1
|
Zhang F, Zhang H, Wang S, Gao M, Du K, Chen X, Lu Y, Hu Q, Du A, Du S, Wang J, Shi K, Chen Z, Li Z, Li Z, Xiao J. A dynamically phase-adaptive regulating hydrogel promotes ultrafast anti-fibrotic wound healing. Nat Commun 2025; 16:3738. [PMID: 40254609 PMCID: PMC12009973 DOI: 10.1038/s41467-025-58987-w] [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: 08/30/2024] [Accepted: 04/04/2025] [Indexed: 04/22/2025] Open
Abstract
Achieving rapid and scar-free wound repair is a key goal in the field of regenerative medicine. Herein, a dynamically Schiff base-crosslinked hydrogel (F/R gel) with phase-adaptive regulating functions is constructed to integratedly promote rapid re-epithelization with suppressed scars on chronic infected wounds. Specifically, the gel effectively eliminates multidrug-resistant bacterial biofilm at infection stage via antimicrobial activity of ε-polylysine firstly dissociated from hydrogel matrix in infectious microenvironment, and interrupts the severe oxidative stress-inflammation cycle at wound site by the released ceria nanozyme, thus stimulating a pro-regenerative environment to ensure tissue repair. Subsequently, fibroblast growth factor/c-Jun siRNA co-loaded microcapsules gradually disintegrate to release drugs, facilitating neoangiogenesis and cell proliferation but simultaneously blocking c-Jun overexpression for fibrotic scar suppression. Notably, the F/R gel facilitates normal-like skin regeneration with no perceptible scars formed on infected male mouse wound and female rabbit ear wound models. Our work offers a promising regenerative strategy emphasizing immunomodulatory and fibroblast subtype modulation for scarless wound repair.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Haijuan Zhang
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Shengfu Wang
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Mingying Gao
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Kaiyi Du
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xinyuan Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yang Lu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Qianqian Hu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Anyu Du
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Shenghu Du
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jian Wang
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Keqing Shi
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zimiao Chen
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhuo Li
- Key Laboratory of Functional Inorganic Materials Chemistry (Ministry of Education), School of Chemistry and Materials Science, Heilongjiang University, Harbin, 150080, P. R. China.
| | - Zhenglin Li
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Jian Xiao
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China.
| |
Collapse
|
2
|
Aydın M, Akyüz S, Yanik H, Yildirim E, Başak AM, Güven HE, Gülap Y, Yilmaz KB. Autologous adipose-derived tissue stromal vascular fraction and intralesional epidermal growth factor combined application in patients with diabetic foot. J Wound Care 2025; 34:xxx-xxxviii. [PMID: 40056382 DOI: 10.12968/jowc.2024.0107] [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: 03/10/2025]
Abstract
OBJECTIVE The aim of this study was to evaluate the effect on wound healing of intralesional epidermal growth factor (iEGF) (Heberprot-p; Hasbiotech, Cuba) and autologous adipose-derived tissue stromal vascular fraction (AD-tSVF) applied in the closure of tissue defects. METHOD The patients included in the study were separated into three approximately equal groups: Group 1 with iEGF+AD-tSVF applied; Group 2 with iEGF only applied; and Group 3 with conventional wound care products applied. Granulation tissue was taken from the wound bed before the application of iEGF and AD-tSVF and at intervals thereafter for flow cytometry analysis. RESULTS Group 1 included 11 patients; Group 2 included 10 patients; and Group 3 included 10 patients. The time to re-epithelialisation was determined as 187.60±68.78 days in Group 3 patients compared with Group 1 (72.27±10.33 days) and Group 2 (70.50±18.02 days) (p<0.001). Following the application of iEGF to the wound bed, an increase was observed in M2 macrophage (CD209+), and M1 macrophage (CD38+) levels. The (CD34+) stem cells obtained from the granulation tissue after the application of AD-tSVF were determined to still be statistically significantly increased in the wound bed on the 21st day. CONCLUSION The results of this study demonstrated that the application of iEGF and iEGF+ AD-tSVF significantly shortened the wound healing period compared with conventional methods. AD-tSVF stands as an effective option, especially in the patient group with halted or delayed wound healing despite the application of iEGF. Moreover, the significant increase (p<0.001) in the level of M2 macrophages (CD209+), M1 macrophages (CD38+) and stem cells (CD34+) provided by this treatment modality showed that it contributed to wound healing at the cellular level.
Collapse
Affiliation(s)
- Mustafa Aydın
- MD, Orthopaedic Surgeon, Department of Orthopedics and Traumatology, University of Health Sciences Gulhane Training and Research Hospital, Ankara, Turkey
| | - Simay Akyüz
- PhD, RN, Assistant Professor, University of Health Sciences, Gulhane Faculty of Nursing, Ankara, Turkey
| | - Hamdullah Yanik
- PhD, Molecular Biology and Genetics Specialist, Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Turkey
| | - Eda Yildirim
- MD, Orthopaedic Surgeon, Department of Orthopedics and Traumatology, University of Health Sciences Gulhane Training and Research Hospital, Ankara, Turkey
| | - Ali Murat Başak
- MD, Orthopaedic Surgeon, Department of Orthopedics and Traumatology, University of Health Sciences Gulhane Training and Research Hospital, Ankara, Turkey
| | - Hikmet Erhan Güven
- MD, Associate Professor, General Surgeon, Department of General Surgery, Etlik City Hospital, Ankara, Turkey
| | - Yasin Gülap
- MD, General Surgeon, Department of General Surgery, University of Health Sciences Gulhane Training and Research Hospital, Ankara, Turkey
| | - Kerim Bora Yilmaz
- PhD, Molecular Biology and Genetics Specialist, Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Turkey
- MD, General Surgeon, Department of General Surgery, University of Health Sciences Gulhane Training and Research Hospital, Ankara, Turkey
- MD, Professor, General Surgeon, Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
| |
Collapse
|
3
|
Yang J, Zhang L, Wang Y, Wang N, Wei H, Zhang S, Ding Q, Sun S, Ding C, Liu W. Dihydromyricetin-loaded oxidized polysaccharide/L-arginine chitosan adhesive hydrogel promotes bone regeneration by regulating PI3K/AKT signaling pathway and MAPK signaling pathway. Carbohydr Polym 2024; 346:122614. [PMID: 39245525 DOI: 10.1016/j.carbpol.2024.122614] [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: 04/26/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024]
Abstract
Bone defects caused by trauma, infection and congenital diseases still face great challenges. Dihydromyricetin (DHM) is a kind of flavone extracted from Ampelopsis grossedentata, a traditional Chinese medicine. DHM can enhance the osteogenic differentiation of human bone marrow mesenchymal stem cells with the potential to promote bone regeneration. Hydrogel can be used as a carrier of DHM to promote bone regeneration due to its unique biochemical characteristics and three-dimensional structure. In this study, oxidized phellinus igniarius polysaccharides (OP) and L-arginine chitosan (CA) are used to develop hydrogel. The pore size and gel strength of the hydrogel can be changed by adjusting the oxidation degree of oxidized phellinus igniarius polysaccharides. The addition of DHM further reduce the pore size of the hydrogel (213 μm), increase the mechanical properties of the hydrogel, and increase the antioxidant and antibacterial activities of the hydrogel. The scavenging rate of DPPH are 72.30 ± 0.33 %, and the inhibition rate of E.coli and S.aureus are 93.12 ± 0.38 % and 94.49 ± 1.57 %, respectively. In addition, PCAD has good adhesion and biocompatibility, and its extract can effectively promote the osteogenic differentiation of MC3T3-E1 cells. Network pharmacology and molecular docking show that the promoting effect of DHM on osteogenesis may be achieved by activating the PI3K/AKT and MAPK signaling pathways. This is confirmed through in vitro cell experiments and in vivo animal experiments.
Collapse
Affiliation(s)
- Jiali Yang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Lifeng Zhang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Yue Wang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Ning Wang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Hewei Wei
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Shuai Zhang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Qiteng Ding
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Shuwen Sun
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Chuanbo Ding
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China.
| | - Wencong Liu
- School of Food and Pharmaceutical Engineering, Wuzhou University, Wuzhou 543002, China.
| |
Collapse
|
4
|
Yin Q, Luo XY, Ma K, Liu ZZ, Gao Y, Zhang JB, Chen W, Yang YJ. Hyaluronic Acid/Gelatin-Based Multifunctional Bioadhesive Hydrogel Loaded with a Broad-Spectrum Bacteriocin for Enhancing Diabetic Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47226-47241. [PMID: 39193631 DOI: 10.1021/acsami.4c09309] [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: 08/29/2024]
Abstract
The development of multifunctional wound adhesives is critical in clinical settings due to the scarcity of dressings with effective adhesive properties while protecting against infection by drug-resistant bacteria. Polysaccharide and gelatin-based hydrogels, known for their biocompatibility and bioactivity, assist in wound healing. This study introduces a multifunctional bioadhesive hydrogel developed through dynamic covalent bonding and light-triggered covalent bonding, comprising oxidized hyaluronic acid, methacrylated gelatin, and the bacteriocin recently discovered by our lab, named jileicin (JC). The adhesion strength of the hydrogel, measured at 180 kPa, was 4.35 times higher than that of the fibrin glue. Furthermore, the hydrogel demonstrated robust platelet adhesion, procoagulant activity, and outstanding hemostatic properties in a mouse liver injury model. Incorporating JC significantly enhanced the phagocytosis and bactericidal capabilities of the macrophages. This immunomodulatory function on host cells, coupled with its potent bacterial membrane-disrupting ability, makes JC an effective killer against methicillin-resistant Staphylococcus aureus. In wound repair experiments on diabetic mice with infected full-thickness skin defects, the hydrogel treatment group showed a notable reduction in bacterial load, accelerated M2-type macrophage polarization, diminished inflammation, and hastened wound healing. Owing to its outstanding biocompatibility, antibacterial activity, and controlled adhesion, this hydrogel presents a promising therapeutic option for treating infected skin wounds.
Collapse
Affiliation(s)
- Qi Yin
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130000, China
| | - Xue-Yue Luo
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130000, China
| | - Ke Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy Agricultural Sciences, Lanzhou 730000, China
| | - Zhen-Zhen Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130000, China
| | - Yu Gao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130000, China
| | - Jia-Bao Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130000, China
| | - Wei Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130000, China
| | - Yong-Jun Yang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun 130000, China
| |
Collapse
|
5
|
Yang C, Yuan W, Liao G, Tang Z, Zhu T, Jia Y, Yu Q, Wang L. Customized Vascular Repair Microenvironment: Poly(lactic acid)-Gelatin Nanofibrous Scaffold Decorated with bFGF and Ag@Fe 3O 4 Core-Shell Nanowires. ACS APPLIED MATERIALS & INTERFACES 2024; 16:40787-40804. [PMID: 39072379 DOI: 10.1021/acsami.4c09269] [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: 07/30/2024]
Abstract
Vascular defects caused by trauma or vascular diseases can significantly impact normal blood circulation, resulting in serious health complications. Vascular grafts have evolved as a popular approach for vascular reconstruction with promising outcomes. However, four of the greatest challenges for successful application of small-diameter vascular grafts are (1) postoperative anti-infection, (2) preventing thrombosis formation, (3) utilizing the inflammatory response to the graft to induce tissue regeneration and repair, and (4) noninvasive monitoring of the scaffold and integration. The present study demonstrated a basic fibroblast growth factor (bFGF) and oleic acid dispersed Ag@Fe3O4 core-shell nanowires (OA-Ag@Fe3O4 CSNWs) codecorated poly(lactic acid) (PLA)/gelatin (Gel) multifunctional electrospun vascular grafts (bAPG). The Ag@Fe3O4 CSNWs have sustained Ag+ release and exceptional photothermal capabilities to effectively suppress bacterial infections both in vitro and in vivo, noninvasive magnetic resonance imaging (MRI) modality to monitor the position of the graft, and antiplatelet adhesion properties to promise long-term patency. The gradually released bFGF from the bAPG scaffold promotes the M2 macrophage polarization and enhances the recruitment of macrophages, endothelial cells (ECs) and fibroblast cells. This significant regulation of diverse cell behavior has been proven to be beneficial to vascular repair and regeneration both in vitro and in vivo. Therefore, this study supplies a method to prepare multifunctional vascular-repair materials and is expected to represent a significant guidance and reference to the development of biomaterials for vascular tissue engineering.
Collapse
Affiliation(s)
- Congyi Yang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, Guangzhou 510640, China
| | - Weiwen Yuan
- Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Guoxing Liao
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, Guangzhou 510640, China
| | - Zhe Tang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, Guangzhou 510640, China
| | - Tong Zhu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, Guangzhou 510640, China
| | - Yifan Jia
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, Guangzhou 510640, China
| | - Qianqian Yu
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, Guangzhou 510640, China
| | - LinGe Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, Guangdong Basic Research Center of Excellence for Energy & Information Polymer Materials, South China University of Technology, Guangzhou 510640, China
- State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
6
|
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
|
7
|
Savitri C, Ha SS, Kwon JW, Kim SH, Kim Y, Park HM, Kwon H, Ji MJ, Park K. Human Fibroblast-Derived Matrix Hydrogel Accelerates Regenerative Wound Remodeling Through the Interactions with Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305852. [PMID: 38476050 PMCID: PMC11095160 DOI: 10.1002/advs.202305852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/21/2024] [Indexed: 03/14/2024]
Abstract
Herein, a novel extracellular matrix (ECM) hydrogel is proposed fabricated solely from decellularized, human fibroblast-derived matrix (FDM) toward advanced wound healing. This FDM-gel is physically very stable and viscoelastic, while preserving the natural ECM diversity and various bioactive factors. Subcutaneously transplanted FDM-gel provided a permissive environment for innate immune cells infiltration. Compared to collagen hydrogel, excellent wound healing indications of FDM-gel treated in the full-thickness wounds are noticed, particularly hair follicle formation via highly upregulated β-catenin. Sequential analysis of the regenerated wound tissues disclosed that FDM-gel significantly alleviated pro-inflammatory cytokine and promoted M2-like macrophages, along with significantly elevated vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) level. A mechanistic study demonstrated that macrophages-FDM interactions through cell surface integrins α5β1 and α1β1 resulted in significant production of VEGF and bFGF, increased Akt phosphorylation, and upregulated matrix metalloproteinase-9 activity. Interestingly, blocking such interactions using specific inhibitors (ATN161 for α5β1 and obtustatin for α1β1) negatively affected those pro-healing growth factors secretion. Macrophages depletion animal model significantly attenuated the healing effect of FDM-gel. This study demonstrates that the FDM-gel is an excellent immunomodulatory material that is permissive for host cells infiltration, resorbable with time, and interactive with macrophages, where it thus enables regenerative matrix remodeling toward a complete wound healing.
Collapse
Affiliation(s)
- Cininta Savitri
- Center for BiomaterialsKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Sang Su Ha
- Center for BiomaterialsKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Jae Won Kwon
- Center for BiomaterialsKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and Technology, KIST SchoolUniversity of Science and Technology (UST)Seoul02792Republic of Korea
| | - Sung Hoon Kim
- Center for BiomaterialsKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Young‐Min Kim
- Center for BiomaterialsKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and Technology, KIST SchoolUniversity of Science and Technology (UST)Seoul02792Republic of Korea
| | - Hyun Mee Park
- Advanced Analysis and Data CenterKISTSeoul02792Republic of Korea
| | - Haejin Kwon
- Advanced Analysis and Data CenterKISTSeoul02792Republic of Korea
| | - Mi Jung Ji
- Advanced Analysis and Data CenterKISTSeoul02792Republic of Korea
| | - Kwideok Park
- Center for BiomaterialsKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science and Technology, KIST SchoolUniversity of Science and Technology (UST)Seoul02792Republic of Korea
| |
Collapse
|
8
|
Vongsakulpaisarn P, Sangkhamanee SS, Rassameemasmaung S, Sritanaudomchai H. Effect of Periodontal Ligament Stem Cells-Derived Conditioned Medium on Gene Expression and Differentiation of Tumor Necrosis Factor-α-Challenged Osteoblasts. Eur J Dent 2024; 18:378-386. [PMID: 37562430 PMCID: PMC10959631 DOI: 10.1055/s-0043-1771337] [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: 08/12/2023] Open
Abstract
OBJECTIVES Tumor necrosis factor-α (TNF-α) causes bone resorption in periodontitis. It induces the production of receptor activator of NF-κB ligand (RANKL) from osteoblasts, leading to the disturbance of bone homeostasis through RANKL, RANK, and osteoprotegerin (OPG) axis. This study aimed to explore the effect of periodontal ligament stem cells-derived conditioned medium (PDLSCs-CM) on gene expression related to bone homeostasis and the differentiation of TNF-α-challenged osteoblasts. MATERIALS AND METHODS Human osteoblasts were cultured with 50 ng/mL of TNF-α and 0, 1, 10, and 100 µg/ mL of PDLSCs-CM. Osteoblasts cultured without TNF-α and PDLSCs-CM were served as control. Gene expression of RANKL, OPG, and interleukin-1β (IL-1β) was evaluated by reverse transcription quantitative polymerase chain reaction at 48 hours. The early-stage and late-stage differentiation of TNF-α-challenged osteoblasts without or with PDLSCs-CM was explored by alkaline phosphatase (ALP) activity and alizarin red staining, respectively, at day 1, 3, 6, 9, and 12. STATISTICAL ANALYSIS Mann-Whitney U test was used to analyze the differences in gene expression of TNF-α-challenged osteoblasts at 24 and 48 hours, and Kruskal-Wallis test was used to analyze the effect of PDLSCs-CM on gene expression and ALP activity among all experimental groups using SPSS software version 21.0. Statistical significance was considered with p-value less than 0.05. RESULTS Expression of RANKL, OPG and IL-1β was significantly upregulated in TNF-α-challenged osteoblasts compared to the untreated control. The PDLSCs-CM at 1 and 10 μg/mL downregulated gene expression of TNF-α-challenged osteoblasts compared to the group without PDLSCs-CM, but the difference did not reach statistical significance. The ALP activity was decreased in TNF-α-challenged osteoblasts. The addition of PDLSCs-CM did not alter ALP activity of TNF-α-challenged osteoblasts. Alizarin red staining was comparable in the TNF-α-challenged osteoblasts cultured without or with PDLSCs-CM. CONCLUSIONS The PDLSCs-CM did not alter gene expression involved in bone homeostasis and differentiation of TNF-α-challenged osteoblasts.
Collapse
Affiliation(s)
- Poranee Vongsakulpaisarn
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | | | - Supanee Rassameemasmaung
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
9
|
Zhao Y, Li M, Mao J, Su Y, Huang X, Xia W, Leng X, Zan T. Immunomodulation of wound healing leading to efferocytosis. SMART MEDICINE 2024; 3:e20230036. [PMID: 39188510 PMCID: PMC11235971 DOI: 10.1002/smmd.20230036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 08/28/2024]
Abstract
Effectively eliminating apoptotic cells is precisely controlled by a variety of signaling molecules and a phagocytic effect known as efferocytosis. Abnormalities in efferocytosis may bring about the development of chronic conditions, including angiocardiopathy, chronic inflammatory diseases and autoimmune diseases. During wound healing, failure of efferocytosis leads to the collection of apoptosis, the release of necrotic material and chronic wounds that are difficult to heal. In addition to the traditional phagocytes-macrophages, other important cell species including dendritic cells, neutrophils, vascular endothelial cells, fibroblasts and keratinocytes contribute to wounding healing. This review summarizes how efferocytosis-mediated immunomodulation plays a repair-promoting role in wound healing, providing new insights for patients suffering from various cutaneous wounds.
Collapse
Affiliation(s)
- Yun Zhao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Cosmetic and Plastic SurgeryAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Minxiong Li
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiayi Mao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yinghong Su
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xin Huang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenzheng Xia
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiangfeng Leng
- Department of Cosmetic and Plastic SurgeryAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Tao Zan
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
10
|
Kumar B, Khatpe AS, Guanglong J, Batic K, Bhat-Nakshatri P, Granatir MM, Addison RJ, Szymanski M, Baldridge LA, Temm CJ, Sandusky G, Althouse SK, Cote ML, Miller KD, Storniolo AM, Nakshatri H. Stromal heterogeneity may explain increased incidence of metaplastic breast cancer in women of African descent. Nat Commun 2023; 14:5683. [PMID: 37709737 PMCID: PMC10502140 DOI: 10.1038/s41467-023-41473-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/05/2023] [Indexed: 09/16/2023] Open
Abstract
The biologic basis of genetic ancestry-dependent variability in disease incidence and outcome is just beginning to be explored. We recently reported enrichment of a population of ZEB1-expressing cells located adjacent to ductal epithelial cells in normal breasts of women of African ancestry compared to those of European ancestry. In this study, we demonstrate that these cells have properties of fibroadipogenic/mesenchymal stromal cells that express PROCR and PDGFRα and transdifferentiate into adipogenic and osteogenic lineages. PROCR + /ZEB1 + /PDGFRα+ (PZP) cells are enriched in normal breast tissues of women of African compared to European ancestry. PZP: epithelial cell communication results in luminal epithelial cells acquiring basal cell characteristics and IL-6-dependent increase in STAT3 phosphorylation. Furthermore, level of phospho-STAT3 is higher in normal and cancerous breast tissues of women of African ancestry. PZP cells transformed with HRasG12V ± SV40-T/t antigens generate metaplastic carcinoma suggesting that these cells are one of the cells-of-origin of metaplastic breast cancers.
Collapse
Affiliation(s)
- Brijesh Kumar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, 221005, India
| | - Aditi S Khatpe
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jiang Guanglong
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Katie Batic
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | | | - Maggie M Granatir
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Rebekah Joann Addison
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Megan Szymanski
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Lee Ann Baldridge
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Constance J Temm
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - George Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sandra K Althouse
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Michele L Cote
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Kathy D Miller
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Anna Maria Storniolo
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- VA Roudebush Medical Center, Indianapolis, IN, 46202, USA.
| |
Collapse
|
11
|
Bai J, Zhang W, Zhou C, Zhao G, Zhong H, Hang K, Xu J, Zhang W, Chen E, Wu J, Liu L, Xue D. MFG-E8 promotes osteogenic differentiation of human bone marrow mesenchymal stem cells through GSK3β/β-catenin signaling pathway. FASEB J 2023; 37:e22950. [PMID: 37144883 DOI: 10.1096/fj.202201417rrr] [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: 08/30/2022] [Revised: 04/13/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023]
Abstract
Fracture nonunion and bone defects are challenging for orthopedic surgeons. Milk fat globule-epidermal growth factor 8 (MFG-E8), a glycoprotein possibly secreted by macrophages in a fracture hematoma, participates in bone development. However, the role of MFG-E8 in the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is unclear. We investigated the osteogenic effect of MFG-E8 in vitro and in vivo. The CCK-8 assay was used to assess the effect of recombinant human MFG-E8 (rhMFG-E8) on the viability of hBMSCs. Osteogenesis was investigated using RT-PCR, Western blotting, and immunofluorescence. Alkaline phosphatase (ALP) and Alizarin red staining were used to evaluate ALP activity and mineralization, respectively. An enzyme-linked immunosorbent assay was conducted to evaluate the secretory MFG-E8 concentration. Knockdown and overexpression of MFG-E8 in hBMSCs were established via siRNA and lentivirus vector transfection, respectively. Exogenous rhMFG-E8 was used to verify the in vivo therapeutic effect in a tibia bone defect model based on radiographic analysis and histological evaluation. Endogenous and secretory MFG-E8 levels increased significantly during the early osteogenic differentiation of hBMSCs. Knockdown of MFG-E8 inhibited the osteogenic differentiation of hBMSCs. Overexpression of MFG-E8 and rhMFG-E8 protein increased the expression of osteogenesis-related genes and proteins and enhanced calcium deposition. The active β-catenin to total β-catenin ratio and the p-GSK3β protein level were increased by MFG-E8. The MFG-E8-induced enhanced osteogenic differentiation of hBMSCs was partially attenuated by a GSK3β/β-catenin signaling inhibitor. Recombinant MFG-E8 accelerated bone healing in a rat tibial-defect model. In conclusion, MFG-E8 promotes the osteogenic differentiation of hBMSCs by regulating the GSK3β/β-catenin signaling pathway and so, is a potential therapeutic target.
Collapse
Affiliation(s)
- Jinwu Bai
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Weijun Zhang
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Chenwei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Guangfeng Zhao
- Department of Emergency, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Huiming Zhong
- Department of Emergency, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Kai Hang
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Jianxiang Xu
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Wei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Erman Chen
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Jiaqi Wu
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| | - Ling Liu
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Deting Xue
- Department of Orthopaedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, People's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, Zhejiang Province, PR China
| |
Collapse
|
12
|
Kuang L, Zhang C, Li B, Deng H, Chen R, Li G. Human Keratinocyte-Derived Exosomal MALAT1 Promotes Diabetic Wound Healing by Upregulating MFGE8 via microRNA-1914-3p. Int J Nanomedicine 2023; 18:949-970. [PMID: 36852184 PMCID: PMC9961177 DOI: 10.2147/ijn.s399785] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Purpose Diabetic wound is a highly prevalent and refractory disease. Extensive studies have confirmed that keratinocytes and macrophages play an important role in the process of wound healing. Additionally, exosomes are regarded as a vital intercellular communication tool. This study aimed to investigate the role of human keratinocyte-derived exosomal MALAT1 in the treatment of diabetic wound by influencing the biological function of macrophages. Methods We mainly assessed the function of MALAT1 on the biological changes of macrophages, and the expression of MALAT1 in the keratinocyte-exosomes analyzed by quantitative real-time polymerase chain reaction (RT-qPCR). The downstream interaction between RNAs or proteins was assessed by mechanistic experiments. Besides, we evaluated the effects of human keratinocyte-derived exosomal MALAT1 on diabetic wound healing in vivo to verify in vitro results. Results We demonstrated that human keratinocyte-derived exosomal MALAT1 enhanced the biological functions of high glucose-injured macrophages, including phagocytosis, converting to a pro-healing phenotype and reducing apoptosis. Mechanistically, MALAT1 accelerated the expression of MFGE8 by competitively binding to miR-1914-3p, thereby affecting the function of macrophages and the signal axis of TGFB1/SMAD3, and finally promoting the healing of diabetic wounds. Human keratinocyte-derived exosomal MALAT1 might promote collagen deposition, ECM remodeling, and expression of MFGE8, VEGF, and CD31 but reduce the expression of TGFB and SMAD3 in an in vivo model of diabetic mice wounds, which accelerated diabetic wound healing and restored its function. Conclusion The current study revealed that human keratinocyte-derived exosomal MALAT1 would suppress miR-1914-3p to activate MFGE8 and eventually promote wound healing by enhancing macrophage phagocytosis, converting to a pro-healing phenotype and reducing apoptosis. It proposed that keratinocyte-derived exosomes might have the capacity to serve as a new method for the clinical treatment of diabetic wound.
Collapse
Affiliation(s)
- Liwen Kuang
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430062, People’s Republic of China
| | - Chenchen Zhang
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430062, People’s Republic of China
| | - Binghui Li
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430062, People’s Republic of China
| | - Haibo Deng
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430062, People’s Republic of China
| | - Ran Chen
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430062, People’s Republic of China
| | - Gongchi Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People’s Republic of China,Correspondence: Gongchi Li, Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Road, Wuhan, Hubei, 430022, People’s Republic of China, Tel +8613618615209, Email
| |
Collapse
|
13
|
Chen K, Henn D, Sivaraj D, Bonham CA, Griffin M, Kussie HC, Padmanabhan J, Trotsyuk AA, Wan DC, Januszyk M, Longaker MT, Gurtner GC. Mechanical Strain Drives Myeloid Cell Differentiation Toward Proinflammatory Subpopulations. Adv Wound Care (New Rochelle) 2022; 11:466-478. [PMID: 34278820 PMCID: PMC9805866 DOI: 10.1089/wound.2021.0036] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 06/27/2021] [Indexed: 01/13/2023] Open
Abstract
Objective: After injury, humans and other mammals heal by forming fibrotic scar tissue with diminished function, and this healing process involves the dynamic interplay between resident cells within the skin and cells recruited from the circulation. Recent studies have provided mounting evidence that external mechanical forces stimulate intracellular signaling pathways to drive fibrotic processes. Innovation: While most studies have focused on studying mechanotransduction in fibroblasts, recent data suggest that mechanical stimulation may also shape the behavior of immune cells, referred to as "mechano-immunomodulation." However, the effect of mechanical strain on myeloid cell recruitment and differentiation remains poorly understood and has never been investigated at the single-cell level. Approach: In this study, we utilized a three-dimensional (3D) in vitro culture system that permits the precise manipulation of mechanical strain applied to cells. We cultured myeloid cells and used single-cell RNA-sequencing to interrogate the effects of strain on myeloid differentiation and transcriptional programming. Results: Our data indicate that myeloid cells are indeed mechanoresponsive, with mechanical stress influencing myeloid differentiation. Mechanical strain also upregulated a cascade of inflammatory chemokines, most notably from the Ccl family. Conclusion: Further understanding of how mechanical stress affects myeloid cells in conjunction with other cell types in the complicated, multicellular milieu of wound healing may lead to novel insights and therapies for the treatment of fibrosis.
Collapse
Affiliation(s)
- Kellen Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Dominic Henn
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Dharshan Sivaraj
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Clark A. Bonham
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michelle Griffin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Hudson C. Kussie
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jagannath Padmanabhan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Artem A. Trotsyuk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Derrick C. Wan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael Januszyk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, California, USA
| | - Geoffrey C. Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
14
|
Geoffroy K, Laplante P, Clairefond S, Azzi F, Trudel D, Lattouf JB, Stagg J, Saad F, Mes-Masson AM, Bourgeois-Daigneault MC, Cailhier JF. High Levels of MFG-E8 Confer a Good Prognosis in Prostate and Renal Cancer Patients. Cancers (Basel) 2022; 14:cancers14112790. [PMID: 35681775 PMCID: PMC9179566 DOI: 10.3390/cancers14112790] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/19/2022] [Accepted: 06/02/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary In the present study, we analyzed the distribution and prognostic impact of milk fat globule-epidermal growth factor-8 (MFG-E8) protein expression in patients with prostate and renal cancers. Our data highlighted MFG-E8 expression by tumor cells in the epithelium. Our results also showed that low levels of MFG-E8 in prostate and renal cancers were associated with worse clinical outcomes. Furthermore, higher numbers of CD206+ cells were found in the peripheral regions of renal clear cell carcinoma that expressed lower MFG-E8 levels. Globally, our results suggest that MFG-E8 expression could potentially be used as a prognostic marker in prostate and renal cancers. Abstract Milk fat globule-epidermal growth factor-8 (MFG-E8) is a glycoprotein secreted by different cell types, including apoptotic cells and activated macrophages. MFG-E8 is highly expressed in a variety of cancers and is classically associated with tumor growth and poor patient prognosis through reprogramming of macrophages into the pro-tumoral/pro-angiogenic M2 phenotype. To date, correlations between levels of MFG-E8 and patient survival in prostate and renal cancers remain unclear. Here, we quantified MFG-E8 and CD68/CD206 expression by immunofluorescence staining in tissue microarrays constructed from renal (n = 190) and prostate (n = 274) cancer patient specimens. Percentages of MFG-E8-positive surface area were assessed in each patient core and Kaplan–Meier analyses were performed accordingly. We found that MFG-E8 was expressed more abundantly in malignant regions of prostate tissue and papillary renal cell carcinoma but was also increased in the normal adjacent regions in clear cell renal carcinoma. In addition, M2 tumor-associated macrophage staining was increased in the normal adjacent tissues compared to the malignant areas in renal cancer patients. Overall, high tissue expression of MFG-E8 was associated with less disease progression and better survival in prostate and renal cancer patients. Our observations provide new insights into tumoral MFG-E8 content and macrophage reprogramming in cancer.
Collapse
Affiliation(s)
- Karen Geoffroy
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
| | - Patrick Laplante
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
| | - Sylvie Clairefond
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
| | - Feryel Azzi
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
- Division of Pathology and Cellular Biology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Dominique Trudel
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
- Division of Pathology and Cellular Biology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Jean-Baptiste Lattouf
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
- Division of Urology, Department of Surgery, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - John Stagg
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
- Faculté de Pharmacie, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Fred Saad
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
- Division of Urology, Department of Surgery, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Anne-Marie Mes-Masson
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
- Department of Medicine, Faculté de Médecine, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
- Department de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Jean-François Cailhier
- Institut du Cancer de Montréal (ICM), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (K.G.); (P.L.); (S.C.); (F.A.); (D.T.); (J.-B.L.); (J.S.); (F.S.); (A.-M.M.-M.); (M.-C.B.-D.)
- Department of Medicine, Faculté de Médecine, Université de Montréal, Montreal, QC H3C 3J7, Canada
- Division of Nephrology, Department of Medicine, Université de Montréal, Montreal, QC H3C 3J7, Canada
- Correspondence: ; Tel.: +1-514-890-8000-x25971; Fax: +1-514-412-7938
| |
Collapse
|
15
|
Milk fat-globule epidermal growth factor 8: A potential Regulator of Cutaneous Wound Healing. Mol Biol Rep 2022; 49:8883-8893. [PMID: 35581508 DOI: 10.1007/s11033-022-07365-6] [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: 12/23/2021] [Revised: 03/02/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
Destroying the integrity of the skin may causes disability and even death from injury or illness. Wound healing is a core mechanism to maintain skin barrier function. Milk fat-globule epidermal growth factor 8 (MFG-E8) is a key factor in wound healing and is involved in regulating blood coagulation, mediating macrophage uptake of apoptotic cells, shifting macrophages from an inflammatory to an anti-inflammatory phenotype, promoting angiogenesis, enhancing vascular endothelial growth factor (VEGF) signaling, and assisting wound tissue perfusion. However, these abilities are dysregulated in pathological conditions, such as glucose disorders and ischemic injury. Restricted application of exogenous MFG-E8 can restore function and play a beneficial role in cutaneous wound healing.
Collapse
|
16
|
Kim H, Back JH, Han G, Lee SJ, Park YE, Gu MB, Yang Y, Lee JE, Kim SH. Extracellular vesicle-guided in situ reprogramming of synovial macrophages for the treatment of rheumatoid arthritis. Biomaterials 2022; 286:121578. [PMID: 35594838 DOI: 10.1016/j.biomaterials.2022.121578] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/02/2022] [Accepted: 05/11/2022] [Indexed: 12/01/2022]
Abstract
Activation state of synovial macrophages is significantly correlated with disease activity and severity of rheumatoid arthritis (RA) and provides valuable clues for RA treatment. Classically activated M1 macrophages in inflamed synovial joints secrete high levels of pro-inflammatory cytokines and chemokines, resulting in bone erosion and cartilage degradation. Herein, we propose extracellular vesicle (EV)-guided in situ macrophage reprogramming toward anti-inflammatory M2 macrophages as a novel RA treatment modality based on the immunotherapeutic concept of reestablishing M1-M2 macrophage equilibrium in synovial tissue. M2 macrophage-derived EVs (M2-EVs) were able to convert activated M1 into reprogrammed M2 (RM2) macrophages with extremely high efficiency (>90%), producing a distinct protein expression pattern characteristic of anti-inflammatory M2 macrophages. In particular, M2-EVs were enriched for proteins known to be involved in the generation and migration of M2 macrophages as well as macrophage reprogramming factors, allowing for rapid and efficient driving of macrophage polarization toward M2 phenotype. After administration of M2-EVs into the joint of a collagen-induced arthritis mouse model, the synovial macrophage polarization was significantly shifted from M1 to M2 phenotype, a process that benefited greatly from the long residence time (>3 days) of M2-EVs in the joint. This superb in situ macrophage-reprogramming ability of EVs resulted in decreased joint swelling, arthritic index score and synovial inflammation, with corresponding reductions in bone erosion and articular cartilage damage and no systemic toxicity. The anti-RA effects of M2-EVs were comparable to those of the conventional disease-modifying antirheumatic drug, Methotrexate, which causes a range of toxic adverse effects, including gastrointestinal mucosal injury. Overall, our EV-guided reprogramming strategy for in situ tuning of macrophage responses holds great promise for the development of anti-inflammatory therapeutics for the treatment of various inflammatory diseases in addition to RA.
Collapse
Affiliation(s)
- Hyosuk Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ji Hyun Back
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea; Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Geonhee Han
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Su Jin Lee
- Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Yae Eun Park
- Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Man Bock Gu
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Yoosoo Yang
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ji Eun Lee
- Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Sun Hwa Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
17
|
Geng R, Lin Y, Ji M, Chang Q, Li Z, Xu L, Zhang W, Lu J. MFG-E8 promotes tendon-bone healing by regualting macrophage efferocytosis and M2 polarization after anterior cruciate ligament reconstruction. J Orthop Translat 2022; 34:11-21. [PMID: 35615640 PMCID: PMC9109120 DOI: 10.1016/j.jot.2022.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/18/2022] [Accepted: 04/15/2022] [Indexed: 11/24/2022] Open
|
18
|
Regenerating Skeletal Muscle Compensates for the Impaired Macrophage Functions Leading to Normal Muscle Repair in Retinol Saturase Null Mice. Cells 2022; 11:cells11081333. [PMID: 35456012 PMCID: PMC9028072 DOI: 10.3390/cells11081333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/05/2022] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Skeletal muscle repair is initiated by local inflammation and involves the engulfment of dead cells (efferocytosis) by infiltrating macrophages at the injury site. Macrophages orchestrate the whole repair program, and efferocytosis is a key event not only for cell clearance but also for triggering the timed polarization of the inflammatory phenotype of macrophages into the healing one. While pro-inflammatory cytokines produced by the inflammatory macrophages induce satellite cell proliferation and differentiation into myoblasts, healing macrophages initiate the resolution of inflammation, angiogenesis, and extracellular matrix formation and drive myoblast fusion and myotube growth. Therefore, improper efferocytosis results in impaired muscle repair. Retinol saturase (RetSat) initiates the formation of various dihydroretinoids, a group of vitamin A derivatives that regulate transcription by activating retinoid receptors. Previous studies from our laboratory have shown that RetSat-null macrophages produce less milk fat globule-epidermal growth factor-factor-8 (MFG-E8), lack neuropeptide Y expression, and are characterized by impaired efferocytosis. Here, we investigated skeletal muscle repair in the tibialis anterior muscle of RetSat-null mice following cardiotoxin injury. Our data presented here demonstrate that, unexpectedly, several cell types participating in skeletal muscle regeneration compensate for the impaired macrophage functions, resulting in normal muscle repair in the RetSat-null mice.
Collapse
|
19
|
Brilland B, Laplante P, Thebault P, Geoffroy K, Brissette MJ, Latour M, Chassé M, Qi S, Hébert MJ, Cardinal H, Cailhier JF. MFG-E8 Reduces Aortic Intimal Proliferation in a Murine Model of Transplant Vasculopathy. Int J Mol Sci 2022; 23:ijms23084094. [PMID: 35456911 PMCID: PMC9027378 DOI: 10.3390/ijms23084094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 12/10/2022] Open
Abstract
Transplant vasculopathy is characterized by endothelial apoptosis, which modulates the local microenvironment. Milk fat globule epidermal growth factor 8 (MFG-E8), which is released by apoptotic endothelial cells, limits tissue damage and inflammation by promoting anti-inflammatory macrophages. We aimed to study its role in transplant vasculopathy using the murine aortic allotransplantation model. BALB/c mice were transplanted with fully mismatched aortic transplants from MFG-E8 knockout (KO) or wild type (WT) C57BL/6J mice. Thereafter, mice received MFG-E8 (or vehicle) injections for 9 weeks prior to histopathological analysis of allografts for intimal proliferation (hematoxylin and eosin staining) and leukocyte infiltration assessment (immunofluorescence). Phenotypes of blood leukocytes and humoral responses were also evaluated (flow cytometry and ELISA). Mice receiving MFG-E8 KO aortas without MFG-E8 injections had the most severe intimal proliferation (p < 0.001). Administration of MFG-E8 decreased intimal proliferation, especially in mice receiving MFG-E8 KO aortas. Administration of MFG-E8 also increased the proportion of anti-inflammatory macrophages among graft-infiltrating macrophages (p = 0.003) and decreased systemic CD4+ and CD8+ T-cell activation (p < 0.001). An increase in regulatory T cells occurred in both groups of mice receiving WT aortas (p < 0.01). Thus, the analarmin MFG-E8 appears to be an important protein for reducing intimal proliferation in this murine model of transplant vasculopathy. MFG-E8 effects are associated with intra-allograft macrophage reprogramming and systemic T-cell activation dampening.
Collapse
Affiliation(s)
- Benoit Brilland
- Service de Néphrologie-Dialyse-Transplantation, CHU d’Angers, F-49000 Angers, France;
- University of Angers, Université de Nantes, CHU Angers, INSERM, CRCINA, SFR ICAT, F-49000 Angers, France
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
| | - Patrick Laplante
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Pamela Thebault
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Karen Geoffroy
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Marie-Joëlle Brissette
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
| | - Mathieu Latour
- Department of Pathology, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 3J4, Canada;
| | - Michaël Chassé
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Department of Medicine, Critical Care Division, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 3J4, Canada
| | - Shijie Qi
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
| | - Marie-Josée Hébert
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Canadian National Transplant Research Program, Edmonton, AB T6G 2E1, Canada
- Department of Medicine, Renal Division, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 3J4, Canada
| | - Héloïse Cardinal
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Canadian National Transplant Research Program, Edmonton, AB T6G 2E1, Canada
- Department of Medicine, Renal Division, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 3J4, Canada
| | - Jean-François Cailhier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
- Canadian National Transplant Research Program, Edmonton, AB T6G 2E1, Canada
- Department of Medicine, Renal Division, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 3J4, Canada
- Correspondence: ; Tel.: +514-890-8000 (ext. 25971); Fax: +514-412-7938
| |
Collapse
|
20
|
Wu L, Kim Y, Seon GM, Choi SH, Park HC, Son G, Kim SM, Lim BS, Yang HC. Effects of RGD-grafted phosphatidylserine-containing liposomes on the polarization of macrophages and bone tissue regeneration. Biomaterials 2021; 279:121239. [PMID: 34753037 DOI: 10.1016/j.biomaterials.2021.121239] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022]
Abstract
Phosphatidylserine-containing liposomes (PSLs) can mimic the anti-inflammatory effects of apoptotic cells by binding to the phosphatidylserine receptors of macrophages. MGF-E8, a bridge molecule between phosphatidylserine and macrophages, can promote M2 polarization by activating macrophage integrin with its arginine-glycine-aspartic acid (RGD) motif. In this study, to mimic MGF-E8, PSLs presenting RGD peptide (RGD-PSLs) were prepared, and their immunomodulatory effects on macrophages and the bone tissue regeneration of rat calvarial defects were investigated. RGD peptides enhanced the phagocytosis of PSLs by macrophages, especially when the PSLs contained 3% RGD. RGD-PSLs were also more effective than PSLs for the suppression of lipopolysaccharide-induced gene expression of proinflammatory cytokines (i.e., IL-1β, IL-6, and TNF-α) as well as CD86 (M1 marker) expression. Furthermore, RGD promoted PSL-induced M2 polarization: 3%-RGD-PSLs significantly enhanced the mRNA expression of Arg-1, FIZZ1, and YM-1, as well as CD206 (M2 marker) expression. In a calvarial defect model, a significant increase in M2 with a decrease in M1 macrophages was observed with 3%-RGD-PSL treatment compared with the effects of PSLs alone. Finally, new bone formation was also accelerated by 3%-RGD-PSLs. Thus, these results suggest that the intensive immunomodulatory effect of RGD-PSLs led to the enhancement of bone tissue regeneration.
Collapse
Affiliation(s)
- Lele Wu
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, 101, Deahak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Yongjoon Kim
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, 101, Deahak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Gyeung Mi Seon
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, 101, Deahak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Sang Hoon Choi
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, 101, Deahak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Hee Chul Park
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, 101, Deahak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Gitae Son
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, 101, Deahak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Soung Min Kim
- Department of Oral and Maxillofacial Surgery, Dental Research Institute, School of Dentistry, Seoul National University, 101, Deahak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Bum-Soon Lim
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, 101, Deahak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Hyeong-Cheol Yang
- Department of Dental Biomaterials Science, Dental Research Institute, School of Dentistry, Seoul National University, 101, Deahak-ro, Jongno-gu, Seoul, 03080, South Korea.
| |
Collapse
|
21
|
Jha MK, Passero JV, Rawat A, Ament XH, Yang F, Vidensky S, Collins SL, Horton MR, Hoke A, Rutter GA, Latremoliere A, Rothstein JD, Morrison BM. Macrophage monocarboxylate transporter 1 promotes peripheral nerve regeneration after injury in mice. J Clin Invest 2021; 131:e141964. [PMID: 34491913 DOI: 10.1172/jci141964] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/02/2021] [Indexed: 12/28/2022] Open
Abstract
Peripheral nerves have the capacity for regeneration, but the rate of regeneration is so slow that many nerve injuries lead to incomplete recovery and permanent disability for patients. Macrophages play a critical role in the peripheral nerve response to injury, contributing to both Wallerian degeneration and nerve regeneration, and their function has recently been shown to be dependent on intracellular metabolism. To date, the impact of their intracellular metabolism on peripheral nerve regeneration has not been studied. We examined conditional transgenic mice with selective ablation in macrophages of solute carrier family 16, member 1 (Slc16a1), which encodes monocarboxylate transporter 1 (MCT1), and found that MCT1 contributed to macrophage metabolism, phenotype, and function, specifically in regard to phagocytosis and peripheral nerve regeneration. Adoptive cell transfer of wild-type macrophages ameliorated the impaired nerve regeneration in macrophage-selective MCT1-null mice. We also developed a mouse model that overexpressed MCT1 in macrophages and found that peripheral nerves in these mice regenerated more rapidly than in control mice. Our study provides further evidence that MCT1 has an important biological role in macrophages and that manipulations of macrophage metabolism can enhance recovery from peripheral nerve injuries, for which there are currently no approved medical therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Samuel L Collins
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Maureen R Horton
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Alban Latremoliere
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
22
|
Yamaguchi R, Guo X, Zheng J, Zhang J, Han J, Shioya A, Uramoto H, Mochizuki T, Yamada S. PRDX4 Improved Aging-Related Delayed Wound Healing in Mice. J Invest Dermatol 2021; 141:2720-2729. [PMID: 34029576 DOI: 10.1016/j.jid.2021.04.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/15/2021] [Accepted: 04/14/2021] [Indexed: 01/09/2023]
Abstract
Aging-related delayed wound healing is an issue of concern worldwide. Oxidative stress is involved in wound healing. Antioxidative enzymes have various roles in this process. PRDX4, a member of the PRDX family, is upregulated after injury. To investigate the effects of PRDX4 on aging-related wound healing, we subjected C57BL/6J (wild-type), human Prdx4‒transgenic (i.e., hPrdx4+/+), Prdx4-knockout (i.e., Prdx4-/y) mice of three age groups (young, adult, and aged) to skin wound formation. The overexpression of PRDX4 accelerated wound healing in adult and aged mice but not in young mice. Aged hPrdx4+/+ mice showed reduced oxidative stress and inflammation, lower numbers of neutrophils, increased macrophage infiltration, increased angiogenesis, and increased GF levels. The granulation tissue of adult and aged hPrdx4+/+ mice was richer in fibroblasts than that in the matched wild-type mice. PRDX4 deficiency was associated with mortality in adult and aged mice. In vitro, the overexpression of PRDX4 promoted the proliferation and migration of fibroblasts derived from adult or aged mice and made fibroblasts more resistant to the cytotoxicity of hydrogen peroxide. PRDX4 is essential for wound healing and can improve the healing process from multiple aspects, suggesting that it may be very beneficial to wound treatment, especially for the elderly.
Collapse
Affiliation(s)
- Reimon Yamaguchi
- Department of Pathology and Laboratory Medicine, School of Medicine, Kanazawa Medical University, Uchinada, Japan; Department of Dermatology, School of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Xin Guo
- Department of Pathology and Laboratory Medicine, School of Medicine, Kanazawa Medical University, Uchinada, Japan.
| | - Jianbo Zheng
- Department of Pathology and Laboratory Medicine, School of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Jing Zhang
- Department of Pathology and Laboratory Medicine, School of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Jia Han
- Department of Pathology and Laboratory Medicine, School of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Akihiro Shioya
- Department of Pathology and Laboratory Medicine, School of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Hidetaka Uramoto
- Department of Thoracic Surgery, School of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Takashi Mochizuki
- Department of Dermatology, School of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Sohsuke Yamada
- Department of Pathology and Laboratory Medicine, School of Medicine, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
23
|
Daghian SG, Farahpour MR, Jafarirad S. Biological fabrication and electrostatic attractions of new layered silver/talc nanocomposite using Lawsonia inermis L. and its chitosan-capped inorganic/organic hybrid: Investigation on acceleration of Staphylococcus aureus and Pseudomonas aeruginosa infected wound healing. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112294. [PMID: 34474845 DOI: 10.1016/j.msec.2021.112294] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 02/01/2023]
Abstract
In the present study, new-layered inorganic/organic hybrid of silver/talc nanocomposites (Ag/Tlc-NPs) and its chitosan-capped derivative (Ag/Tlc/Csn NCs) were biochemically synthesized utilizing Lawsonia inermis L. extract. The silver nanoparticles (Ag NPs) were synthesized employing green method on the exterior surface layer of talc mineral as a solid substrate. The negatively charged surface layer of talc might function as templates and can attract the chitosan cations from a solution to yield a layered hybrid structure, whose inorganic phase is formed by Si-O-Ag bonds. Our results revealed that Ag NPs were formed on the exterior surface of talc with a diameter with size of 124-215 nm. In addition, cytotoxicity, in vitro antibacterial activity, and clinical effects of wound-healing ointments containing talc were investigated. The results implied the successful synthesis of Ag/Tlc/Csn NCs using the extract. The structures were safe up to 0.50 mg/mL. In vitro studies confirmed antioxidant and antibacterial properties of Ag/Tlc/Csn NCs. In sum, our findings showed that the ointments improve wound healing process by inducing an anti-inflammatory M2 phenotype and bFGF, CD206, collagen1A, and IL-10 production that causes fibroblast migration and wound closure through influencing M2 macrophage. Ag/Tlc/Csn is suggested to be taken into consideration as a medical combination for improving infected wound healing and as a promising agent for clinical administration.
Collapse
Affiliation(s)
- Sajjad Ghanbarzadeh Daghian
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, Iran
| | - Mohammad Reza Farahpour
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, Iran.
| | - Saeed Jafarirad
- Department of organic and biochemistry, Faculty of chemistry, University of Tabriz, Tabriz, Iran; Research center of bioscience and biotechnology, University of Tabriz, Tabriz, Iran
| |
Collapse
|
24
|
Multivalent effects of heptamannosylated β-cyclodextrins on macrophage polarization to accelerate wound healing. Colloids Surf B Biointerfaces 2021; 208:112071. [PMID: 34461486 DOI: 10.1016/j.colsurfb.2021.112071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 02/05/2023]
Abstract
Macrophages have high plasticity and heterogeneity, and can suppress or mediate inflammation, depending on their cytokine secretion and phenotype. Regulating macrophage polarization into its M2 phenotype has a remarkable effect on inflammatory inhibition, inducing the regeneration of injured tissues. Here, we synthesized two heptamannosylated β-cyclodextrin derivatives (CD-Man7 and C3-CD-Man7) and demonstrated that their multivalent mannose ligands could induce M2 macrophage polarization to accelerate wound healing. Unlike hydrophilic CD-Man7, amphiphilic C3-CD-Man7 can self-assemble to form nanoparticles (CD-Man-NPs) in aqueous solution. Further, in vitro results confirmed that multivalent mannose ligands of either CD-Man7 or CD-Man-NPs stimulated RAW264.7 macrophages to differentiate into the M2 phenotype, which promoted fibroblast migration via a paracrine mechanism. In vivo results confirmed that both CD-Man7 and CD-Man-NPs reduced the inflammatory response in wound tissue and accelerated wound healing. The present study demonstrates multivalent effects of CD-Man7 and CD-Man-NPs on M2 macrophage polarization, indicating the therapeutic potential of these β-cyclodextrin glycoconjugates in the treatment of inflammatory diseases and wound healing.
Collapse
|
25
|
Chu C, Zhao X, Rung S, Xiao W, Liu L, Qu Y, Man Y. Application of biomaterials in periodontal tissue repair and reconstruction in the presence of inflammation under periodontitis through the foreign body response: Recent progress and perspectives. J Biomed Mater Res B Appl Biomater 2021; 110:7-17. [PMID: 34142745 DOI: 10.1002/jbm.b.34891] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
Periodontitis would cause dental tissue damage locally. Biomaterials substantially affect the surrounding immune microenvironment through treatment-oriented local inflammatory remodeling in dental periodontitis. This remodeling process is conducive to wound healing and periodontal tissue regeneration. Recent progress in understanding the foreign body response (FBR) and immune regulation, including cell heterogeneity, and cell-cell and cell-material interactions, has provided new insights into the design criteria for biomaterials applied in treatment of periodontitis. This review discusses recent progress and perspectives in the immune regulation effects of biomaterials to augment or reconstruct soft and hard tissue in an inflammatory microenvironment of periodontitis.
Collapse
Affiliation(s)
- Chenyu Chu
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiwen Zhao
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shengan Rung
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenlan Xiao
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Li Liu
- State Key Laboratory of Biotherapy and Laboratory, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Yili Qu
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Man
- Department of Oral Implantology & National Clinical Research Center for Oral Diseases & State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Zhang M, Liu H, Shi M, Zhang T, Lu W, Yang S, Cui Q, Li Z. Potential Mechanisms of the Impact of Hepatocyte Growth Factor Gene-Modified Tendon Stem Cells on Tendon Healing. Front Cell Dev Biol 2021; 9:659389. [PMID: 34222233 PMCID: PMC8250428 DOI: 10.3389/fcell.2021.659389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
The therapeutic impact of stem cells is potentially largely attributable to secretion of exosomes and soluble factors. The present study evaluates the impact of hepatocyte growth factor (HGF)-expressing tendon stem cells (TSCs) on tendon healing in a rat model. Patellar tendon TSCs were isolated and underwent transfection with lentiviral vectors containing HGF or green fluorescent protein (GFP) genes. In vivo, immunohistochemistry of tendons sampled 1 week postsurgery demonstrated that all stem cell-treated groups exhibited higher numbers of CD163+ M2 monocytes and IL-10+ cells (anti-inflammatory), and lower numbers of CCR7+ M1 monocytes and IL-6+ as well as COX-2+ cells (pro-inflammatory). Effects were most pronounced in the HGF-expressing TSCs (TSCs + HGF) treated group. Histology ± immunohistochemistry of tendons sampled 4 and 8 weeks postsurgery demonstrated that all stem cell-treated groups exhibited more ordered collagen fiber arrangement and lower levels of COLIII, α-SMA, TGF-β1, and fibronectin (proteins relevant to fibroscarring). Effects were most pronounced in the TSCs + HGF-treated group. For the in vitro study, isolated tendon fibroblasts pretreated with TGF-β1 to mimic the in vivo microenvironment of tendon injury were indirectly cocultured with TSCs, TSCs + GFP, or TSCs + HGF using a transwell system. Western blotting demonstrated that all stem cell types decreased TGF-β1-induced increases in fibroblast levels of COX-2, COLIII, and α-SMA, concomitant with decreased activation of major TGF-β1 signaling pathways (p38 MAPK, ERK1/2, but not Smad2/3). This effect was most pronounced for TSCs + HGF, which also decreased the TGF-β1-induced increase in activation of the Smad2/3 signaling pathway. The presence of specific inhibitors of these pathways during fibroblast TGF-β1 stimulation also attenuated increases in levels of COX-2, COLIII, and α-SMA. In conclusion, TSCs + HGF, which exhibit HGF overexpression, may promoting tendon healing via decreasing inflammation and fibrosis, perhaps partly via inhibiting TGF-β1-induced signaling. These findings identify a novel potential therapeutic strategy for tendon injuries, warranting additional research.
Collapse
Affiliation(s)
- Mingzhao Zhang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hengchen Liu
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Manyu Shi
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tingting Zhang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjun Lu
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shulong Yang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qingbo Cui
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhaozhu Li
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
27
|
PARP inhibitors promote stromal fibroblast activation by enhancing CCL5 autocrine signaling in ovarian cancer. NPJ Precis Oncol 2021; 5:49. [PMID: 34108603 PMCID: PMC8190269 DOI: 10.1038/s41698-021-00189-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 05/13/2021] [Indexed: 11/21/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) play significant roles in drug resistance through different ways. Antitumor therapies, including molecular targeted interventions, not only effect tumor cells but also modulate the phenotype and characteristics of CAFs, which can in turn blunt the therapeutic response. Little is known about how stromal fibroblasts respond to poly (ADP-ribose) polymerase inhibitors (PARPis) in ovarian cancer (OC) and subsequent effects on tumor cells. This is a study to evaluate how CAFs react to PARPis and their potential influence on PARPi resistance in OC. We discovered that OC stromal fibroblasts exhibited intrinsic resistance to PARPis and were further activated after the administration of PARPis. PARPi-challenged fibroblasts displayed a specific secretory profile characterized by increased secretion of CCL5, MIP-3α, MCP3, CCL11, and ENA-78. Mechanistically, increased secretion of CCL5 through activation of the NF-κB signaling pathway was required for PARPi-induced stromal fibroblast activation in an autocrine manner. Moreover, neutralizing CCL5 partly reversed PARPi-induced fibroblast activation and boosted the tumor inhibitory effect of PARPis in both BRCA1/2-mutant and BRCA1/2-wild type xenograft models. Our study revealed that PARPis could maintain and improve stromal fibroblast activation involving CCL5 autocrine upregulation. Targeting CCL5 might offer a new treatment modality in overcoming the reality of PARPi resistance in OC.
Collapse
|
28
|
Lu Y, Liu L, Pan J, Luo B, Zeng H, Shao Y, Zhang H, Guan H, Guo D, Zeng C, Zhang R, Bai X, Zhang H, Cai D. MFG-E8 regulated by miR-99b-5p protects against osteoarthritis by targeting chondrocyte senescence and macrophage reprogramming via the NF-κB pathway. Cell Death Dis 2021; 12:533. [PMID: 34031369 PMCID: PMC8144578 DOI: 10.1038/s41419-021-03800-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/27/2022]
Abstract
Milk fat globule-epidermal growth factor (EGF) factor 8 (MFG-E8), as a necessary bridging molecule between apoptotic cells and phagocytic cells, has been widely studied in various organs and diseases, while the effect of MFG-E8 in osteoarthritis (OA) remains unclear. Here, we identified MFG-E8 as a key factor mediating chondrocyte senescence and macrophage polarization and revealed its role in the pathology of OA. We found that MFG-E8 expression was downregulated both locally and systemically as OA advanced in patients with OA and in mice after destabilization of the medial meniscus surgery (DMM) to induce OA. MFG-E8 loss caused striking progressive articular cartilage damage, synovial hyperplasia, and massive osteophyte formation in OA mice, which was relieved by intra-articular administration of recombinant mouse MFG-E8 (rmMFG-E8). Moreover, MFG-E8 restored chondrocyte homeostasis, deferred chondrocyte senescence and reprogrammed macrophages to the M2 subtype to alleviate OA. Further studies showed that MFG-E8 was inhibited by miR-99b-5p, expression of which was significantly upregulated in OA cartilage, leading to exacerbation of experimental OA partially through activation of NF-κB signaling in chondrocytes. Our findings established an essential role of MFG-E8 in chondrocyte senescence and macrophage reprogramming during OA, and identified intra-articular injection of MFG-E8 as a potential therapeutic target for OA prevention and treatment.
Collapse
Affiliation(s)
- Yuheng Lu
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Liangliang Liu
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Jianying Pan
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Bingsheng Luo
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Hua Zeng
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Yan Shao
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Hongbo Zhang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Hong Guan
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Dong Guo
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Chun Zeng
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Rongkai Zhang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Xiaochun Bai
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Haiyan Zhang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China. .,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China. .,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.
| | - Daozhang Cai
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China. .,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China. .,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.
| |
Collapse
|
29
|
Datta R, Lizama CO, Soltani AK, Mckleroy W, Podolsky MJ, Yang CD, Huynh TL, Cautivo KM, Wang B, Koliwad SK, Abumrad NA, Atabai K. Autoregulation of insulin receptor signaling through MFGE8 and the αvβ5 integrin. Proc Natl Acad Sci U S A 2021; 118:e2102171118. [PMID: 33903257 PMCID: PMC8106306 DOI: 10.1073/pnas.2102171118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The role of integrins, in particular αv integrins, in regulating insulin resistance is incompletely understood. We have previously shown that the αvβ5 integrin ligand milk fat globule epidermal growth factor like 8 (MFGE8) regulates cellular uptake of fatty acids. In this work, we evaluated the impact of MFGE8 on glucose homeostasis. We show that acute blockade of the MFGE8/β5 pathway enhances while acute augmentation dampens insulin-stimulated glucose uptake. Moreover, we find that insulin itself induces cell-surface enrichment of MFGE8 in skeletal muscle, which then promotes interaction between the αvβ5 integrin and the insulin receptor leading to dampening of skeletal-muscle insulin receptor signaling. Blockade of the MFGE8/β5 pathway also enhances hepatic insulin sensitivity. Our work identifies an autoregulatory mechanism by which insulin-stimulated signaling through its cognate receptor is terminated through up-regulation of MFGE8 and its consequent interaction with the αvβ5 integrin, thereby establishing a pathway that can potentially be targeted to improve insulin sensitivity.
Collapse
Affiliation(s)
- Ritwik Datta
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
| | - Carlos O Lizama
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
| | - Amin K Soltani
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Lung Biology Center, University of California, San Francisco, CA 94158
| | - William Mckleroy
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Lung Biology Center, University of California, San Francisco, CA 94158
- Divisions of Pulmonary and Critical Care and Endocrinology, Department of Medicine, University of California, San Francisco, CA 94143
| | - Michael J Podolsky
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Divisions of Pulmonary and Critical Care and Endocrinology, Department of Medicine, University of California, San Francisco, CA 94143
| | - Christopher D Yang
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
| | - Tony L Huynh
- Department of Radiology and Biomedical imaging, University of California, San Francisco, CA 94107
| | - Kelly M Cautivo
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Biao Wang
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158
- Department of Physiology, University of California, San Francisco, CA 94158
| | - Suneil K Koliwad
- Divisions of Pulmonary and Critical Care and Endocrinology, Department of Medicine, University of California, San Francisco, CA 94143
- Diabetes Center, University of California, San Francisco, CA 94143
| | - Nada A Abumrad
- Diabetes Research Center, Department of Medicine and Cell Biology, Washington University in St. Louis, St. Louis, MO 63110
| | - Kamran Atabai
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158;
- Lung Biology Center, University of California, San Francisco, CA 94158
- Divisions of Pulmonary and Critical Care and Endocrinology, Department of Medicine, University of California, San Francisco, CA 94143
- Department of Physiology, University of California, San Francisco, CA 94158
| |
Collapse
|
30
|
Choodari Gharehpapagh A, Farahpour MR, Jafarirad S. The biological synthesis of gold/perlite nanocomposite using Urtica dioica extract and its chitosan-capped derivative for healing wounds infected with methicillin-resistant Staphylococcus aureus. Int J Biol Macromol 2021; 183:447-456. [PMID: 33932414 DOI: 10.1016/j.ijbiomac.2021.04.150] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/14/2021] [Accepted: 04/24/2021] [Indexed: 01/14/2023]
Abstract
The preparation of ointments from natural compounds is essential for accelerating infected wounds. This study investigated the effects of topical uses of gold nanoparticles (Au)/perlite (Au/Perl) nanocomposites (NCs) by the help of Urtica dioica extract and its chitosan-capped derivative (Chit) on methicillin-resistant Staphylococcus aureus (MRSA)-infected wound healing in a mouse model. Furthermore, Au/Perl/Chit nanocomposite was prepared using protonated chitosan solution. The physicochemical properties of the as-synthesized nanocomposites were also investigated. The effects of Au/Perl/Chit NC were assessed by antibacterial, histopathological parameters as well as molecular evaluations. Then, they were compared with synthetic agent of mupirocin. The results revealed that Au/Perl NC was mesoporous and spherical in a range of 13-15 nm. Topical administration of Au/Perl/Chit ointment accelerated wound healing by reducing bacteria colonization and wound rate enhancing collagen biosynthesis and re-epithelialization, the expressions of IL-10, PI3K, AKT, bFGF, and COL1A genes, which is in agreement with the obtained results for mupirocin. In conclusion, the results strongly demonstrated that administration of ointments prepared from Au/Perl and Au/Perl/Chit nanocomposites stimulates MRSA-infected wound healing by decreasing the length of healing time and regulating PI3K/AKT/bFGF signaling pathway and is a promising candidate in stimulating MRSA-infected wound regeneration.
Collapse
Affiliation(s)
- Aryan Choodari Gharehpapagh
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, Iran
| | - Mohammad Reza Farahpour
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, Iran.
| | - Saeed Jafarirad
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran; Research Institute of Bioscience and Biotechnology, University of Tabriz, Tabriz, Iran
| |
Collapse
|
31
|
Huang CY, Vesvoranan O, Yin X, Montoya A, Londono V, Sawatari Y, Garcia-Godoy F. Anti-Inflammatory Effects of Conditioned Medium of Periodontal Ligament-Derived Stem Cells on Chondrocytes, Synoviocytes, and Meniscus Cells. Stem Cells Dev 2021; 30:537-547. [PMID: 33757298 DOI: 10.1089/scd.2021.0010] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis (OA) is the most common type of arthritis, afflicting millions of people in the world. Elevation of inflammatory mediators and enzymatic matrix destruction is often associated with OA. Therefore, the objective of this study was to investigate the effects of conditioned medium from periodontal ligament-derived stem cells (PDLSCs) on inflammatory and catabolic gene expressions of chondrocytes, synoviocytes, and meniscus cells under in vitro inflammatory condition. Stem cells were isolated from human periodontal ligaments. Conditioned medium was collected and concentrated 20 × . Chondrocytes, synoviocytes, and meniscus cells were isolated from pig knees and divided into four experimental groups: serum-free media, serum-free media+interleukin-1β (IL-1β) (10 ng/mL), conditioned media (CM), and CM+IL-1β. Protein content and extracellular vesicle (EV) miRNAs of CM were analyzed by liquid chromatography-tandem mass spectrometry and RNA sequencing, respectively. It was found that the IL-1β treatment upregulated the expression of IL-1β, tumor necrosis factor-α (TNF-α), MMP-13, and ADAMTS-4 genes in the three cell types, whereas PDLSC-conditioned medium prevented the upregulation of gene expression by IL-1β in all three cell types. This study also found that there was consistency in anti-inflammatory effects of PDLSC CM across donors and cell subcultures, while PDLSCs released several anti-inflammatory factors and EV miRNAs at high levels. OA has been suggested as an inflammatory disease in which all intrasynovial tissues are involved. PDLSC-conditioned medium is a cocktail of trophic factors and EV miRNAs that could mediate different inflammatory processes in various tissues in the joint. Introducing PDLSC-conditioned medium to osteoarthritic joints could be a potential treatment to prevent OA progression by inhibiting inflammation.
Collapse
Affiliation(s)
- Chun-Yuh Huang
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Oraya Vesvoranan
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Xue Yin
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Amanda Montoya
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Valeria Londono
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Yoh Sawatari
- Division of Oral and Maxillofacial Surgery, University of Miami/Jackson Memorial Hospital, Miami, Florida, USA
| | - Franklin Garcia-Godoy
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
32
|
Romana-Souza B, Chen L, Leonardo TR, Chen Z, DiPietro LA. Dermal fibroblast phagocytosis of apoptotic cells: A novel pathway for wound resolution. FASEB J 2021; 35:e21443. [PMID: 33749877 PMCID: PMC8670562 DOI: 10.1096/fj.202002078r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/20/2021] [Accepted: 02/01/2021] [Indexed: 01/23/2023]
Abstract
The effective clearance of apoptotic cells is an essential step in the resolution of healing wounds. In particular, blood vessel regression during wound resolution produces a significant number of apoptotic endothelial cells (ApoEC) that must be cleared. In considering the fate of ApoEC and the presence of fibroblasts during wound resolution, we hypothesized that fibroblasts might serve as phagocytes involved in endothelial cell removal. The current study investigated whether dermal fibroblasts engulf ApoEC, whether this uptake alters the phenotype of dermal fibroblasts, and the biological molecules involved. In both in vitro and in vivo studies, following ApoEC engulfment, fibroblasts acquired a pro-healing phenotype (increased cell migration, contractility, α-smooth muscle actin expression, and collagen deposition). In addition, fibroblast uptake of ApoEC was shown to be mediated in part by the milk fat globule-EGF factor 8 protein/integrin αv β5 pathway. Our study demonstrates a novel function of fibroblasts in the clearance of ApoEC and suggests that this capability has significant implications for tissue repair and fibrosis.
Collapse
Affiliation(s)
- Bruna Romana-Souza
- Tissue Repair Laboratory, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Trevor R. Leonardo
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zhenlong Chen
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Luisa A. DiPietro
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
33
|
Small extracellular vesicles derived from interferon-γ pre-conditioned mesenchymal stromal cells effectively treat liver fibrosis. NPJ Regen Med 2021; 6:19. [PMID: 33785758 PMCID: PMC8010072 DOI: 10.1038/s41536-021-00132-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are used for ameliorating liver fibrosis and aiding liver regeneration after cirrhosis; Here, we analyzed the therapeutic potential of small extracellular vesicles (sEVs) derived from interferon-γ (IFN-γ) pre-conditioned MSCs (γ-sEVs). γ-sEVs effectively induced anti-inflammatory macrophages with high motility and phagocytic abilities in vitro, while not preventing hepatic stellate cell (HSC; the major source of collagen fiber) activation in vitro. The proteome analysis of MSC-derived sEVs revealed anti-inflammatory macrophage inducible proteins (e.g., annexin-A1, lactotransferrin, and aminopeptidase N) upon IFN-γ stimulation. Furthermore, by enabling CX3CR1+ macrophage accumulation in the damaged area, γ-sEVs ameliorated inflammation and fibrosis in the cirrhosis mouse model more effectively than sEVs. Single cell RNA-Seq analysis revealed diverse effects, such as induction of anti-inflammatory macrophages and regulatory T cells, in the cirrhotic liver after γ-sEV administration. Overall, IFN-γ pre-conditioning altered sEVs resulted in efficient tissue repair indicating a new therapeutic strategy.
Collapse
|
34
|
Bu HF, Subramanian S, Geng H, Wang X, Liu F, Chou PM, Du C, De Plaen IG, Tan XD. MFG-E8 Plays an Important Role in Attenuating Cerulein-Induced Acute Pancreatitis in Mice. Cells 2021; 10:728. [PMID: 33806041 PMCID: PMC8064467 DOI: 10.3390/cells10040728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 11/23/2022] Open
Abstract
Milk fat globule-EGF factor 8 (MFG-E8) is a secreted glycoprotein that regulates tissue homeostasis, possesses potent anti-inflammatory properties, and protects against tissue injury. The human pancreas expresses MFG-E8; however, the role of MFG-E8 in the pancreas remains unclear. We examined the expression of MFG-E8 in the pancreas at baseline and during cerulein-induced acute pancreatitis in mice and determined whether MFG-E8 attenuates the progression of pancreatitis, a serious inflammatory condition that can be life-threatening. We administered cerulein to wild-type (WT) and Mfge8 knockout (KO) mice to induce pancreatitis. Immunoblot analysis showed that MFG-E8 is constitutively expressed in the murine pancreas and is increased in mice with cerulein-induced acute pancreatitis. In situ hybridization revealed that ductal epithelial cells in the mouse pancreas express Mfge8 transcripts at baseline. During pancreatitis, Mfge8 transcripts were abundantly expressed in acinar cells and endothelial cells in addition to ductal epithelial cells. Knocking out Mfge8 in mice exacerbated the severity of cerulein-induced acute pancreatitis and delayed its resolution. In contrast, administration of recombinant MFG-E8 attenuated cerulein-induced acute pancreatitis and promoted repair of pancreatic injury in Mfge8 KO mice. Taken together, our study suggests that MFG-E8 protects the pancreas against inflammatory injury and promotes pancreatic tissue repair. MFG-E8 may represent a novel therapeutic target in acute pancreatitis.
Collapse
Affiliation(s)
- Heng-Fu Bu
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.-F.B.); (S.S.); (H.G.); (X.W.); (F.L.); (C.D.)
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Saravanan Subramanian
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.-F.B.); (S.S.); (H.G.); (X.W.); (F.L.); (C.D.)
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Hua Geng
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.-F.B.); (S.S.); (H.G.); (X.W.); (F.L.); (C.D.)
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Xiao Wang
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.-F.B.); (S.S.); (H.G.); (X.W.); (F.L.); (C.D.)
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Fangyi Liu
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.-F.B.); (S.S.); (H.G.); (X.W.); (F.L.); (C.D.)
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Pauline M. Chou
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Chao Du
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.-F.B.); (S.S.); (H.G.); (X.W.); (F.L.); (C.D.)
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Isabelle G. De Plaen
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
- Division of Neonatology, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Xiao-Di Tan
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.-F.B.); (S.S.); (H.G.); (X.W.); (F.L.); (C.D.)
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
- Department of Research & Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
35
|
Toita R, Shimizu E, Murata M, Kang JH. Protective and healing effects of apoptotic mimic-induced M2-like macrophage polarization on pressure ulcers in young and middle-aged mice. J Control Release 2021; 330:705-714. [DOI: 10.1016/j.jconrel.2020.12.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/16/2020] [Accepted: 12/28/2020] [Indexed: 12/29/2022]
|
36
|
DiPietro LA, Wilgus TA, Koh TJ. Macrophages in Healing Wounds: Paradoxes and Paradigms. Int J Mol Sci 2021; 22:950. [PMID: 33477945 PMCID: PMC7833402 DOI: 10.3390/ijms22020950] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages are prominent cells in normally healing adult skin wounds, yet their exact functions and functional significance to healing outcomes remain enigmatic. Many functional attributes are ascribed to wound macrophages, including host defense and support of the proliferation of new tissue to replace that lost by injury. Indeed, the depletion of macrophages is unmistakably detrimental to normal skin healing in adult mammals. Yet in certain systems, dermal wounds seem to heal well with limited or even no functional macrophages, creating an apparent paradox regarding the function of this cell in wounds. Recent advances in our understanding of wound macrophage phenotypes, along with new information about cellular plasticity in wounds, may provide some explanation for the apparently contradictory findings and suggest new paradigms regarding macrophage function in wounds. Continued study of this remarkable cell is needed to develop effective therapeutic options to improve healing outcomes.
Collapse
Affiliation(s)
- Luisa A. DiPietro
- College of Dentistry, University of Illinois at Chicago, 801 S. Paulina, Chicago, IL 60612, USA
| | - Traci A. Wilgus
- Department of Pathology, The Ohio State University, 129 Hamilton Hall, 1645 Neil Ave, Columbus, OH 43210, USA;
| | - Timothy J. Koh
- College of Applied Health Sciences, University of Illinois at Chicago, 1919 W. Taylor, Chicago, IL 60612, USA;
| |
Collapse
|
37
|
Zhang M, Liu H, Cui Q, Han P, Yang S, Shi M, Zhang T, Zhang Z, Li Z. Tendon stem cell-derived exosomes regulate inflammation and promote the high-quality healing of injured tendon. Stem Cell Res Ther 2020; 11:402. [PMID: 32943109 PMCID: PMC7499865 DOI: 10.1186/s13287-020-01918-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/30/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Tendon stem cells (TSCs) have been reported to hold promises for tendon repair and regeneration. However, less is known about the effects of exosomes derived from TSCs. Therefore, we aimed to clarify the healing effects of TSC-derived exosomes (TSC-Exos) on tendon injury. METHODS The Achilles tendons of Sprague-Dawley male rats were used for primary culture of TSCs and tenocytes, and exosomes were isolated from TSCs. The proliferation of tenocytes induced by TSC-Exos was analyzed using an EdU assay; cell migration was measured by cell scratch and transwell assays. We used western blot to analyze the role of the PI3K/AKT and MAPK/ERK1/2 signaling pathways. In vivo, Achilles tendon injury models were created in Sprague-Dawley rats. Rats (n = 54) were then randomly assigned to three groups: the TSC-Exos group, the GelMA group, and the control group. We used immunofluorescence to detect changes in the expression of inflammatory and apoptotic markers at 1 week after surgery. Histology and changes in expression of extracellular matrix (ECM)-related indices were assessed by hematoxylin-eosin (H&E) staining and immunohistochemistry at 2 and 8 weeks. The collagen fiber diameter of the healing tendon was analyzed at 8 weeks by transmission electron microscopy (TEM). RESULTS TSC-Exos were taken up by tenocytes, which promoted the proliferation and migration of cells in a dose-dependent manner; this process may depend on the activation of the PI3K/AKT and MAPK/ERK1/2 signaling pathways. At 1 week after surgery, we found that inflammation and apoptosis were significantly suppressed by TSC-Exos. At 2 and 8 weeks, tendons treated with TSC-Exos showed more continuous and regular arrangement in contrast to disorganized tendons in the GelMA and control groups, and TSC-Exos may help regulate ECM balance and inhibited scar formation. Further, at 8 weeks, the TSC-Exos group had a larger diameter of collagen compared to the control group. CONCLUSIONS Our data suggest that TSC-Exos could promote high-quality healing of injured tendon, which may be a promising therapeutic approach for tendon injury.
Collapse
Affiliation(s)
- Mingzhao Zhang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin, 150001, China
| | - Hengchen Liu
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin, 150001, China
| | - Qingbo Cui
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin, 150001, China
| | - Peilin Han
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin, 150001, China
| | - Shulong Yang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin, 150001, China
| | - Manyu Shi
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin, 150001, China
| | - Tingting Zhang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin, 150001, China
| | - Zenan Zhang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin, 150001, China
| | - Zhaozhu Li
- Department of Pediatric Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin, 150001, China.
| |
Collapse
|
38
|
Huang W, Jiao J, Liu J, Huang M, Hu Y, Ran W, Yan L, Xiong Y, Li M, Quan Z, Rao Y, Chen J, Huang Y, Zhang D. MFG-E8 accelerates wound healing in diabetes by regulating "NLRP3 inflammasome-neutrophil extracellular traps" axis. Cell Death Discov 2020; 6:84. [PMID: 32963812 PMCID: PMC7484765 DOI: 10.1038/s41420-020-00318-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/06/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
Sustained activation of NLRP3 inflammasome and release of neutrophil extracellular traps (NETs) impair wound healing of diabetic foot ulcers (DFUs). Our previous study reported that milk fat globule epidermal growth factor VIII (MFG-E8) attenuates tissue damage in systemic lupus erythematosus. However, the functional effect of MFG-E8 on "NLRP3 inflammasome-NETs" inflammatory loop in wound healing of diabetes is not completely elucidated. In this study, neutrophils from DFU patients are susceptible to undergo NETosis, releasing more NETs. The circulating levels of NET components neutrophil elastase and proteinase 3 and inflammatory cytokines IL-1β and IL-18 were significantly elevated in DFU patients compared with healthy controls or diabetic patients, in spite of higher levels of MFG-E8 in DFU patients. In Mfge8-/- diabetic mice, skin wound displayed exaggerated inflammatory response, including leukocyte infiltration, excessive activation of NLRP3 inflammasome (release of higher IL-1β, IL-18, and TNF-α), largely lodged NETs, resulting in poor angiogenesis and wound closure. When stimulated with high-dose glucose or IL-18, MFG-E8-deficient neutrophils release more NETs than WT neutrophils. After administration of recombinant MFG-E8, IL-18-primed NETosis of WT or Mfge8-/- neutrophils was significantly inhibited. Furthermore, NET and mCRAMP (component of NETs, the murine equivalent of cathelicidin LL-37 in human)-mediated activation of NLRP3 inflammasome and production of IL-1β/IL-18 were significantly elevated in Mfge8-/- macrophages compared with WT macrophages, which were also significantly dampened by the administration of rmMFG-E8. Therefore, our study demonstrated that as inhibitor of the "NLRP3 inflammasome-NETs" inflammatory loop, exogenous rMFG-E8 improves angiogenesis and accelerates wound healing, highlighting possible therapeutic potential for DFUs.
Collapse
Affiliation(s)
- Wei Huang
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Jinyu Jiao
- Department of Cardiac Function, Wuhan Asian Heart Hospital, Wuhan University of Science and Technology, Wuhan, Hubei China
| | - Ju Liu
- Department of Geriatrics, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Meng Huang
- Department of Dermatology, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Yanyan Hu
- Department of Dermatology, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Wenzhuo Ran
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Li Yan
- Clinical Laboratory of Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yin Xiong
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Mei Li
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Ziyao Quan
- Clinical Laboratory of Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yahua Rao
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Jiayi Chen
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Yan Huang
- Department of Laboratory Medicine, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Dongxin Zhang
- Department of Clinical Laboratory, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| |
Collapse
|
39
|
De Maeyer RPH, van de Merwe RC, Louie R, Bracken OV, Devine OP, Goldstein DR, Uddin M, Akbar AN, Gilroy DW. Blocking elevated p38 MAPK restores efferocytosis and inflammatory resolution in the elderly. Nat Immunol 2020; 21:615-625. [PMID: 32251403 PMCID: PMC7983074 DOI: 10.1038/s41590-020-0646-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 03/02/2020] [Indexed: 01/26/2023]
Abstract
Increasing age alters innate immune-mediated responses; however, the mechanisms underpinning these changes in humans are not fully understood. Using a human dermal model of acute inflammation, we found that, although inflammatory onset is similar between young and elderly individuals, the resolution phase was substantially impaired in elderly individuals. This arose from a reduction in T cell immunoglobulin mucin receptor-4 (TIM-4), a phosphatidylserine receptor expressed on macrophages that enables the engulfment of apoptotic bodies, so-called efferocytosis. Reduced TIM-4 in elderly individuals was caused by an elevation in macrophage p38 mitogen-activated protein kinase (MAPK) activity. Administering an orally active p38 inhibitor to elderly individuals rescued TIM-4 expression, cleared apoptotic bodies and restored a macrophage resolution phenotype. Thus, inhibiting p38 in elderly individuals rejuvenated their resolution response to be more similar to that of younger people. This is the first resolution defect identified in humans that has been successfully reversed, thereby highlighting the tractability of targeting pro-resolution biology to treat diseases driven by chronic inflammation.
Collapse
Affiliation(s)
| | | | - Rikah Louie
- Division of Medicine, University College London, London, UK
| | | | - Oliver P Devine
- Division of Infection and Immunity, University College London, London, UK
| | - Daniel R Goldstein
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mohib Uddin
- Respiratory Global Medicines Development, AstraZeneca, Gothenburg, Sweden
| | - Arne N Akbar
- Division of Infection and Immunity, University College London, London, UK
| | - Derek W Gilroy
- Division of Medicine, University College London, London, UK.
| |
Collapse
|
40
|
A Comparison of the Wound Healing Efficacy of Trolamine Emulsion, Manuka Gel, and Polymyxin-Bacitracin Ointment. Adv Skin Wound Care 2020; 33:217-220. [DOI: 10.1097/01.asw.0000655468.08618.fc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
41
|
Comparative Analysis of KGF-2 and bFGF in Prevention of Excessive Wound Healing and Scar Formation in a Corneal Alkali Burn Model. Cornea 2020; 38:1430-1437. [PMID: 31490279 DOI: 10.1097/ico.0000000000002134] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE Basic fibroblast growth factor (bFGF) is an effective drug for corneal injury. However, the explicit role of bFGF in corneal scar formation still remains unclear. Keratinocyte growth factor-2 (KGF-2) is associated with the treatment of wound healing. We aimed to compare the efficacy of bFGF and KGF-2 in prevention of excessive wound healing and consequent scar formation in a rat alkali burn model, which provides important clues on the significance of KGF-2 to be developed as a new drug for such injuries. METHODS The epithelial defect area was evaluated using fluorescein sodium at a concentration of 0.5%. The therapeutic effect of KGF-2 and bFGF on proliferation of rabbit corneal fibroblasts (RCFs) was evaluated by methylthiazoletetrazolium. RCF migration assays were performed with a modified scratch method. Activation of mitogen-activated protein kinase (MAPK) was evaluated by Western blot with specific antibodies. RESULTS All corneal wounds treated with KGF-2 were found closed within 7 days; however, the wounds treated with bFGF or phosphate buffer saline (PBS) required 14 days to close. RCFs treated with KGF-2 or bFGF showed similar dose-dependent proliferation. The KGF-2 group significantly promoted cell migration compared with the bFGF group. The KGF-2 group showed less expression of α-smooth muscle actin (SMA) and numbers of myofibroblasts compared with the bFGF group. Our findings suggested identification of cascade reaction of extracellular regulated protein kinases (ERK)1/2 and p38 signals in KGF-2- and bFGF-induced proliferation and migration of RCFs. In addition, KGF-2 showed stronger effects during ERK1/2 and p38 phosphorylation in methylthiazoletetrazolium proliferation assay and scratch migration assay. CONCLUSIONS KGF-2 exhibited better effects than bFGF in reepithelialization, acceleration of migration, and reduction of scar formation, which has potential to become a new drug to cure corneal injury.
Collapse
|
42
|
Zhu Y, Yao Z, Liu Y, Zhang W, Geng L, Ni T. Incorporation of ROS-Responsive Substance P-Loaded Zeolite Imidazolate Framework-8 Nanoparticles into a Ca 2+-Cross-Linked Alginate/Pectin Hydrogel for Wound Dressing Applications. Int J Nanomedicine 2020; 15:333-346. [PMID: 32021183 PMCID: PMC6980861 DOI: 10.2147/ijn.s225197] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 01/02/2020] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Wound healing, especially of extensive full-thickness wounds, is one of the most difficult problems in clinical studies. In this study, we prepared a novel substance P (SP)-delivery system using zeolite imidazolate framework-8 (ZIF-8) nanoparticles. METHODS We synthesized ZIF-8 nanoparticles using a modified biomimetic mineralization method. We then coated SP-loaded ZIF-8 nanoparticles (SP@ZIF-8) with polyethylene glycol-thioketal (PEG-TK) to fabricate SP@ZIF-8-PEG-TK nanoparticles, and encapsulated them in injectable hydrogel composed of sodium alginate and pectin and cross-linked using calcium chloride. The final hydrogel wound dressing containing SP@ZIF-8-PEG-TK nanoparticles was called SP@ZIF-8-PEG-TK@CA. RESULTS The fabricated ZIF-8 nanoparticles had high SP-loading efficiency. SP-release assay showed that the SP@ZIF-8-PEG-TK nanoparticles maintained drug activity and showed responsive release under stimulation by reactive oxygen species. The SP@ZIF-8-PEG-TK nanoparticles promoted proliferation of human dermal fibroblasts, up-regulated expression levels of inflammation-related genes in macrophages, and exhibited favorable cytocompatibility in vitro. Full-thickness excision wound models in vivo confirmed that SP@ZIF-8-PEG-TK@CA dressings had excellent wound-healing efficacy by promoting an early inflammatory response and subsequent M2 macrophage polarization in the wound-healing process. CONCLUSION In conclusion, these findings indicated that SP@ZIF-8-PEG-TK@CA dressings might be useful for wound dressing applications in the clinic.
Collapse
Affiliation(s)
- Yiming Zhu
- Department of General Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| | - Zuochao Yao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| | - Yushu Liu
- Department of Burns and Plastic Surgery, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China
| | - Wen Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| | - Lele Geng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| | - Tao Ni
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Huangpu, Shanghai, China
| |
Collapse
|
43
|
Wang J, Zuo Y, Zhuang K, Luo K, Yan X, Li J, Zhang JH, Liu F. Recombinant Human Milk Fat Globule-Epidermal Growth Factor 8 Attenuates Microthrombosis after Subarachnoid Hemorrhage in Rats. J Stroke Cerebrovasc Dis 2019; 29:104536. [PMID: 31883781 DOI: 10.1016/j.jstrokecerebrovasdis.2019.104536] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/28/2019] [Accepted: 11/09/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Microthrombosis after subarachnoid hemorrhage has an adverse effect on prognosis. Milk fat globule-epidermal growth factor 8 promotes phagocytosis of phagocytic cells and may reduce microthrombosis. This study investigated the effects of recombinant human milk fat globule-epidermal growth factor 8 on microthrombosis and neurological function after subarachnoid hemorrhage. METHODS Rats subarachnoid hemorrhage model was induced by intravascular puncture method. Western blot was performed to measure the expression of endogenous milk fat globule-epidermal growth factor 8 after subarachnoid hemorrhage. Microthrombosis was quantified by microthrombi count using immunohistochemistry and immunofluorescence. The neuroprotective effect of recombinant human milk fat globule-epidermal growth factor 8 administration was evaluated by modified Garcia score, beam balance, Rotarod test, and Morris water maze. RESULTS Endogenous milk fat globule-epidermal growth factor 8 protein level increased after subarachnoid hemorrhage. Microthrombosis was significantly increased in subarachnoid hemorrhage rats brain, while recombinant human milk fat globule-epidermal growth factor 8 dramatically reduced microthrombosis as well as improve short- and long- term neurobehavior after subarachnoid hemorrhage. CONCLUSIONS Recombinant human milk fat globule-epidermal growth factor 8 reduces microthrombosis and improves neurological function after subarachnoid hemorrhage, which may be an effective strategy for treating subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Jikai Wang
- Department of Neurosurgery, Third XiangYa Hospital, Central South University, Changsha, Hunan, China
| | - Yuchun Zuo
- Department of Neurosurgery, XiangYa Hospital, Central South University, Changsha, Hunan, China
| | - Kai Zhuang
- Department of Neurosurgery, Third XiangYa Hospital, Central South University, Changsha, Hunan, China
| | - Kui Luo
- Department of Neurosurgery, Third XiangYa Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxin Yan
- Department of Anatomy and Neurobiology, XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Jianming Li
- Neuroscience Research Center, Changsha Medical University, Changsha, Hunan, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California.
| | - Fei Liu
- Department of Neurosurgery, Third XiangYa Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
44
|
Kim H, Wang SY, Kwak G, Yang Y, Kwon IC, Kim SH. Exosome-Guided Phenotypic Switch of M1 to M2 Macrophages for Cutaneous Wound Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900513. [PMID: 31637157 PMCID: PMC6794619 DOI: 10.1002/advs.201900513] [Citation(s) in RCA: 304] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/09/2019] [Indexed: 05/10/2023]
Abstract
Macrophages (Mϕs) critically contribute to wound healing by coordinating inflammatory, proliferative, and angiogenic processes. A proper switch from proinflammatory M1 to anti-inflammatory M2 dominant Mϕs accelerates the wound healing processes leading to favorable wound-care outcomes. Herein, an exosome-guided cell reprogramming technique is proposed to directly convert M1 to M2 Mϕs for effective wound management. The M2 Mϕ-derived exosomes (M2-Exo) induce a complete conversion of M1 to M2 Mϕs in vitro. The reprogrammed M2 Mϕs turn Arginase (M2-marker) and iNOS (M1-marker) on and off, respectively, and exhibit distinct phenotypic and functional features of M2 Mϕs. M2-Exo has not only Mϕ reprogramming factors but also various cytokines and growth factors promoting wound repair. After subcutaneous administration of M2-Exo into the wound edge, the local populations of M1 and M2 Mϕs are markedly decreased and increased, respectively, showing a successful exosome-guided switch to M2 Mϕ polarization. The direct conversion of M1 to M2 Mϕs at the wound site accelerates wound healing by enhancing angiogenesis, re-epithelialization, and collagen deposition. The Mϕ phenotype switching induced by exosomes possessing the excellent cell reprogramming capability and innate biocompatibility can be a promising therapeutic approach for various inflammation-associated disorders by regulating the balance between pro- versus anti-inflammatory Mϕs.
Collapse
Affiliation(s)
- Hyosuk Kim
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Sun Young Wang
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Gijung Kwak
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Yoosoo Yang
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Ick Chan Kwon
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Sun Hwa Kim
- Center for TheragnosisBiomedical Research InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| |
Collapse
|
45
|
Shi Z, Zhang Y, Wang Q, Jiang D. MFG‐E8 regulates inflammation and apoptosis in tendon healing, and promotes tendon repair: A histological and biochemical evaluation. IUBMB Life 2019; 71:1986-1993. [DOI: 10.1002/iub.2143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Zhengzhou Shi
- Department of Urology, Shanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Youbo Zhang
- Department of Pediatric SurgeryNantong Maternal and Child Health Hospital Nantong Jiangsu China
| | - Qi Wang
- Department of Urology, Shanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Dapeng Jiang
- Department of Urology, Shanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
46
|
Shi Z, Wang Q, Jiang D. Extracellular vesicles from bone marrow-derived multipotent mesenchymal stromal cells regulate inflammation and enhance tendon healing. J Transl Med 2019; 17:211. [PMID: 31238964 PMCID: PMC6593555 DOI: 10.1186/s12967-019-1960-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023] Open
Abstract
Background Extracellular vesicles from bone marrow-derived multipotent mesenchymal stromal cells (BMSC-EVs) can play important roles in the repair of injured tissues. However, no reports have investigated the role and underlying mechanisms of BMSCs-EVs in the tendon repair process. We hypothesized that BMSC-EVs may play a role in modulating inflammation during tendon healing and improving tendon repair in a rat model of patellar tendon injury. Methods First, we created window defects in the patellar tendons of Sprague–Dawley rats. Rats (n = 16) were then randomly assigned to three groups: BMSC-EVs group, Fibrin group, and control group. Rats in the BMSC-EVs group were treated with BMSC-EVs and fibrin glue (25 µg in 10 µL). Rats in the fibrin group were treated with fibrin only, and those in the control group received no treatment. Histopathology, immunohistochemistry, and gene expression analyses were performed at 2 and 4 weeks after surgery. Results At 4 weeks, tendons treated with BMSC-EVs showed regularly aligned and compact collagen fibers as compared with the disrupted scar-like healing in rats in the fibrin and control groups. The expression of genes related to tendon matrix formation and tenogenic differentiation: collagen (COL)-1a1, scleraxis (SCX), and tenomodulin (TNMD) was significantly higher in the BMSC-EVs group than in the other two groups. With histopathology, we observed significantly higher numbers of CD146+ tendon stem cells and fewer numbers of apoptotic cells and C–C chemokine receptor type 7 (CCR7)-positive proinflammatory macrophages in the BMSC-EVs group. BMSC-EVs treatment also led to an increase in the expression of anti-inflammatory mediators (IL-10 and IL-4) at 2 weeks after surgery. Conclusions Overall, our findings show that the local administration of BMSC-EVs promotes tendon healing by suppressing inflammation and apoptotic cell accumulation and increasing the proportion of tendon-resident stem/progenitor cells. These findings provide a basis for the potential clinical use of BMSC-EVs in tendon repair.
Collapse
Affiliation(s)
- Zhengzhou Shi
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Qi Wang
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Dapeng Jiang
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
47
|
Engineered delivery strategies for enhanced control of growth factor activities in wound healing. Adv Drug Deliv Rev 2019; 146:190-208. [PMID: 29879493 DOI: 10.1016/j.addr.2018.06.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/18/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022]
Abstract
Growth factors (GFs) are versatile signalling molecules that orchestrate the dynamic, multi-stage process of wound healing. Delivery of exogenous GFs to the wound milieu to mediate healing in an active, physiologically-relevant manner has shown great promise in laboratories; however, the inherent instability of GFs, accompanied with numerous safety, efficacy and cost concerns, has hindered the clinical success of GF delivery. In this article, we highlight that the key to overcoming these challenges is to enhance the control of the activities of GFs throughout the delivering process. We summarise the recent strategies based on biomaterials matrices and molecular engineering, which aim to improve the conditions of GFs for delivery (at the 'supply' end of the delivery), increase the stability and functions of GFs in extracellular matrix (in transportation to target cells), as well as enhance the GFs/receptor interaction on the cell membrane (at the 'destination' end of the delivery). Many of these investigations have led to encouraging outcomes in various in vitro and in vivo regenerative models with considerable translational potential.
Collapse
|
48
|
McCoy AM, Arrington J, Yau PM. Effect of Preparation Method on the Protein Profile of Equine Amnion Dressings. J Proteome Res 2019; 18:2676-2685. [PMID: 31117638 DOI: 10.1021/acs.jproteome.9b00240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The protein content of amnion is thought to be the primary contributor to its efficacy as a biological dressing for wounds. Protein elution into antibiotic processing media has been reported, but the effect of antiseptic-based processing methods is unknown. Amniotic membranes were collected from eight healthy mares. Samples were collected after removal of gross debris. Tissues were subsequently divided and processed with either 0.05% chlorhexidine or 2% iodine/0.25% acetic acid. After protein extraction and trypsin digestion, the proteins were labeled with 8-plex iTRAQ tags, combined, and analyzed by high-resolution liquid chromatography-mass spectrometry. The MaxQuant-Perseus software suite was used to identify and quantify sample proteins, with functional annotation performed in PANTHER. There were 220 unique proteins identified, of which 144 were found in all individuals and across all conditions, several with a known role in wound healing. Contrary to expectations, processing did not significantly alter the protein content of the amnion tissue. Limitations include the small sample size and single time point. These results suggest that either processing method is acceptable for use in the preparation of equine amnion dressings. The role of expressed proteins in the biological activity of amnion dressings remains to be elucidated.
Collapse
Affiliation(s)
- Annette M McCoy
- Department of Veterinary Clinical Medicine , University of Illinois Urbana-Champaign , 1008 West Hazelwood Drive , Urbana , Illinois 61802 , United States
| | - Justine Arrington
- Protein Sciences Facility, Roy J. Carver Biotechnology Center , University of Illinois Urbana-Champaign , 505 South Matthews Avenue , Urbana , Illinois 61801 , United States
| | - Peter M Yau
- Protein Sciences Facility, Roy J. Carver Biotechnology Center , University of Illinois Urbana-Champaign , 505 South Matthews Avenue , Urbana , Illinois 61801 , United States
| |
Collapse
|
49
|
Panahipour L, Kochergina E, Kreissl A, Haiden N, Gruber R. Milk modulates macrophage polarization in vitro. Cytokine X 2019; 1:100009. [PMID: 33604549 PMCID: PMC7885867 DOI: 10.1016/j.cytox.2019.100009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/17/2019] [Accepted: 05/17/2019] [Indexed: 02/01/2023] Open
Abstract
Objective Milk holds an anti-inflammatory response that is particularly important to protecting infants against necrotizing enterocolitis. Milk might also exert anti-inflammatory effects in adulthood, including the oral cavity where macrophages of the oral mucosal control innate immunity defense. It remains unknown, however, whether milk can modulate the local inflammatory response by affecting the polarization of macrophages. Material and Methods To determine whether pasteurized human milk and pasteurized cow milk can provoke macrophage polarization, murine bone marrow macrophages and RAW264.7 cells were exposed to human saliva or the inflammatory cytokines IL1β and TNFα. Activation of pro-(M1) inflammatory response is indicated by the expression of IL1 and IL8. To determine polarization towards a M2 phenotype, the expression of arginase 1 (ARG1) and chitinase-like 3 (Chil3) was determined by reverse transcriptase PCR and immunoassay. Western blot was done on phosphorylated p38 and JNK. Results Aqueous fractions of human milk and cow milk from different donors, respectively, significantly decreased the inflammatory response of primary macrophages and RAW264.7 cells when exposed to saliva or IL1 and TNFα. Similar to IL4, human milk and cow milk caused a robust expression of ARG1 and Chil3 in primary macrophages. The polarization of macrophages by pasteurized milk occurred independent of the phosphorylation of p38 and JNK. Conclusion These data suggest that pasteurized milk, independent of the origin, can cause the polarization of macrophages from a pro-inflammatory M1 towards a pro-resolving M2 phenotype. Thus, milk might have a protective role for the oral cavity by modulation of the macrophage-based innate immune system.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria
| | - Evgeniya Kochergina
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria
| | - Alexandra Kreissl
- Department of Paediatrics and Adolescent Medicine, Division of Neonatology, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria
| | - Nadja Haiden
- Department of Clinical Pharmacology, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria.,Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland.,Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1200 Vienna, Austria
| |
Collapse
|
50
|
Yu L, Zhao L, Jia Z, Bi J, Wei Q, Song X, Jiang L, Lin S, Wei M. MFG-E8 overexpression is associated with poor prognosis in breast cancer patients. Pathol Res Pract 2018; 215:490-498. [PMID: 30612778 DOI: 10.1016/j.prp.2018.12.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/10/2018] [Accepted: 12/30/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND MFG-E8(Milk fat globule-EGF factor 8), a secreted glycoprotein, plays an exceptional role in various diseases. MFG-E8 overexpression is found in a variety of cancers. However, it remains unclear whether MFG-E8 overexpression is associated with the clinicopathological characteristics and prognosis of human breast cancer. MATERIALS AND METHODS In this study, we detected the expression and localization of MFG-E8 protein in breast cancer and cancer-adjacent tissues using immunohistochemical staining, Western blot analysis and immunofluorescence. We analyzed the association between MFG-E8 expression and clinical characteristics and outcomes of breast cancer patients with different HR and HER2 statuses. RESULTS Our results confirmed that MFG-E8 expression increased significantly in breast cancer compared with cancer-adjacent tissues by immunohistochemical staining (P < 0.001). Similarly, the Western blot results further confirmed the increased expression of MFG-E8 in breast cancer compared with cancer-adjacent tissues (P = 0.001). Immunofluorescence staining showed that MFG-E8 was mainly localized in the cytoplasm and membrane of tumor cells, consistent with the immunohistochemical staining results. The high expression levels of MFG-E8 showed a greater association with lymph node metastasis, TNM stage and histological grade (P < 0.001). Moreover, high MFG-E8 expression was related to a shortened overall survival (OS) (P < 0.001) and disease-free survival (DFS) (P < 0.001). Bioinformatics analysis with a Kaplan-Meier plotter also demonstrated a strong association of MFG-E8 mRNA overexpression with a short OS and DFS compared with low MFG-E8 expression (P = 0.040, P = 0.005). CONCLUSIONS Our findings indicate that MFG-E8 may be a potential marker for poor prognosis and survival in breast cancer.
Collapse
Affiliation(s)
- Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, 110122, China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, 110122, China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Zhen Jia
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, 110122, China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Jia Bi
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, 110122, China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, 110122, China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Xinyue Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, 110122, China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Longyang Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, 110122, China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Shu Lin
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, 110122, China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, 110122, China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.
| |
Collapse
|