaken to explore the potential role of inflammation in cancer, from tumor development, invasion, and metastasis to the resistance of tumors to treatment. Inflammation-targeted agents not only demonstrate the potential to suppress cancer development, but also to improve the efficacy of other therapeutic modalities. In this review, we describe the highly dynamic and complex inflammatory tumor microenvironment, with discussion on key inflammation mediators in cancer including inflammatory cells, inflammatory cytokines, and their downstream intracellular pathways. In addition, we especially address the role of inflammation in cancer development and highlight the action mechanisms of inflammation-targeted therapies in antitumor response. Finally, we summarize the results from both preclinical and clinical studies up to date to illustrate the translation potential of inflammation-targeted therapies.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Siyuan Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
31
|
Wang H, Yin B, Sun W, Geng H, Wang M, Li Y, Sun H, Yang X, Ni S. CO 2-Mediated Alkali-Neutralization Curdlan Hydrogels for Potential Wound Healing Application. Biomacromolecules 2024; 25:1738-1748. [PMID: 38340076 DOI: 10.1021/acs.biomac.3c01233] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Physical hydrogels of natural polysaccharides are considered as ideal candidates for wound dressing due to their natural biological activity and no harmful cross-linking agents. However, it remains a challenge to fabricate such hydrogel dressings in a facile and low-cost way. Herein, we reported an easy and cost-effective method to construct CO2-mediated alkali-neutralization Curdlan (CR) hydrogels without using an external cross-linking agent. Two types of hydrogels (denoted as CR-NaOH and CR-Na3PO4, respectively) were fabricated by dissolving CR powders in a NaOH or Na3PO4 aqueous solution, followed by keeping the CR alkaline solutions in air. The obtained pure CR hydrogels possessed a tunable porous structure with walls containing different forms of nanofibrils. These hydrogels exhibited much higher gel strength by comparison with the gels prepared by conventional heating treatment. They were flexible, stretchable, twistable, and conformable to arbitrarily curved skins. Moreover, they exhibited ideal swellability, proper degradability, and water vapor transmission rate, and their physicochemical properties were closely related to CR concentration in the alkaline solution. These two hydrogels also supported the growth of L929 cells. Importantly, studies on wound healing revealed that both 3CR-NaOH and 3CR-Na3PO4 hydrogels were capable of accelerating the wound healing process through recruiting more macrophages/fibroblasts, inducing more collagen deposition and neovascularization (α-SMA and CD31) without carrying any exogenous bioactive components. In conclusion, the present work not only reported promising materials for application in wound therapy but also offered a facile and safe manufacturing procedure for generating pure CR physical hydrogels with better performance.
Collapse
Affiliation(s)
- Haiyan Wang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P.R. China
| | - Bohao Yin
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 YiShan Road, Shanghai 200233, P. R. China
| | - Wenjun Sun
- Shanghai Xuhui District Dental Center, Shanghai 200030, P. R. China
| | - Huanna Geng
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P.R. China
| | - Mingyue Wang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P.R. China
| | - Yahui Li
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P.R. China
| | - Hui Sun
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 YiShan Road, Shanghai 200233, P. R. China
| | - Xuexia Yang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P.R. China
| | - Siyu Ni
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P.R. China
| |
Collapse
|
32
|
Chan HL, Zhang XHF. Nutrient Sensing in Macrophages Linked to Reorganized Tumor Vasculature. Cancer Res 2024; 84:650-651. [PMID: 38241708 DOI: 10.1158/0008-5472.can-24-0201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/21/2024]
Abstract
Macrophages are plastic immune cells that have varying functions dependent on stimulation from their environment. In a recent issue of Immunity, Do and colleagues demonstrated that activating mechanistic target of rapamycin complex 1 signaling in tumor macrophages alters their metabolism, localization, and function. Specifically, these tumor macrophages promote vascular remodeling that develops a hypoxic environment toxic to cancer cells. This culminates in a tangible reduction in tumor burden in a murine model of breast cancer. Their findings reveal a unique strategy to promote vascular remodeling through macrophage polarization and thereby highlight the intimate connections between macrophage metabolism and function. Additionally, their model highlights parallels between tumor progression and wound healing contexts while emphasizing the amplified effect of small perturbations to a tumor ecosystem.
Collapse
Affiliation(s)
- Hilda L Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Baylor College of Medicine Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas
- The Robert and Janice McNair Foundation MD/PhD Scholars, Baylor College of Medicine, Houston, Texas
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- McNair Medical Institute, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
33
|
Shiraki A, Tsuboi K, Wakabayashi T, Shiraki N, Nishida K. Reperfusion of retinal nonperfusion by neovascular-vascular anastomosis in proliferative diabetic retinopathy. Eur J Ophthalmol 2024; 34:NP28-NP32. [PMID: 37915118 DOI: 10.1177/11206721231210896] [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] [Indexed: 11/03/2023]
Abstract
BACKGROUND Retinal nonperfusion is a significant cause of vision loss in patients with proliferative diabetic retinopathy (PDR). Therefore, reperfusion of a nonperfusion has been a matter of strong interest, but few previous studies have demonstrated the potential benefits of reperfusion. CASE REPORTS Here, we report longitudinal optical coherence tomography angiographic analysis of two cases of PDR, in which the retinal neovascularization (RNV) that developed in response to retinal ischemia formed anastomoses with pre-existing physiological retinal vessels, resulting in both superficial and deep capillary reperfusion within the nonperfusion. We named this interesting finding "neovascular-vascular anastomosis." Retinal reperfusion due to neovascular-vascular anastomosis differed from recanalization, defined as reperfusion of once-occluded blood vessels, and has not been reported previously. CONCLUSION Our observation highlights the potential of RNV to rescue retinal ischemia by the formation of neovascular-vascular anastomoses.
Collapse
Affiliation(s)
- Akihiko Shiraki
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
| | - Kotaro Tsuboi
- Casey Eye Institute, Oregon Health and Science University, Portland, USA
- Department of Ophthalmology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Taku Wakabayashi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
- Wills Eye Hospital, Mid Atlantic Retina, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Nobuhiko Shiraki
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Suita, Japan
| |
Collapse
|
34
|
Simkin J, Aloysius A, Adam M, Safaee F, Donahue RR, Biswas S, Lakhani Z, Gensel JC, Thybert D, Potter S, Seifert AW. Tissue-resident macrophages specifically express Lactotransferrin and Vegfc during ear pinna regeneration in spiny mice. Dev Cell 2024; 59:496-516.e6. [PMID: 38228141 PMCID: PMC10922778 DOI: 10.1016/j.devcel.2023.12.017] [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: 01/17/2022] [Revised: 05/30/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024]
Abstract
The details of how macrophages control different healing trajectories (regeneration vs. scar formation) remain poorly defined. Spiny mice (Acomys spp.) can regenerate external ear pinnae tissue, whereas lab mice (Mus musculus) form scar tissue in response to an identical injury. Here, we used this dual species system to dissect macrophage phenotypes between healing modes. We identified secreted factors from activated Acomys macrophages that induce a pro-regenerative phenotype in fibroblasts from both species. Transcriptional profiling of Acomys macrophages and subsequent in vitro tests identified VEGFC, PDGFA, and Lactotransferrin (LTF) as potential pro-regenerative modulators. Examining macrophages in vivo, we found that Acomys-resident macrophages secreted VEGFC and LTF, whereas Mus macrophages do not. Lastly, we demonstrate the requirement for VEGFC during regeneration and find that interrupting lymphangiogenesis delays blastema and new tissue formation. Together, our results demonstrate that cell-autonomous mechanisms govern how macrophages react to the same stimuli to differentially produce factors that facilitate regeneration.
Collapse
Affiliation(s)
- Jennifer Simkin
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA; Department of Orthopaedic Surgery, LSU Health-New Orleans, New Orleans, LA 70112, USA.
| | - Ajoy Aloysius
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Mike Adam
- Department of Pediatrics, University of Cincinnati Children's Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Fatemeh Safaee
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Renée R Donahue
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Shishir Biswas
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Zohaib Lakhani
- Department of Orthopaedic Surgery, LSU Health-New Orleans, New Orleans, LA 70112, USA
| | - John C Gensel
- Department of Physiology, University of Kentucky, Lexington, KY 40506, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40506, USA
| | - David Thybert
- European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Steven Potter
- Department of Pediatrics, University of Cincinnati Children's Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
35
|
Xu L, Wang X, Wu Y, Zhang Z, Li X, Zhang J. Effectiveness of APG and Honey Gauze in Pressure Injury of Elderly: A Randomized Control Trial. INT J LOW EXTR WOUND 2024:15347346241234420. [PMID: 38403980 DOI: 10.1177/15347346241234420] [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: 02/27/2024]
Abstract
This study was designed to evaluate the efficiency of the combination of autologous platelet-rich plasma gel (APG) and Manuka honey gauze in the treatment of Stages 3-4 pressure injury of older adults. Patients were divided into four groups: Manuka honey gauze and APG (M + A), Manuka honey gauze (M), APG (A), and a control group (C). Different treatments were given, then wound bed coverage with granulation tissue, wound size reduction, and Pressure Ulcer Scale for Healing (PUSH) score were examined. Paraffin-embedded sections of wound tissues were analyzed and wound swab cultures were assessed. Kruskal-Wallis test and Mann-Whitney U test were performed in statistical analysis at a 5% significance level. A total of 42 patients were accepted. Significant increase of wound bed coverage with granulation tissue (51.24%, P = .004, Kruskal-Wallis test) and decrease of PUSH score (-5) were observed in the M + A group at the end of the observation (P = .032, Mann-Whitney U test). The hematoxylin-eosin staining of wound tissues showed that typical squamous epithelium was seen in wound bed of patient in M + A group. Manuka honey gauze and APG were proved to be superior treatments for pressure injury of old patient. Increase of granulation tissue coverage, reduction of PUSH score, and improved growth of epithelium were observed in M + A group. There was no side-effect, and the treatment would not cause infection.
Collapse
Affiliation(s)
- Lulu Xu
- Department of Geriatrics, Chongqing Clinical Research Center for Geriatric Diseases, Chongqing General Hospital, Chongqing, China
| | - Xinmeng Wang
- Chinese Academy of Sciences, Chongqing Medical University & Chongqing Institute of Green and Intelligent Technology, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, China
| | - Yongmei Wu
- Department of Geriatrics, Chongqing Clinical Research Center for Geriatric Diseases, Chongqing General Hospital, Chongqing, China
| | - Zhen Zhang
- Department of Clinical Laboratory, Chongqing General Hospital, Chongqing, China
| | - Xiafei Li
- Department of Geriatrics, Chongqing Clinical Research Center for Geriatric Diseases, Chongqing General Hospital, Chongqing, China
| | - Jie Zhang
- Department of Geriatrics, Chongqing Clinical Research Center for Geriatric Diseases, Chongqing General Hospital, Chongqing, China
- Graduate School, Chongqing Medical University, Chongqing, China
| |
Collapse
|
36
|
Azubuike-Osu SO, Kuhs A, Götz P, Faro A, Preissner KT, Arnholdt C, Deindl E. Treatment with Cobra Venom Factor Decreases Ischemic Tissue Damage in Mice. Biomedicines 2024; 12:309. [PMID: 38397911 PMCID: PMC10886846 DOI: 10.3390/biomedicines12020309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Tissue ischemia, caused by the blockage of blood vessels, can result in substantial damage and impaired tissue performance. Information regarding the functional contribution of the complement system in the context of ischemia and angiogenesis is lacking. To investigate the influence of complement activation and depletion upon femoral artery ligation (FAL), Cobra venom factor (CVF) (that functionally resembles C3b, the activated form of complement component C3) was applied in mice in comparison to control mice. Seven days after induction of muscle ischemia through FAL, gastrocnemius muscles of mice were excised and subjected to (immuno-)histological analyses. H&E and apoptotic cell staining (TUNEL) staining revealed a significant reduction in ischemic tissue damage in CVF-treated mice compared to controls. The control mice, however, exhibited a significantly higher capillary-to-muscle fiber ratio and a higher number of proliferating endothelial cells (CD31+/CD45-/BrdU+). The total number of leukocytes (CD45+) substantially decreased in CVF-treated mice versus control mice. Moreover, the CVF-treated group displayed a shift towards the M2-like anti-inflammatory and regenerative macrophage phenotype (CD68+/MRC1+). In conclusion, our findings suggest that treatment with CVF leads to reduced ischemic tissue damage along with decreased leukocyte recruitment but increased numbers of M2-like polarized macrophages, thereby enhancing tissue regeneration, repair, and healing.
Collapse
Affiliation(s)
- Sharon O. Azubuike-Osu
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany or (S.O.A.-O.); (A.K.); (P.G.); (A.F.); (C.A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University Ndufu Alike, Abakaliki 482131, Ebonyi, Nigeria
| | - Amelie Kuhs
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany or (S.O.A.-O.); (A.K.); (P.G.); (A.F.); (C.A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Philipp Götz
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany or (S.O.A.-O.); (A.K.); (P.G.); (A.F.); (C.A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Anna Faro
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany or (S.O.A.-O.); (A.K.); (P.G.); (A.F.); (C.A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Klaus T. Preissner
- Department of Cardiology, Kerckhoff-Heart Research Institute, Faculty of Medicine, Justus Liebig University, 35392 Giessen, Germany;
| | - Christoph Arnholdt
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany or (S.O.A.-O.); (A.K.); (P.G.); (A.F.); (C.A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Elisabeth Deindl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany or (S.O.A.-O.); (A.K.); (P.G.); (A.F.); (C.A.)
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
37
|
Suh SB, Suh JY, Cho SB. Analyzing secretory proteins in human dermal fibroblast-conditioned medium for angiogenesis: A bioinformatic approach. Skin Res Technol 2024; 30:e13568. [PMID: 38200622 PMCID: PMC10781896 DOI: 10.1111/srt.13568] [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: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND The conditioned medium from human dermal fibroblasts (dermal fibroblast-conditioned medium; DFCM) contains a diverse array of secretory proteins, including growth factors and wound repair-promoting proteins. Angiogenesis, a crucial process that facilitates the infiltration of inflammatory cells during wound repair, is induced by a hypoxic environment and inflammatory cytokines. METHODS In this study, we conducted a comprehensive bioinformatic analysis of 337 proteins identified through proteomics analysis of DFCM. We specifically focused on 64 DFCM proteins with potential involvement in angiogenesis. These proteins were further classified based on their characteristics, and we conducted a detailed analysis of their protein-protein interactions. RESULTS Gene Ontology protein classification categorized these 64 DFCM proteins into various classes, including metabolite interconversion enzymes (N = 11), protein modifying enzymes (N = 10), protein-binding activity modulators (N = 9), cell adhesion molecules (N = 6), extracellular matrix proteins (N = 6), transfer/carrier proteins (N = 3), calcium-binding proteins (N = 2), chaperones (N = 2), cytoskeletal proteins (N = 2), RNA metabolism proteins (N = 1), intercellular signal molecules (N = 1), transporters (N = 1), scaffold/adaptor proteins (N = 1), and unclassified proteins (N = 9). Furthermore, our protein-protein interaction network analysis of DFCM proteins revealed two distinct networks: one with medium confidence level interaction scores, consisting of 60 proteins with significant connections, and another at a high confidence level, comprising 52 proteins with significant interactions. CONCLUSIONS Our bioinformatic analysis highlights the presence of a multitude of secretory proteins in DFCM that form significant protein-protein interaction networks crucial for regulating angiogenesis. These findings underscore the critical roles played by DFCM proteins in various stages of angiogenesis during the wound repair process.
Collapse
Affiliation(s)
| | | | - Sung Bin Cho
- Yonsei Seran Dermatology and Laser ClinicSeoulSouth Korea
| |
Collapse
|
38
|
Pereira M, Pinto J, Arteaga B, Guerra A, Jorge RN, Monteiro FJ, Salgado CL. A Comprehensive Look at In Vitro Angiogenesis Image Analysis Software. Int J Mol Sci 2023; 24:17625. [PMID: 38139453 PMCID: PMC10743557 DOI: 10.3390/ijms242417625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
One of the complex challenges faced presently by tissue engineering (TE) is the development of vascularized constructs that accurately mimic the extracellular matrix (ECM) of native tissue in which they are inserted to promote vessel growth and, consequently, wound healing and tissue regeneration. TE technique is characterized by several stages, starting from the choice of cell culture and the more appropriate scaffold material that can adequately support and supply them with the necessary biological cues for microvessel development. The next step is to analyze the attained microvasculature, which is reliant on the available labeling and microscopy techniques to visualize the network, as well as metrics employed to characterize it. These are usually attained with the use of software, which has been cited in several works, although no clear standard procedure has been observed to promote the reproduction of the cell response analysis. The present review analyzes not only the various steps previously described in terms of the current standards for evaluation, but also surveys some of the available metrics and software used to quantify networks, along with the detection of analysis limitations and future improvements that could lead to considerable progress for angiogenesis evaluation and application in TE research.
Collapse
Affiliation(s)
- Mariana Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Jéssica Pinto
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Belén Arteaga
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Faculty of Medicine, University of Granada, Parque Tecnológico de la Salud, Av. de la Investigación 11, 18016 Granada, Spain
| | - Ana Guerra
- INEGI—Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, 4200-465 Porto, Portugal; (A.G.); (R.N.J.)
| | - Renato Natal Jorge
- INEGI—Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, 4200-465 Porto, Portugal; (A.G.); (R.N.J.)
- LAETA—Laboratório Associado de Energia, Transportes e Aeronáutica, Universidade do Porto, 4200-165 Porto, Portugal
- FEUP—Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e de Materiais, Universidade do Porto, 4200-165 Porto, Portugal
| | - Fernando Jorge Monteiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- FEUP—Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e de Materiais, Universidade do Porto, 4200-165 Porto, Portugal
- PCCC—Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
| | - Christiane Laranjo Salgado
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (M.P.); (J.P.); (B.A.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
39
|
Zhang J, Hu C, Zhang R, Xu J, Zhang Y, Yuan L, Zhang S, Pan S, Cao M, Qin J, Cheng X, Xu Z. The role of macrophages in gastric cancer. Front Immunol 2023; 14:1282176. [PMID: 38143746 PMCID: PMC10746385 DOI: 10.3389/fimmu.2023.1282176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/24/2023] [Indexed: 12/26/2023] Open
Abstract
As one of the deadliest cancers of the gastrointestinal tract, there has been limited improvement in long-term survival rates for gastric cancer (GC) in recent decades. The poor prognosis is attributed to difficulties in early detection, minimal opportunity for radical resection and resistance to chemotherapy and radiation. Macrophages are among the most abundant infiltrating immune cells in the GC stroma. These cells engage in crosstalk with cancer cells, adipocytes and other stromal cells to regulate metabolic, inflammatory and immune status, generating an immunosuppressive tumour microenvironment (TME) and ultimately promoting tumour initiation and progression. In this review, we summarise recent advances in our understanding of the origin of macrophages and their types and polarisation in cancer and provide an overview of the role of macrophages in GC carcinogenesis and development and their interaction with the GC immune microenvironment and flora. In addition, we explore the role of macrophages in preclinical and clinical trials on drug resistance and in treatment of GC to assess their potential therapeutic value in this disease.
Collapse
Affiliation(s)
- Jiaqing Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Can Hu
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ruolan Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Jingli Xu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yanqiang Zhang
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Li Yuan
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shengjie Zhang
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Siwei Pan
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Mengxuan Cao
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiangjiang Qin
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
40
|
Chen X, Zhang L, Chai W, Tian P, Kim J, Ding J, Zhang H, Liu C, Wang D, Cui X, Pan H. Hypoxic Microenvironment Reconstruction with Synergistic Biofunctional Ions Promotes Diabetic Wound Healing. Adv Healthc Mater 2023; 12:e2301984. [PMID: 37740829 DOI: 10.1002/adhm.202301984] [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: 06/22/2023] [Revised: 09/14/2023] [Indexed: 09/25/2023]
Abstract
Chronic hypoxia and ischemia make diabetic wounds non-healing. Cellular functions of diabetic chronic wounds are inhibited under a pathological environment. Therefore, this work develops a composite hydrogel system to promote diabetic wound healing. The composite hydrogel system consists of ε-poly-lysine (EPL), calcium peroxide (CP), and borosilicate glass (BG). The hydrogel supplies continuous dissolved oxygen molecules to the wound that can penetrate the skin tissue to restore normal cellular function and promote vascular regeneration. Biofunctional ions released from BGs can recruit more macrophages through neovascularization and modulate macrophage phenotypic transformation. Combining oxygen-mediated vascular regeneration and ion-mediated inflammatory regulation significantly accelerated diabetic wound healing. These findings indicate that this composite hydrogel system holds promise as a novel tissue engineering material.
Collapse
Affiliation(s)
- Xiaochen Chen
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Liyan Zhang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Wenwen Chai
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Pengfei Tian
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Jua Kim
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Jingxin Ding
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Hao Zhang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Chunyu Liu
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Deping Wang
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Xu Cui
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd, Shenzhen, 518071, P. R. China
| |
Collapse
|
41
|
Shi Z, Yao C, Shui Y, Li S, Yan H. Research progress on the mechanism of angiogenesis in wound repair and regeneration. Front Physiol 2023; 14:1284981. [PMID: 38089479 PMCID: PMC10711283 DOI: 10.3389/fphys.2023.1284981] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/13/2023] [Indexed: 12/10/2024] Open
Abstract
Poor wound healing and pathological healing have been pressing issues in recent years, as they impact human quality of life and pose risks of long-term complications. The study of neovascularization has emerged as a prominent research focus to address these problems. During the process of repair and regeneration, the establishment of a new vascular system is an indispensable stage for complete healing. It provides favorable conditions for nutrient delivery, oxygen supply, and creates an inflammatory environment. Moreover, it is a key manifestation of the proliferative phase of wound healing, bridging the inflammatory and remodeling phases. These three stages are closely interconnected and inseparable. This paper comprehensively integrates the regulatory mechanisms of new blood vessel formation in wound healing, focusing on the proliferation and migration of endothelial cells and the release of angiogenesis-related factors under different healing outcomes. Additionally, the hidden link between the inflammatory environment and angiogenesis in wound healing is explored.
Collapse
Affiliation(s)
- Zhuojun Shi
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Chong Yao
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Yujie Shui
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Site Li
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Hong Yan
- Laboratory of Plastic Surgery, Department of Plastic Surgery and Reconstruction, Second Hospital of West China, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Xu Y, Jia Y, Wu N, Wang J, He L, Yang D. CD93 Ameliorates Diabetic Wounds by Promoting Angiogenesis via the p38MAPK/MK2/HSP27 Axis. Eur J Vasc Endovasc Surg 2023; 66:707-721. [PMID: 37295599 DOI: 10.1016/j.ejvs.2023.06.001] [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: 01/05/2023] [Revised: 04/04/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
OBJECTIVE Diabetic wounds are a complication of diabetes mellitus, which is characterised by microcirculation dysfunction caused by decreased local blood supply and insufficient metabolic exchange. Clinically, in addition to glycaemic control, the most important treatment for diabetic wounds is to promote local angiogenesis, which accelerates wound healing. The authors previous study demonstrated that CD93, which is specifically expressed on vascular endothelial cells (ECs), redundantly regulates angiogenesis in zebrafish, suggesting that CD93 is a potential angiogenic molecule. However, the role of CD93 in diabetic wounds has not yet been elucidated. METHODS The angiogenic effects of CD93 were studied from four aspects: exogenous, endogenous, in vitro, and in vivo. CD93 recombinant protein was used in microvascular ECs and in mice to observe angiogenesis in vitro and in vivo. The wound model was established in CD93-/- and wild type diabetic mice, and the degree of wound healing as well as the amount and maturity of neovascularisation were investigated. The possible mechanism of CD93 in angiogenesis was determined by CD93 overexpression in cultured ECs. RESULTS CD93 recombinant protein was found to exogenously promote tube formation and sprouting of ECs. It also recruited cells to promote the formation of vascular like structures in subcutaneous tissue and accelerated wound healing by optimising angiogenesis and re-epithelisation. Furthermore, CD93 deficiency was observed to delay wound repair, characterised by reduced neovascularisation, vascular maturity, and re-epithelisation level. Mechanically, CD93 activated the p38MAPK/MK2/HSP27 signalling pathway, positively affecting the angiogenic functions of ECs. CONCLUSION This study demonstrated that CD93 promotes angiogenesis both in vitro and in vivo and that its angiogenic role in vitro is mediated by the p38MAPK/MK2/HSP27 signalling pathway. It was also found that CD93 exerts beneficial effects on wound healing in diabetic mice by promoting angiogenesis and re-epithelisation.
Collapse
Affiliation(s)
- Yuan Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yuhuan Jia
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Na Wu
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Wang
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Liwen He
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Deqin Yang
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
43
|
Oates TCL, Moura PL, Cross S, Roberts K, Baum HE, Haydn‐Smith KL, Wilson MC, Heesom KJ, Severn CE, Toye AM. Defining the proteomic landscape of cultured macrophages and their polarization continuum. Immunol Cell Biol 2023; 101:947-963. [PMID: 37694300 PMCID: PMC10953363 DOI: 10.1111/imcb.12687] [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: 05/10/2023] [Revised: 07/28/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023]
Abstract
Macrophages have previously been characterized based on phenotypical and functional differences into suggested simplified subtypes of MØ, M1, M2a and M2c. These macrophage subtypes can be generated in a well-established primary monocyte culture model that produces cells expressing accepted subtype surface markers. To determine how these subtypes retain functional similarities and better understand their formation, we generated all four subtypes from the same donors. Comparative whole-cell proteomics confirmed that four distinct macrophage subtypes could be induced from the same donor material, with > 50% of 5435 identified proteins being significantly altered in abundance between subtypes. Functional assessment highlighted that these distinct protein expression profiles are primed to enable specific cell functions, indicating that this shifting proteome is predictive of meaningful changes in cell characteristics. Importantly, the 2552 proteins remained consistent in abundance across all macrophage subtypes examined, demonstrating maintenance of a stable core proteome that likely enables swift polarity changes. We next explored the cross-polarization capabilities of preactivated M1 macrophages treated with dexamethasone. Importantly, these treated cells undergo a partial repolarization toward the M2c surface markers but still retain the M1 functional phenotype. Our investigation of polarized macrophage subtypes therefore provides evidence of a sliding scale of macrophage functionality, with these data sets providing a valuable benchmark resource for further studies of macrophage polarity, with relevance for cell therapy development and drug discovery.
Collapse
Affiliation(s)
- Tiah CL Oates
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| | - Pedro L Moura
- Center for Haematology and Regenerative Medicine, Department of Medicine (MedH)Karolinska InstitutetHuddingeSweden
| | | | - Kiren Roberts
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | - Holly E Baum
- Max Planck Bristol Centre for Minimal Biology, School of ChemistryUniversity of BristolBristolUK
| | - Katy L Haydn‐Smith
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | | | - Kate J Heesom
- Proteomics Facility, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | - Charlotte E Severn
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| |
Collapse
|
44
|
Sarcinella A, Femminò S, Brizzi MF. Extracellular Vesicles: Emergent and Multiple Sources in Wound Healing Treatment. Int J Mol Sci 2023; 24:15709. [PMID: 37958693 PMCID: PMC10650196 DOI: 10.3390/ijms242115709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Non-healing wound- and tissue-injury are commonly experienced worldwide by the aging population. The persistence of disease commonly leads to tissue infection, resulting in severe clinical complications. In the last decade, extracellular vesicles (EVs) have been considered promising and emergent therapeutic tools to improve the healing processes. Therefore, efforts have been directed to develop a cell-free therapeutic platform based on EV administration to orchestrate tissue repair. EVs derived from different cell types, including fibroblast, epithelial, and immune cells are recruited to the injured sites and in turn take part in scar formation. EVs are nano-sized particles containing a heterogeneous cargo consisting of lipids, proteins, and nucleic acids protected from degradation by their lipid bilayer. Noteworthy, since EVs have natural biocompatibility and low immunogenicity, they represent the ideal therapeutic candidates for regenerative purposes. Indeed, EVs are released by several cell types, and even if they possess unique biological properties, their functional capability can be further improved by engineering their content and functionalizing their surface, allowing a specific cell cargo delivery. Herein, we provide an overview of preclinical data supporting the contribution of EVs in the repair and regenerative processes, focusing on different naïve EV sources, as well as on their engineering, to offer a scalable and low-cost therapeutic option for tissue repair.
Collapse
Affiliation(s)
| | | | - Maria Felice Brizzi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (A.S.); (S.F.)
| |
Collapse
|
45
|
Kourouklis AP, Kaemmel J, Wu X, Baños M, Chanfon A, de Brot S, Ferrari A, Cesarovic N, Falk V, Mazza E. Transdermal wires for improved integration in vivo. BIOMATERIALS ADVANCES 2023; 153:213568. [PMID: 37591177 DOI: 10.1016/j.bioadv.2023.213568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 08/19/2023]
Abstract
Alternative engineering approaches have led the design of implants with controlled physical features to minimize adverse effects in biological tissues. Similar efforts have focused on optimizing the design features of percutaneous VAD drivelines with the aim to prevent infection, omitting however a thorough look on the implant-skin interactions that govern local tissue reactions. Here, we utilized an integrated approach for the biophysical modification of transdermal implants and their evaluation by chronic sheep implantation in comparison to the standard of care VAD drivelines. We developed a novel method for the transfer of breath topographical features on thin wires with modular size. We examined the impact of implant's diameter, surface topography, and chemistry on macroscopic, histological, and physical markers of inflammation, fibrosis, and mechanical adhesion. All implants demonstrated infection-free performance. The fibrotic response was enhanced by the increasing diameter of implants but not influenced by their surface properties. The implants of small diameter promoted mild inflammatory responses with improved mechanical adhesion and restricted epidermal downgrowth, in both silicone and polyurethane coated transdermal wires. On the contrary, the VAD drivelines with larger diameter triggered severe inflammatory reactions with frequent epidermal downgrowth. We validated these effects by quantifying the infiltration of macrophages and the level of vascularization in the fibrotic zone, highlighting the critical role of size reduction for the benign integration of transdermal implants with skin. This insight on how the biophysical properties of implants impact local tissue reactions could enable new solutions on the transdermal transmission of power, signal, and mass in a broad range of medical devices.
Collapse
Affiliation(s)
- Andreas P Kourouklis
- Department of Mechanical and Process Engineering, Institute for Mechanical Systems, ETH Zurich, 8092 Zurich, Switzerland
| | - Julius Kaemmel
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany
| | - Xi Wu
- Department of Mechanical and Process Engineering, Institute for Mechanical Systems, ETH Zurich, 8092 Zurich, Switzerland
| | - Miguel Baños
- Department of Mechanical and Process Engineering, Institute for Mechanical Systems, ETH Zurich, 8092 Zurich, Switzerland
| | - Astrid Chanfon
- COMPATH, Institute of Animal Pathology, University of Bern, 3012 Bern, Switzerland
| | - Simone de Brot
- COMPATH, Institute of Animal Pathology, University of Bern, 3012 Bern, Switzerland
| | - Aldo Ferrari
- EMPA, Swiss Federal Laboratories for Material Science and Technology, 8600 Dübendorf, Switzerland
| | - Nikola Cesarovic
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany; Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, 13353 Berlin, Germany; Department of Health Sciences and Technology, ETH Zürich, 8093 Zürich, Switzerland
| | - Edoardo Mazza
- Department of Mechanical and Process Engineering, Institute for Mechanical Systems, ETH Zurich, 8092 Zurich, Switzerland; EMPA, Swiss Federal Laboratories for Material Science and Technology, 8600 Dübendorf, Switzerland.
| |
Collapse
|
46
|
Agarwal P, Cadart C, Fort L, Gahan J, Greenspan L, Juan T, Kameneva P, Miao Y. Pathway to Independence: the future of developmental biology. Development 2023; 150:dev202360. [PMID: 37812057 PMCID: PMC10705336 DOI: 10.1242/dev.202360] [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: 10/10/2023]
Abstract
In 2022, Development launched its Pathway to Independence (PI) Programme, aimed at supporting postdocs as they transition to their first independent position. We selected eight talented researchers as the first cohort of PI Fellows. In this article, each of our Fellows provides their perspective on the future of their field. Together, they paint an exciting picture of the current state of and open questions in developmental biology.
Collapse
Affiliation(s)
- Priti Agarwal
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Clotilde Cadart
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - Loic Fort
- Vanderbilt University School of Medicine, 465 21st Avenue South, U 3200 MRB III, Nashville, TN 37240-7935, USA
| | - James Gahan
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Leah Greenspan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Thomas Juan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - Polina Kameneva
- The Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Yuchuan Miao
- Department of Genetics, Harvard Medical School and Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
47
|
Ward J, Martin P. Live-imaging studies reveal how microclots and the associated inflammatory response enhance cancer cell extravasation. J Cell Sci 2023; 136:jcs261225. [PMID: 37671502 PMCID: PMC10561694 DOI: 10.1242/jcs.261225] [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: 04/11/2023] [Accepted: 08/31/2023] [Indexed: 09/07/2023] Open
Abstract
Previous clinical studies and work in mouse models have indicated that platelets and microclots might enable the recruitment of immune cells to the pre-metastatic cancer niche, leading to efficacious extravasation of cancer cells through the vessel wall. Here, we investigated the interaction between platelets, endothelial cells, inflammatory cells, and engrafted human and zebrafish cancer cells by live-imaging studies in translucent zebrafish larvae, and show how clotting (and clot resolution) act as foci and as triggers for extravasation. Fluorescent tagging in each lineage revealed their dynamic behaviour and potential roles in these events, and we tested function by genetic and drug knockdown of the contributing players. Morpholino knockdown of fibrinogen subunit α (fga) and warfarin treatment to inhibit clotting both abrogated extravasation of cancer cells. The inflammatory phenotype appeared fundamental, and we show that forcing a pro-inflammatory, tnfa-positive phenotype is inhibitory to extravasation of cancer cells.
Collapse
Affiliation(s)
- Juma Ward
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
48
|
Zhong J, Xiao C, Chen Q, Pan X, Xu T, Wang Y, Hou W, Liu L, Cao F, Wang Y, Li X, Zhou L, Yang H, Yang Y, Zhao C. Zebrafish functional xenograft vasculature platform identifies PF-502 as a durable vasculature normalization drug. iScience 2023; 26:107734. [PMID: 37680473 PMCID: PMC10480778 DOI: 10.1016/j.isci.2023.107734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/21/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Tumor vasculature often exhibits disorder and inefficiency. Vascular normalization offers potential for alleviating hypoxia and optimizing drug delivery in tumors. However, identifying effective agents is hindered by a lack of robust screening. We aimed to establish a comprehensive method using the zebrafish functional xenograft vasculature platform (zFXVP) to visualize and quantify tumor vasculature changes. Employing zFXVP, we systematically screened compounds, identifying PF-502 as a robust vascular normalization agent. Mechanistic studies showed PF-502 induces endothelial cell-cycle arrest, streamlines vasculature, and activates Notch1 signaling, enhancing stability and hemodynamics. In murine models, PF-502 exhibited pronounced vascular normalization and improved drug delivery at a sub-maximum tolerated dose. These findings highlight zFXVP's utility and suggest PF-502 as a promising adjunctive for vascular normalization in clinical settings.
Collapse
Affiliation(s)
- Jian Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Chaoxin Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Qin Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Xiangyu Pan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Tongtong Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yiyun Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Wanting Hou
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Lu Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Fujun Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yulin Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Xiaoying Li
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Lin Zhou
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yu Yang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| |
Collapse
|
49
|
Attiogbe E, Larochelle S, Chaib Y, Mainzer C, Mauroux A, Bordes S, Closs B, Gilbert C, Moulin VJ. An in vitro autologous, vascularized, and immunocompetent Tissue Engineered Skin model obtained by the self-assembled approach. Acta Biomater 2023; 168:361-371. [PMID: 37419164 DOI: 10.1016/j.actbio.2023.06.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023]
Abstract
A complete in vitro skin model, containing resident cell types is needed to understand physiology and to consider the role of immune and endothelial cells in dermal drug testing. In this study, a cell extraction technique was developed to isolate resident skin cells from the same human donor while preserving the immune and endothelial cells. Then those cells were used to reconstruct an autologous, vascularized, and immunocompetent Tissue-Engineered Skin model, aviTES. Phenotypic characterization of the viable cells was performed on freshly isolated cells and after thawing through flow cytometry. Dermal cell extracts were characterized as fibroblasts, endothelial and immune cells, and the average amount of each cell type represents 4, 0.5, and 1 million viable cells per g of the dermis, respectively. The 3D models, TES and aviTES, were characterized by a fully differentiated epidermis that showed an increase in the presence of Ki67+ cells in the basolateral layer of the aviTES model. Capillary-like network formation, through the self-assembly of endothelial cells, and the presence of functional immune cells were identified through immunofluorescence staining in aviTES. In addition, the aviTES model was immunocompetent, as evidenced by its capacity to increase the production of pro-inflammatory cytokines TNF-α, MIP-1α, and GM-CSF following LPS stimulation. This study describes an autologous skin model containing a functional resident skin immune system and a capillary network. It provides a relevant tool to study the contribution of the immune system to skin diseases and inflammatory responses and to investigate resident skin cell interactions and drug development. STATEMENT OF SIGNIFICANCE: There is an urgent need for a complete in vitro skin model containing the resident cell types to better understand the role of immune and endothelial cells in skin and to be able to use it for drug testing. Actual 3D models of human skin most often contain only fibroblasts and keratinocytes with a limited number of models containing endothelial cells or a limited variety of immune cells. This study describes an autologous skin model containing a functional resident skin immune system and a capillary network. It provides a relevant tool to study the contribution of the immune system to skin diseases and inflammatory responses and to investigate interactions between resident skin cell, improving our capacity to develop new drugs.
Collapse
Affiliation(s)
- Emilie Attiogbe
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Sébastien Larochelle
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Yanis Chaib
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | | | | | | | | | - Caroline Gilbert
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Véronique J Moulin
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
50
|
Zheng H, Cheng X, Jin L, Shan S, Yang J, Zhou J. Recent advances in strategies to target the behavior of macrophages in wound healing. Biomed Pharmacother 2023; 165:115199. [PMID: 37517288 DOI: 10.1016/j.biopha.2023.115199] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 08/01/2023] Open
Abstract
Chronic wounds and scar formation are widespread due to limited suitable remedies. The macrophage is a crucial regulator in wound healing, controlling the onset and termination of inflammation and regulating other processes related to wound healing. The current breakthroughs in developing new medications and drug delivery methods have enabled the accurate targeting of macrophages in oncology and rheumatic disease therapies through clinical trials. These successes have cleared the way to utilize drugs targeting macrophages in various disorders. This review thus summarizes macrophage involvement in normal and pathologic wound healing. It further details the targets available for macrophage intervention and therapeutic strategies for targeting the behavior of macrophages in tissue repair and regeneration.
Collapse
Affiliation(s)
- Hongkun Zheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xinwei Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lu Jin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Jia Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|