1
|
Rajasekar V, Abdalla MM, Neelakantan P, Yiu CKY. Cellular dynamics and signalling mechanisms in dentine repair: A narrative review. Int Endod J 2025. [PMID: 40491185 DOI: 10.1111/iej.14261] [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: 01/26/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND Bioactive molecules have gained significant attention in regenerative medicine due to their ability to boost the reparative properties of stem cells, including those in the dental pulp. This narrative review aims to deepen our understanding of the dynamics of bioactive molecules in the dental pulp and their role in enhancing hard tissue reparative processes. OBJECTIVES (i) To discuss the role of different cells and the critical pathways involved in dentine formation through direct (reparative) or indirect (infection control and immunomodulatory) mechanisms. (ii) To highlight how innovative therapeutic strategies could be employed to target key molecules for successful dentine repair and regeneration. METHODS The review encompassed all years up to the search period. Databases such as PubMed, Scopus and Medline were utilized to gather relevant studies. The search strategy involved specific signalling molecules such as Transforming growth factor-β1 (TGF-β), Bone Morphogenetic Proteins (BMP), Small Integrin Binding Ligand N-linked Glycoproteins (SIBLING) and growth factors. Cell types including odontoblasts, fibroblasts, immune cells and dental pulp stem cells (DPSCs) were of interest. Additionally, signalling pathways like Wnt, Notch, Shh, amongst others, were investigated for their roles in repair mechanisms. Key terms were combined using Boolean operators [Cell type] AND [signalling molecules] AND/OR [dentine], [Cell type] AND/OR [signalling pathways] AND/OR [dentine] to include studies addressing the interaction of these components in enhancing repair processes. DISCUSSION Key molecules such as TGF-β1, BMP and SIBLING proteins effectively enhance the dentine reparative response, whilst other molecules such as complement proteins and antimicrobial peptides primarily activate immune cells and facilitate pathogen clearance to promote the regenerative capabilities of DPSCs. This well-orchestrated interaction emphasizes the need to investigate the effects of these molecules on all cells within the dental pulp. Morphogenic signalling molecules such as BMP-2, -4 and -7, and Wnt show temporal, yet significant regenerative properties, whilst Shh and Notch present inconsistent effects on dentine regeneration, and a consensus on their roles and properties in dentine repair has yet to be reached. CONCLUSIONS This review highlights the critical role of bioactive molecules in dentine repair to guide the development of next-generation bioinspired therapeutics for vital pulp therapy.
Collapse
Affiliation(s)
- Vidhyashree Rajasekar
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - Mohamed Mahmoud Abdalla
- Dental Biomaterials, Faculty of Dental Medicine Al-Azhar University, Cairo, Egypt
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - Prasanna Neelakantan
- Faculty of Medicine and Dentistry, Mike Petryk School of Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Cynthia K Y Yiu
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, Hong Kong
| |
Collapse
|
2
|
Ghosh S, Bishayi B. Neutralization of IL-17 and CXCR1 Protects Septic Arthritis by Regulating CXCL8-CXCR1 Pathway Along With Functional Activities in Neutrophils. Int J Rheum Dis 2025; 28:e70144. [PMID: 40195600 DOI: 10.1111/1756-185x.70144] [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: 10/30/2024] [Revised: 02/04/2025] [Accepted: 02/13/2025] [Indexed: 04/09/2025]
Abstract
AIM The main objective of this study is to elucidate the role of CXCR1 Ab, IL-17 Ab, and gentamicin in protecting septic arthritis by regulating neutrophil functional responses while evaluating the contribution of the CXCL8-CXCR1 pathway. METHODS Eighty-four experimental swiss albino mice were utilized to study septic arthritis. They were divided into eight groups. After developing sepsis, respective mice groups were treated with CXCR1 Ab, IL-17 Ab, and gentamicin doses. Doses were administered on days 1, 8, and 13 of the experimental schedule. At the early, middle, and late phases of the experiment i.e. at 3, 10, and 15 DPI (Days Post Infection), mice were sacrificed and blood and tissues were collected for further experimental evaluations. Different functional studies were performed on isolated blood neutrophils, spleen, and synovial tissues. Histological evaluation, immunofluorescence study, sepsis profile, and downstream signaling pathway analysis were done to obtain data. RESULTS Infected mice group exhibited high inflammatory responses while treatment helped to mitigate them. IL-17 neutralization helped to lower bacterial burden, neutrophil ROS activity, MPO activity, and ALP activity. However, the combined neutralization of CXCR1 and IL-17 greatly influenced PMN chemotactic activity and lysozyme activity. At the early phase of the experiment, IL-17 neutralization's impact was more prominent, while later, CXCR1 neutralization gained the upper hand. CONCLUSIONS We can conclude that IL-17 Ab in combination with gentamicin is potent in modulating neutrophil activities positively to cure sepsis, while CXCR1 Ab, through the CXCL8/CXCR1 pathway, regulates neutrophil functional activities by impacting different downstream signaling cascades.
Collapse
MESH Headings
- Animals
- Neutrophils/drug effects
- Neutrophils/immunology
- Neutrophils/metabolism
- Neutrophils/microbiology
- Signal Transduction/drug effects
- Interleukin-17/immunology
- Interleukin-17/antagonists & inhibitors
- Interleukin-17/metabolism
- Arthritis, Infectious/immunology
- Arthritis, Infectious/microbiology
- Arthritis, Infectious/prevention & control
- Arthritis, Infectious/metabolism
- Receptors, Interleukin-8A/metabolism
- Receptors, Interleukin-8A/immunology
- Receptors, Interleukin-8A/antagonists & inhibitors
- Disease Models, Animal
- Interleukin-8/metabolism
- Mice
- Time Factors
- Male
- Arthritis, Experimental/immunology
- Arthritis, Experimental/prevention & control
- Arthritis, Experimental/microbiology
- Arthritis, Experimental/metabolism
- Anti-Bacterial Agents/pharmacology
Collapse
Affiliation(s)
- Sharmistha Ghosh
- Immunology Laboratory, Department of Physiology, University of Calcutta, University Colleges of Science and Technology, Kolkata, West Bengal, India
| | - Biswadev Bishayi
- Immunology Laboratory, Department of Physiology, University of Calcutta, University Colleges of Science and Technology, Kolkata, West Bengal, India
| |
Collapse
|
3
|
Livingstone EJ, Cartwright JA, Campana L, Lewis PJS, Dwyer BJ, Aird R, Man TY, Vermeren M, Rossi AG, Boulter L, Forbes SJ. Semaphorin 7a is protective through immune modulation during acetaminophen-induced liver injury. J Inflamm (Lond) 2025; 22:13. [PMID: 40114253 PMCID: PMC11927371 DOI: 10.1186/s12950-025-00429-x] [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: 10/04/2024] [Accepted: 01/13/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND AND AIM Acetaminophen (APAP) induced acute liver injury (ALI), the leading cause acute liver failure in the western world, has limited treatment options. APAP toxicity results in massive hepatic necrosis and secondary infiltrating monocytes and neutrophils, which contribute to pathogenesis. Semaphorin 7a (Sema7a), a chemoattractant and modulator of monocytes and neutrophils, is a potential therapeutic target in other conditions, but its role in APAP-ALI is unexplored. METHODS Wild-type (WT) and Sema7a knockout (KO) mice were examined during APAP-ALI. Serum liver function tests, histological analysis and cellular localisation of Sema7a and its receptors, Plexin C1 and Integrin β1, were examined. Serum cytokines were quantified, tissue macrophages and neutrophils were localised, and in vivo phenotype, including phagocytosis, was assessed by immunohistochemistry and flow cytometry. RESULTS Sema7a was expressed by HNF4α + peri-necrotic hepatocytes circumferentially during APAP-ALI injury phases, and serum concentrations were increased, and correlated with hepatic injury. Sema7a KO mice had increased circulating inflammatory cytokines and significantly less hepatic F4/80 + macrophages, a cell type required for hepatic repair. Sema7a KO mice had higher necrotic area neutrophils, and increased neutrophil chemoattractant CXCL1. Without Sema7a expression, mice displayed increased necrosis and liver injury markers compared to Sema7a WT mice. Without peri-necrotic hepatocyte Sema7a expression, we also identified increased cell death and hepatic cellular stress outside of necrosis. CONCLUSION We have identified a novel protective role of Sema7a during injury phases of APAP-ALI. Without peri-necrotic hepatocyte Sema7a expression and secretion, there is increased inflammation, time specific worsened hepatic necrosis and increased hepatic cell stress and death outside of the necrotic zone.
Collapse
Affiliation(s)
- Eilidh J Livingstone
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Jennifer A Cartwright
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
- The Royal (Dick) School of Veterinary Studiesand Theaq , Roslin Institute, University of Edinburgh, Edinburgh, UK.
| | - Lara Campana
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Philip J Starkey Lewis
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Benjamin J Dwyer
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Rhona Aird
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Tak Yung Man
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Matthieu Vermeren
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Adriano Giorgio Rossi
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Stuart John Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
Huang X, Jannu AJ, Song Z, Jury‐Garfe N, Lasagna‐Reeves CA, Alzheimer's Disease Neuroimaging Initiative, Johnson TS, Huang K, Zhang J. Predicting Alzheimer's disease subtypes and understanding their molecular characteristics in living patients with transcriptomic trajectory profiling. Alzheimers Dement 2025; 21:e14241. [PMID: 39812331 PMCID: PMC11772740 DOI: 10.1002/alz.14241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/18/2024] [Accepted: 08/02/2024] [Indexed: 01/16/2025]
Abstract
INTRODUCTION Deciphering the diverse molecular mechanisms in living Alzheimer's disease (AD) patients is a big challenge but is pivotal for disease prognosis and precision medicine development. METHODS Utilizing an optimal transport approach, we conducted graph-based mapping of transcriptomic profiles to transfer AD subtype labels from ROSMAP monocyte samples to ADNI and ANMerge peripheral blood mononuclear cells. Subsequently, differential expression followed by comparative pathway and diffusion pseudotime analysis were applied to each cohort to infer the progression trajectories. Survival analysis with real follow-up time was used to obtain potential biomarkers for AD prognosis. RESULTS AD subtype labels were accurately transferred onto the blood samples of ADNI and ANMerge living patients. Pathways and associated genes in neutrophil degranulation-like immune process, immune acute phase response, and IL-6 signaling were significantly associated with AD progression. DISCUSSION The work enhanced our understanding of AD progression in different subtypes, offering insights into potential biomarkers and personalized interventions for improved patient care. HIGHLIGHTS We applied an innovative optimal transport-based approach to map transcriptomic data from different Alzheimer's disease (AD) cohort studies and transfer known AD subtype labels from ROSMAP monocyte samples to peripheral blood mononuclear cell (PBMC) samples within ADNI and ANMerge cohorts. Through comprehensive trajectory and comparative analysis, we investigated the molecular mechanisms underlying different disease progression trajectories in AD. We validated the accuracy of our AD subtype label transfer and identified prognostic genetic markers associated with disease progression, facilitating personalized treatment strategies. By identifying and predicting distinctive AD subtypes and their associated pathways, our study contributes to a deeper understanding of AD heterogeneity.
Collapse
Affiliation(s)
- Xiaoqing Huang
- Department of Biostatistics & Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Asha Jacob Jannu
- Department of Biohealth InformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Ziyan Song
- Department of Biostatistics & Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Nur Jury‐Garfe
- Department of Anatomy, Cell Biology & PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Cristian A. Lasagna‐Reeves
- Department of Anatomy, Cell Biology & PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | | | - Travis S. Johnson
- Department of Biostatistics & Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kun Huang
- Department of Biostatistics & Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jie Zhang
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
5
|
Pallod S, Aguilera Olvera R, Ghosh D, Rai L, Brimo S, DeCambra W, Sant HG, Ristich E, Singh V, Abedin MR, Chang N, Yarger JL, Lee JK, Kilbourne J, Yaron JR, Haydel SE, Rege K. Skin repair and infection control in diabetic, obese mice using bioactive laser-activated sealants. Biomaterials 2024; 311:122668. [PMID: 38908232 PMCID: PMC11562812 DOI: 10.1016/j.biomaterials.2024.122668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Conventional wound approximation devices, including sutures, staples, and glues, are widely used but risk of wound dehiscence, local infection, and scarring can be exacerbated in these approaches, including in diabetic and obese individuals. This study reports the efficacy and quality of tissue repair upon photothermal sealing of full-thickness incisional skin wounds using silk fibroin-based laser-activated sealants (LASEs) containing copper chloride salt (Cu-LASE) or silver nanoprisms (AgNPr-LASE), which absorb and convert near-infrared (NIR) laser energy to heat. LASE application results in rapid and effective skin sealing in healthy, immunodeficient, as well as diabetic and obese mice. Although lower recovery of epidermal structure and function was seen with AgNPr-LASE sealing, likely because of the hyperthermia induced by laser and presence of this material in the wound space, this approach resulted in higher enhancement in recovery of skin biomechanical strength compared to sutures and Cu-LASEs in diabetic, obese mice. Histological and immunohistochemical analyses revealed that AgNPr-LASEs resulted in significantly lower neutrophil migration to the wound compared to Cu-LASEs and sutures, indicating a more muted inflammatory response. Cu-LASEs resulted in local tissue toxicity likely because of effects of copper ions as manifested in the form of a significant epidermal gap and a 'depletion zone', which was a region devoid of viable cells proximal to the wound. Compared to sutures, LASE-mediated sealing, in later stages of healing, resulted in increased angiogenesis and diminished myofibroblast activation, which can be indicative of lower scarring. AgNPr-LASE loaded with vancomycin, an antibiotic drug, significantly lowered methicillin-resistant Staphylococcus aureus (MRSA) load in a pathogen challenge model in diabetic and obese mice and also reduced post-infection inflammation of tissue compared to antibacterial sutures. Taken together, these attributes indicate that AgNPr-LASE demonstrated a more balanced quality of tissue sealing and repair in diabetic and obese mice and can be used for combating local infections, that can result in poor healing in these individuals.
Collapse
Affiliation(s)
- Shubham Pallod
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, USA
| | - Rodrigo Aguilera Olvera
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, USA
| | - Deepanjan Ghosh
- Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, USA
| | - Lama Rai
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; College of Health Solutions, Arizona State University, USA
| | - Souzan Brimo
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Arizona State University, USA
| | | | - Harsh Girish Sant
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Arizona State University, USA
| | - Eron Ristich
- School of Molecular Sciences, Arizona State University, USA; School of Computing and Augmented Intelligence, Arizona State University, USA
| | - Vanshika Singh
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Arizona State University, USA
| | - Muhammad Raisul Abedin
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA
| | - Nicolas Chang
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Biomedical Engineering, School for Biological and Health Systems Engineering, Arizona State University, USA
| | | | - Jung Keun Lee
- Departments of Pathology and Population Medicine, Midwestern University, College of Veterinary Medicine, 5725 West Utopia Rd., Glendale, AZ, 85308, USA
| | | | - Jordan R Yaron
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA
| | - Shelley E Haydel
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, USA; School of Life Sciences, Arizona State University, 501 E. Tyler Mall ECG 303, Tempe, AZ, 85287-6106, USA
| | - Kaushal Rege
- Center for Biomaterials Innovation and Translation, Biodesign Institute, Arizona State University, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Arizona State University, USA.
| |
Collapse
|
6
|
Gong P, Ding Y, Li W, Yang J, Su X, Tian R, Zhou Y, Wang T, Jiang J, Liu R, Fang J, Feng C, Shao C, Shi Y, Li P. Neutrophil-Driven M2-Like Macrophages Are Critical for Skin Fibrosis in a Systemic Sclerosis Model. J Invest Dermatol 2024; 144:2426-2439.e3. [PMID: 38580106 DOI: 10.1016/j.jid.2024.03.031] [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: 04/28/2023] [Revised: 01/04/2024] [Accepted: 03/03/2024] [Indexed: 04/07/2024]
Abstract
Systemic sclerosis (SSc) is a challenging autoimmune disease characterized by progressive fibrosis affecting the skin and internal organs. Despite the known infiltration of macrophages and neutrophils, their precise contributions to SSc pathogenesis remain elusive. In this study, we elucidated that CD206hiMHCIIlo M2-like macrophages constitute the predominant pathogenic immune cell population in the fibrotic skin of a bleomycin-induced SSc mouse model. These cells emerged as pivotal contributors to the profibrotic response by orchestrating the production of TGF-β1 through a MerTK signaling-dependent manner. Notably, we observed that neutrophil infiltration was a prerequisite for accumulation of M2-like macrophages. Strategies such as neutrophil depletion or inhibition of CXCR1/2 were proven effective in reducing M2-like macrophages, subsequently mitigating SSc progression. Detailed investigations revealed that in fibrotic skin, neutrophil-released neutrophil extracellular traps were responsible for the differentiation of M2-like macrophages. Our findings illuminate the significant involvement of the neutrophil-macrophage-fibrosis axis in SSc pathogenesis, offering critical information for the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Pixia Gong
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China; Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yayun Ding
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wen Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jie Yang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Xiao Su
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Ruifeng Tian
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yipeng Zhou
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Tingting Wang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Junjie Jiang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Rui Liu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jiankai Fang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chao Feng
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Peishan Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
7
|
Chen Z, Zou Y, Sun H, He Y, Ye K, Li Y, Qiu L, Mai Y, Chen X, Mao Z, Yi C, Wang W. Engineered Enucleated Mesenchymal Stem Cells Regulating Immune Microenvironment and Promoting Wound Healing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2412253. [PMID: 39295480 DOI: 10.1002/adma.202412253] [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: 08/19/2024] [Indexed: 09/21/2024]
Abstract
Persistent excessive inflammation caused by neutrophil and macrophage dysfunction in the wound bed leads to refractory response during wound healing. However, previous studies using cytokines or drugs often suffer from short half-lives and limited targeting, resulting in unsatisfactory therapeutic effects. Herein, the enucleated mesenchymal stem cell is engineered by aptamer bioorthogonal chemistry to modify the cell membrane and mRNA loading in the cell cytoplasm as a novel delivery vector (Cargocyte) with accurate targeting and sustained cytokine secretion. Cargocytes can successfully reduce NETosis by targeting the nuclear chromatin protein DEK protein with aptamers and sustaining interleukin (IL)-4 expression to overcome the challenges associated with the high cost and short half-life of IL-4 protein and significantly prevent the transition of macrophages into the M1 phenotype. Therapeutic effects have been demonstrated in murine and porcine wound models and have powerful potential to improve wound immune microenvironments effectively. Overall, the use of engineered enucleated mesenchymal stem cells as a delivery system may be a promising approach for wound healing.
Collapse
Affiliation(s)
- Zhengtai Chen
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
| | - Yang Zou
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Hanxiao Sun
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
| | - Yan He
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
| | - Kai Ye
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
| | - Yi Li
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
| | - Lihong Qiu
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
| | - Yuexue Mai
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
| | - Xinghong Chen
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Chenggang Yi
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
| | - Wei Wang
- Department of Plastic Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310000, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, Zhejiang, 311215, China
| |
Collapse
|
8
|
Gao H, Jiang Y, Zeng G, Huda N, Thoudam T, Yang Z, Liangpunsakul S, Ma J. Cell-to-cell and organ-to-organ crosstalk in the pathogenesis of alcohol-associated liver disease. EGASTROENTEROLOGY 2024; 2:e100104. [PMID: 39735421 PMCID: PMC11674000 DOI: 10.1136/egastro-2024-100104] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/08/2024] [Indexed: 12/31/2024]
Abstract
Alcohol-associated liver disease (ALD) is a growing global health concern and its prevalence and severity are increasing steadily. While bacterial endotoxin translocation into the portal circulation is a well-established key factor, recent evidence highlights the critical role of sterile inflammation, triggered by diverse stimuli, in alcohol-induced liver injury. This review provides a comprehensive analysis of the complex interactions within the hepatic microenvironment in ALD. It examines the contributions of both parenchymal cells, like hepatocytes, and non-parenchymal cells, such as hepatic stellate cells, Kupffer cells, neutrophils, and liver sinusoidal endothelial cells, in driving the progression of the disease. Additionally, we explored the involvement of key mediators, including cytokines, chemokines and inflammasomes, which regulate inflammatory responses and promote liver injury and fibrosis. A particular focus has been placed on extracellular vesicles (EVs) as essential mediators of intercellular communication both within and beyond the liver. These vesicles facilitate the transfer of signalling molecules, such as microRNAs and proteins, which modulate immune responses, fibrogenesis and lipid metabolism, thereby influencing disease progression. Moreover, we underscore the importance of organ-to-organ crosstalk, particularly in the gut-liver axis, where dysbiosis and increased intestinal permeability lead to microbial translocation, exacerbating hepatic inflammation. The adipose-liver axis is also highlighted, particularly the impact of adipokines and free fatty acids from adipose tissue on hepatic steatosis and inflammation in the context of alcohol consumption.
Collapse
Affiliation(s)
- Hui Gao
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yanchao Jiang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ge Zeng
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Infectious Diseases, Southern Medical University, Guangzhou, China
| | - Nazmul Huda
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Themis Thoudam
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Zhihong Yang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Suthat Liangpunsakul
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| | - Jing Ma
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
9
|
Xu H, Lotfy P, Gelb S, Pragana A, Hehnly C, Byer LIJ, Shipley FB, Zawadzki ME, Cui J, Deng L, Taylor M, Webb M, Lidov HGW, Andermann ML, Chiu IM, Ordovas-Montanes J, Lehtinen MK. The choroid plexus synergizes with immune cells during neuroinflammation. Cell 2024; 187:4946-4963.e17. [PMID: 39089253 PMCID: PMC11458255 DOI: 10.1016/j.cell.2024.07.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 04/19/2024] [Accepted: 07/01/2024] [Indexed: 08/03/2024]
Abstract
The choroid plexus (ChP) is a vital brain barrier and source of cerebrospinal fluid (CSF). Here, we use longitudinal two-photon imaging in awake mice and single-cell transcriptomics to elucidate the mechanisms of ChP regulation of brain inflammation. We used intracerebroventricular injections of lipopolysaccharides (LPS) to model meningitis in mice and observed that neutrophils and monocytes accumulated in the ChP stroma and surged across the epithelial barrier into the CSF. Bi-directional recruitment of monocytes from the periphery and, unexpectedly, macrophages from the CSF to the ChP helped eliminate neutrophils and repair the barrier. Transcriptomic analyses detailed the molecular steps accompanying this process and revealed that ChP epithelial cells transiently specialize to nurture immune cells, coordinating their recruitment, survival, and differentiation as well as regulation of the tight junctions that control the permeability of the ChP brain barrier. Collectively, we provide a mechanistic understanding and a comprehensive roadmap of neuroinflammation at the ChP brain barrier.
Collapse
Affiliation(s)
- Huixin Xu
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Peter Lotfy
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sivan Gelb
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Aja Pragana
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Christine Hehnly
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Lillian I J Byer
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Frederick B Shipley
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Miriam E Zawadzki
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Harvard MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA
| | - Jin Cui
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Liwen Deng
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Milo Taylor
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard College, Harvard University, Cambridge, MA 02138, USA
| | - Mya Webb
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Hart G W Lidov
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mark L Andermann
- Harvard MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Isaac M Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA; Harvard MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
10
|
Porubska B, Plevakova M, Fikarova N, Vasek D, Somova V, Sanovec O, Simonik O, Komrskova K, Krylov V, Tlapakova T, Krulova M, Krulova M. Therapeutic potential of Sertoli cells in vivo: alleviation of acute inflammation and improvement of sperm quality. Stem Cell Res Ther 2024; 15:282. [PMID: 39227878 PMCID: PMC11373210 DOI: 10.1186/s13287-024-03897-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Inflammation-induced testicular damage is a significant contributing factor to the increasing incidence of infertility. Traditional treatments during the inflammatory phase often fail to achieve the desired fertility outcomes, necessitating innovative interventions such as cell therapy. METHODS We explored the in vivo properties of intravenously administered Sertoli cells (SCs) in an acute lipopolysaccharide (LPS)-induced inflammatory mouse model. Infiltrating and resident myeloid cell phenotypes were assessed using flow cytometry. The impact of SC administration on testis morphology and germ cell quality was evaluated using computer-assisted sperm analysis (CASA) and immunohistochemistry. RESULTS SCs demonstrated a distinctive migration pattern, importantly they preferentially concentrated in the testes and liver. SC application significantly reduced neutrophil infiltration as well as preserved the resident macrophage subpopulations. SCs upregulated MerTK expression in both interstitial and peritubular macrophages. Applied SC treatment exhibited protective effects on sperm including their motility and kinematic parameters, and maintained the physiological testicular morphology. CONCLUSION Our study provides compelling evidence of the therapeutic efficacy of SC transplantation in alleviating acute inflammation-induced testicular damage. These findings contribute to the expanding knowledge on the potential applications of cell-based therapies for addressing reproductive health challenges and offer a promising approach for targeted interventions in male infertility.
Collapse
Affiliation(s)
- Bianka Porubska
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic
| | - Marie Plevakova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic
| | - Natalie Fikarova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic
| | - Daniel Vasek
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic
| | - Veronika Somova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic
| | - Ondrej Sanovec
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Prumyslova 595, Prague, 252 50, Czech Republic
- Department of Physiology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic
| | - Ondrej Simonik
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Prumyslova 595, Prague, 252 50, Czech Republic
| | - Katerina Komrskova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Prumyslova 595, Prague, 252 50, Czech Republic
- Department of Zoology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic
| | - Vladimir Krylov
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic
| | - Tereza Tlapakova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic
| | - Magdalena Krulova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic.
| | - Magdalena Krulova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague, 2, 128 00, Czech Republic
| |
Collapse
|
11
|
Ignes-Romeu A, Weppner HK, Kaur T, Singh M, Hind LE. THP-1 Macrophages Limit Neutrophil Transendothelial Migration in a Model Infection. Cell Mol Bioeng 2024; 17:279-293. [PMID: 39372553 PMCID: PMC11450111 DOI: 10.1007/s12195-024-00813-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/09/2024] [Indexed: 10/08/2024] Open
Abstract
Introduction Dysregulated neutrophil function plays a significant role in the pathology of infections, cancer, cardiovascular diseases, and autoimmune disorders. Neutrophil activity is influenced by various cell populations, including macrophages, which are crucial regulators. However, the exact role of human macrophages in controlling neutrophil function remains unclear due to a scarcity of studies utilizing human cells in physiologically relevant models. Methods We adapted our "Infection-on-a-Chip" microfluidic device to incorporate macrophages within the collagen extracellular matrix, allowing for the study of interactions between human neutrophils and macrophages in a context that mimics in vivo conditions. The integration of THP-1 macrophages was optimized and their effect on the endothelial lumen was characterized, focusing on permeability and structural integrity. The device was then employed to examine the influence of macrophages on neutrophil response to infection with the bacterial pathogen Pseudomonas aeruginosa. Results Integration of THP-1 macrophages into the microfluidic device was successfully optimized, showing no increase in endothelial permeability or structural damage. The presence of macrophages was found to significantly reduce neutrophil transendothelial migration in response to Pseudomonas aeruginosa infection. Conclusions Our findings highlight the regulatory role of macrophages in modulating neutrophil responses, suggesting potential therapeutic targets to control neutrophil function in various diseases. The modified microfluidic platform offers a valuable tool for mechanistic studies into macrophage-neutrophil interactions in disease contexts. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00813-2.
Collapse
Affiliation(s)
- Aitana Ignes-Romeu
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303 USA
| | - Hannah K. Weppner
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303 USA
| | - Tanisha Kaur
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303 USA
| | - Maya Singh
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303 USA
- Present Address: Department of Bioengineering, University of Washington, Seattle, WA 98195 USA
| | - Laurel E. Hind
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO 80303 USA
| |
Collapse
|
12
|
Qin M, Ou R, He W, Han H, Zhang Y, Huang Y, Chen Z, Pan X, Chi Y, He S, Gao L. Salvianolic acid B enhances tissue repair and regeneration by regulating immune cell migration and Caveolin-1-mediated blastema formation in zebrafish. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155553. [PMID: 38820664 DOI: 10.1016/j.phymed.2024.155553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/01/2024] [Accepted: 03/19/2024] [Indexed: 06/02/2024]
Abstract
INTRODUCTION Non-healing wounds resulting from trauma, surgery, and chronic diseases annually affect millions of individuals globally, with limited therapeutic strategies available due to the incomplete understanding of the molecular processes governing tissue repair and regeneration. Salvianolic acid B (Sal B) has shown promising bioactivities in promoting angiogenesis and inhibiting inflammation. However, its regulatory mechanisms in tissue regeneration remain unclear. PURPOSE This study aims to investigate the effects of Sal B on wound healing and regeneration processes, along with its underlying molecular mechanisms, by employing zebrafish as a model organism. METHODS In this study, we employed a multifaceted approach to evaluate the impact of Sal B on zebrafish tail fin regeneration. We utilized whole-fish immunofluorescence, TUNEL staining, mitochondrial membrane potential (MMP), and Acridine Orange (AO) probes to analyze the tissue repair and regenerative under Sal B treatment. Additionally, we utilized transgenic zebrafish strains to investigate the migration of inflammatory cells during different phases of fin regeneration. To validate the importance of Caveolin-1 (Cav1) in tissue regeneration, we delved into its functional role using molecular docking and Morpholino-based gene knockdown techniques. Additionally, we quantified Cav1 expression levels through the application of in situ hybridization. RESULTS Our findings demonstrated that Sal B expedites zebrafish tail fin regeneration through a multifaceted mechanism involving the promotion of cell proliferation, suppression of apoptosis, and enhancement of MMP. Furthermore, Sal B was found to exert regulatory control over the dynamic aggregation and subsequent regression of immune cells during tissue regenerative processes. Importantly, we observed that the knockdown of Cav1 significantly compromised tissue regeneration, leading to an excessive infiltration of immune cells and increased levels of apoptosis. Moreover, the knockdown of Cav1 also affects blastema formation, a critical process influenced by Cav1 in tissue regeneration. CONCLUSION The results of this study showed that Sal B facilitated tissue repair and regeneration through regulating of immune cell migration and Cav1-mediated fibroblast activation, promoting blastema formation and development. This study highlighted the potential pharmacological effects of Sal B in promoting tissue regeneration. These findings contributed to the advancement of regenerative medicine research and the development of novel therapeutic approaches for trauma.
Collapse
Affiliation(s)
- Mengchen Qin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Rouxuan Ou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Weiyi He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Haoyang Han
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuxue Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yan Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhaohan Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoyan Pan
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yali Chi
- Department of Obstetrics & Gynecology, Nanfang Hospital, Southern Medical University (SMU), Guangzhou, China.
| | - Songqi He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
13
|
Shu LZ, Zhang XL, Ding YD, Lin H. From inflammation to bone formation: the intricate role of neutrophils in skeletal muscle injury and traumatic heterotopic ossification. Exp Mol Med 2024; 56:1523-1530. [PMID: 38945957 PMCID: PMC11297321 DOI: 10.1038/s12276-024-01270-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 03/22/2024] [Accepted: 04/16/2024] [Indexed: 07/02/2024] Open
Abstract
Neutrophils are emerging as an important player in skeletal muscle injury and repair. Neutrophils accumulate in injured tissue, thus releasing inflammatory factors, proteases and neutrophil extracellular traps (NETs) to clear muscle debris and pathogens when skeletal muscle is damaged. During the process of muscle repair, neutrophils can promote self-renewal and angiogenesis in satellite cells. When neutrophils are abnormally overactivated, neutrophils cause collagen deposition, functional impairment of satellite cells, and damage to the skeletal muscle vascular endothelium. Heterotopic ossification (HO) refers to abnormal bone formation in soft tissue. Skeletal muscle injury is one of the main causes of traumatic HO (tHO). Neutrophils play a pivotal role in activating BMPs and TGF-β signals, thus promoting the differentiation of mesenchymal stem cells and progenitor cells into osteoblasts or osteoclasts to facilitate HO. Furthermore, NETs are specifically localized at the site of HO, thereby accelerating the formation of HO. Additionally, the overactivation of neutrophils contributes to the disruption of immune homeostasis to trigger HO. An understanding of the diverse roles of neutrophils will not only provide more information on the pathogenesis of skeletal muscle injury for repair and HO but also provides a foundation for the development of more efficacious treatment modalities for HO.
Collapse
Affiliation(s)
- Lin-Zhen Shu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Xian-Lei Zhang
- Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Yi-Dan Ding
- Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Hui Lin
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China.
| |
Collapse
|
14
|
Zhao C, Yang Z, Li Y, Wen Z. Macrophages in tissue repair and regeneration: insights from zebrafish. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:12. [PMID: 38861103 PMCID: PMC11166613 DOI: 10.1186/s13619-024-00195-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Macrophages play crucial and versatile roles in regulating tissue repair and regeneration upon injury. However, due to their complex compositional heterogeneity and functional plasticity, deciphering the nature of different macrophage subpopulations and unraveling their dynamics and precise roles during the repair process have been challenging. With its distinct advantages, zebrafish (Danio rerio) has emerged as an invaluable model for studying macrophage development and functions, especially in tissue repair and regeneration, providing valuable insights into our understanding of macrophage biology in health and diseases. In this review, we present the current knowledge and challenges associated with the role of macrophages in tissue repair and regeneration, highlighting the significant contributions made by zebrafish studies. We discuss the unique advantages of the zebrafish model, including its genetic tools, imaging techniques, and regenerative capacities, which have greatly facilitated the investigation of macrophages in these processes. Additionally, we outline the potential of zebrafish research in addressing the remaining challenges and advancing our understanding of the intricate interplay between macrophages and tissue repair and regeneration.
Collapse
Affiliation(s)
- Changlong Zhao
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhiyong Yang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Division of Life Science, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yunbo Li
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Division of Life Science, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Zilong Wen
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
15
|
Farzamfar S, Garcia LM, Rahmani M, Bolduc S. Navigating the Immunological Crossroads: Mesenchymal Stem/Stromal Cells as Architects of Inflammatory Harmony in Tissue-Engineered Constructs. Bioengineering (Basel) 2024; 11:494. [PMID: 38790361 PMCID: PMC11118848 DOI: 10.3390/bioengineering11050494] [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: 03/26/2024] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
In the dynamic landscape of tissue engineering, the integration of tissue-engineered constructs (TECs) faces a dual challenge-initiating beneficial inflammation for regeneration while avoiding the perils of prolonged immune activation. As TECs encounter the immediate reaction of the immune system upon implantation, the unique immunomodulatory properties of mesenchymal stem/stromal cells (MSCs) emerge as key navigators. Harnessing the paracrine effects of MSCs, researchers aim to craft a localized microenvironment that not only enhances TEC integration but also holds therapeutic promise for inflammatory-driven pathologies. This review unravels the latest advancements, applications, obstacles, and future prospects surrounding the strategic alliance between MSCs and TECs, shedding light on the immunological symphony that guides the course of regenerative medicine.
Collapse
Affiliation(s)
- Saeed Farzamfar
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
| | - Luciana Melo Garcia
- Department of Medicine, Université Laval, Québec, QC G1V 0A6, Canada;
- Hematology-Oncology Service, CHU de Québec—Université Laval, Québec, QC G1V 0A6, Canada
| | - Mahya Rahmani
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
| | - Stephane Bolduc
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
16
|
Cevallos C, Jarmoluk P, Sviercz F, López CAM, Freiberger RN, Delpino MV, Quarleri J. Bystander Effects and Profibrotic Interactions in Hepatic Stellate Cells during HIV and HCV Coinfection. J Immunol Res 2024; 2024:6343757. [PMID: 38715844 PMCID: PMC11074826 DOI: 10.1155/2024/6343757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/21/2025] Open
Abstract
This study aims to explore the influence of coinfection with HCV and HIV on hepatic fibrosis. A coculture system was set up to actively replicate both viruses, incorporating CD4 T lymphocytes (Jurkat), hepatic stellate cells (LX-2), and hepatocytes (Huh7.5). LX-2 cells' susceptibility to HIV infection was assessed through measurements of HIV receptor expression, exposure to cell-free virus, and cell-to-cell contact with HIV-infected Jurkat cells. The study evaluated profibrotic parameters, including programed cell death, ROS imbalance, cytokines (IL-6, TGF-β, and TNF-α), and extracellular matrix components (collagen, α-SMA, and MMP-9). The impact of HCV infection on LX-2/HIV-Jurkat was examined using soluble factors released from HCV-infected hepatocytes. Despite LX-2 cells being nonsusceptible to direct HIV infection, bystander effects were observed, leading to increased oxidative stress and dysregulated profibrotic cytokine release. Coculture with HIV-infected Jurkat cells intensified hepatic fibrosis, redox imbalance, expression of profibrotic cytokines, and extracellular matrix production. Conversely, HCV-infected Huh7.5 cells exhibited elevated profibrotic gene transcriptions but without measurable effects on the LX-2/HIV-Jurkat coculture. This study highlights how HIV-infected lymphocytes worsen hepatic fibrosis during HCV/HIV coinfection. They increase oxidative stress, profibrotic cytokine levels, and extracellular matrix production in hepatic stellate cells through direct contact and soluble factors. These insights offer valuable potential therapies for coinfected individuals.
Collapse
Affiliation(s)
- Cintia Cevallos
- Consejo de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires (UBA), Paraguay, 2155, piso 11, C1121 ABG, Buenos Aires, Argentina
| | - Patricio Jarmoluk
- Consejo de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires (UBA), Paraguay, 2155, piso 11, C1121 ABG, Buenos Aires, Argentina
| | - Franco Sviercz
- Consejo de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires (UBA), Paraguay, 2155, piso 11, C1121 ABG, Buenos Aires, Argentina
| | - Cinthya A. M. López
- Consejo de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires (UBA), Paraguay, 2155, piso 11, C1121 ABG, Buenos Aires, Argentina
| | - Rosa N. Freiberger
- Consejo de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires (UBA), Paraguay, 2155, piso 11, C1121 ABG, Buenos Aires, Argentina
| | - M. Victoria Delpino
- Consejo de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires (UBA), Paraguay, 2155, piso 11, C1121 ABG, Buenos Aires, Argentina
| | - Jorge Quarleri
- Consejo de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires (UBA), Paraguay, 2155, piso 11, C1121 ABG, Buenos Aires, Argentina
| |
Collapse
|
17
|
Conedera FM, Kokona D, Zinkernagel MS, Stein JV, Lin CP, Alt C, Enzmann V. Macrophages coordinate immune response to laser-induced injury via extracellular traps. J Neuroinflammation 2024; 21:68. [PMID: 38500151 PMCID: PMC10949579 DOI: 10.1186/s12974-024-03064-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Retinal degeneration results from disruptions in retinal homeostasis due to injury, disease, or aging and triggers peripheral leukocyte infiltration. Effective immune responses rely on coordinated actions of resident microglia and recruited macrophages, critical for tissue remodeling and repair. However, these phagocytes also contribute to chronic inflammation in degenerated retinas, yet the precise coordination of immune response to retinal damage remains elusive. Recent investigations have demonstrated that phagocytic cells can produce extracellular traps (ETs), which are a source of self-antigens that alter the immune response, which can potentially lead to tissue injury. METHODS Innovations in experimental systems facilitate real-time exploration of immune cell interactions and dynamic responses. We integrated in vivo imaging with ultrastructural analysis, transcriptomics, pharmacological treatments, and knockout mice to elucidate the role of phagocytes and their modulation of the local inflammatory response through extracellular traps (ETs). Deciphering these mechanisms is essential for developing novel and enhanced immunotherapeutic approaches that can redirect a specific maladaptive immune response towards favorable wound healing in the retina. RESULTS Our findings underscore the pivotal role of innate immune cells, especially macrophages/monocytes, in regulating retinal repair and inflammation. The absence of neutrophil and macrophage infiltration aids parenchymal integrity restoration, while their depletion, particularly macrophages/monocytes, impedes vascular recovery. We demonstrate that macrophages/monocytes, when recruited in the retina, release chromatin and granular proteins, forming ETs. Furthermore, the pharmacological inhibition of ETosis support retinal and vascular repair, surpassing the effects of blocking innate immune cell recruitment. Simultaneously, the absence of ETosis reshapes the inflammatory response, causing neutrophils, helper, and cytotoxic T-cells to be restricted primarily in the superficial capillary plexus instead of reaching the damaged photoreceptor layer. CONCLUSIONS Our data offer novel insights into innate immunity's role in responding to retinal damage and potentially help developing innovative immunotherapeutic approaches that can shift the immune response from maladaptive to beneficial for retinal regeneration.
Collapse
Affiliation(s)
- Federica M Conedera
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland.
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland.
| | - Despina Kokona
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens Alt
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Volker Enzmann
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
18
|
Wang J, Zhao Z, Yang K, Bai Y. Research progress in cell therapy for oral diseases: focus on cell sources and strategies to optimize cell function. Front Bioeng Biotechnol 2024; 12:1340728. [PMID: 38515628 PMCID: PMC10955105 DOI: 10.3389/fbioe.2024.1340728] [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: 11/20/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024] Open
Abstract
In recent years, cell therapy has come to play an important therapeutic role in oral diseases. This paper reviews the active role of mesenchymal stem cells, immune cell sources, and other cells in oral disorders, and presents data supporting the role of cell therapy in oral disorders, including bone and tooth regeneration, oral mucosal disorders, oral soft tissue defects, salivary gland dysfunction, and orthodontic tooth movement. The paper will first review the progress of cell optimization strategies for oral diseases, including the use of hormones in combination with stem cells, gene-modified regulatory cells, epigenetic regulation of cells, drug regulation of cells, cell sheets/aggregates, cell-binding scaffold materials and hydrogels, nanotechnology, and 3D bioprinting of cells. In summary, we will focus on the therapeutic exploration of these different cell sources in oral diseases and the active application of the latest cell optimization strategies.
Collapse
Affiliation(s)
| | | | | | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Zhao X, Deng H, Feng Y, Wang Y, Yao X, Ma Y, Zhang L, Jie J, Yang P, Yang Y. Immune-cell-mediated tissue engineering strategies for peripheral nerve injury and regeneration. J Mater Chem B 2024; 12:2217-2235. [PMID: 38345580 DOI: 10.1039/d3tb02557h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
During the process of peripheral nerve repair, there are many complex pathological and physiological changes, including multi-cellular responses and various signaling molecules, and all these events establish a dynamic microenvironment for axon repair, regeneration, and target tissue/organ reinnervation. The immune system plays an indispensable role in the process of nerve repair and function recovery. An effective immune response not only involves innate-immune and adaptive-immune cells but also consists of chemokines and cytokines released by these immune cells. The elucidation of the orchestrated interplay of immune cells with nerve regeneration and functional restoration is meaningful for the exploration of therapeutic strategies. This review mainly enumerates the general immune cell response to peripheral nerve injury and focuses on their contributions to functional recovery. The tissue engineering-mediated strategies to regulate macrophages and T cells through physical and biochemical factors combined with scaffolds are discussed. The dynamic immune responses during peripheral nerve repair and immune-cell-mediated tissue engineering methods are presented, which provide a new insight and inspiration for immunomodulatory therapies in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Xueying Zhao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Hui Deng
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yuan Feng
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yuehan Wang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Xiaomin Yao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yuyang Ma
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Jing Jie
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nantong University, 226001, Nantong, P. R. China.
| | - Pengxiang Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 226001, Nantong, P. R. China.
| |
Collapse
|
20
|
Thind MK, Uhlig HH, Glogauer M, Palaniyar N, Bourdon C, Gwela A, Lancioni CL, Berkley JA, Bandsma RHJ, Farooqui A. A metabolic perspective of the neutrophil life cycle: new avenues in immunometabolism. Front Immunol 2024; 14:1334205. [PMID: 38259490 PMCID: PMC10800387 DOI: 10.3389/fimmu.2023.1334205] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Neutrophils are the most abundant innate immune cells. Multiple mechanisms allow them to engage a wide range of metabolic pathways for biosynthesis and bioenergetics for mediating biological processes such as development in the bone marrow and antimicrobial activity such as ROS production and NET formation, inflammation and tissue repair. We first discuss recent work on neutrophil development and functions and the metabolic processes to regulate granulopoiesis, neutrophil migration and trafficking as well as effector functions. We then discuss metabolic syndromes with impaired neutrophil functions that are influenced by genetic and environmental factors of nutrient availability and usage. Here, we particularly focus on the role of specific macronutrients, such as glucose, fatty acids, and protein, as well as micronutrients such as vitamin B3, in regulating neutrophil biology and how this regulation impacts host health. A special section of this review primarily discusses that the ways nutrient deficiencies could impact neutrophil biology and increase infection susceptibility. We emphasize biochemical approaches to explore neutrophil metabolism in relation to development and functions. Lastly, we discuss opportunities and challenges to neutrophil-centered therapeutic approaches in immune-driven diseases and highlight unanswered questions to guide future discoveries.
Collapse
Affiliation(s)
- Mehakpreet K Thind
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
| | - Holm H Uhlig
- Translational Gastroenterology Unit, Experimental Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Department of Dental Oncology and Maxillofacial Prosthetics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Nades Palaniyar
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Celine Bourdon
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
| | - Agnes Gwela
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Centre for Geographic Medicine Research, Kilifi, Kenya
| | - Christina L Lancioni
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - James A Berkley
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Centre for Geographic Medicine Research, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - Robert H J Bandsma
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
- Laboratory of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON, Canada
| | - Amber Farooqui
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
- Omega Laboratories Inc, Mississauga, ON, Canada
| |
Collapse
|
21
|
Zhou L, Li Y, You J, Wu C, Zuo L, Chen Y, Kang L, Zhou Z, Huang R, Wu S. Salmonella spvC gene suppresses macrophage/neutrophil antibacterial defense mediated by gasdermin D. Inflamm Res 2024; 73:19-33. [PMID: 38135851 DOI: 10.1007/s00011-023-01818-9] [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: 08/29/2023] [Revised: 10/15/2023] [Accepted: 11/06/2023] [Indexed: 12/24/2023] Open
Abstract
OBJECTIVE Salmonella enterica serovar Typhimurium (S. Typhimurium) is a representative model organism for investigating host-pathogen interactions. It was reported that S. Typhimurium spvC gene alleviated intestinal inflammation to aggravate systemic infection, while the precise mechanisms remain unclear. In this study, the influence of spvC on the antibacterial defense of macrophage/neutrophil mediated by gasdermin D (GSDMD) was investigated. METHODS Mouse macrophage-like cell lines J774A.1 and RAW264.7, neutrophil-like cells derived from HL-60 cells (human promyletic leukemia cell lines) were infected with S. Typhimurium wild type, spvC deletion and complemented strains. Cell death was evaluated by LDH release and Annexin V-FITC/PI staining. Macrophage pyroptosis and neutrophil NETosis were detected by western blotting, live cell imaging and ELISA. Flow cytometry was used to assess the impact of spvC on macrophage-neutrophil cooperation in macrophage (dTHP-1)-neutrophil (dHL-60) co-culture model pretreated with GSDMD inhibitor disulfiram. Wild-type and Gsdmd-/- C57BL/6J mice were utilized for in vivo assay. The degree of phagocytes infiltration and inflammation were analyzed by immunofluorescence and transmission electron microscopy. RESULTS Here we find that spvC inhibits pyroptosis in macrophages via Caspase-1/Caspase-11 dependent canonical and non-canonical pathways, and restrains neutrophil extracellular traps extrusion in GSDMD-dependent manner. Moreover, spvC could ameliorate macrophages/neutrophils infiltration and cooperation in the inflammatory response mediated by GSDMD to combat Salmonella infection. CONCLUSIONS Our findings highlight the antibacterial activity of GSDMD in phagocytes and reveal a novel pathogenic mechanism employed by spvC to counteract this host defense, which may shed new light on designing effective therapeutics to control S. Typhimurium infection.
Collapse
Affiliation(s)
- Liting Zhou
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
- Center of Clinical Laboratory, Dushu Lake Hospital, Affiliated to Soochow University, Suzhou, China
| | - Yuanyuan Li
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Medical Microbiology, Experimental Center, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jiayi You
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chaoyi Wu
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Lingli Zuo
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
- Medical Research Center, The People's Hospital of Suzhou New District, Suzhou, China
| | - Yilin Chen
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Li Kang
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhengyu Zhou
- Laboratory Animal Center, Suzhou Medical College of Soochow University, Suzhou, China
| | - Rui Huang
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Shuyan Wu
- Department of Medical Microbiology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
22
|
Arciola CR, Ravaioli S, Mirzaei R, Dolzani P, Montanaro L, Daglia M, Campoccia D. Biofilms in Periprosthetic Orthopedic Infections Seen through the Eyes of Neutrophils: How Can We Help Neutrophils? Int J Mol Sci 2023; 24:16669. [PMID: 38068991 PMCID: PMC10706149 DOI: 10.3390/ijms242316669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Despite advancements in our knowledge of neutrophil responses to planktonic bacteria during acute inflammation, much remains to be elucidated on how neutrophils deal with bacterial biofilms in implant infections. Further complexity transpires from the emerging findings on the role that biomaterials play in conditioning bacterial adhesion, the variety of biofilm matrices, and the insidious measures that biofilm bacteria devise against neutrophils. Thus, grasping the entirety of neutrophil-biofilm interactions occurring in periprosthetic tissues is a difficult goal. The bactericidal weapons of neutrophils consist of the following: ready-to-use antibacterial proteins and enzymes stored in granules; NADPH oxidase-derived reactive oxygen species (ROS); and net-like structures of DNA, histones, and granule proteins, which neutrophils extrude to extracellularly trap pathogens (the so-called NETs: an allusive acronym for "neutrophil extracellular traps"). Neutrophils are bactericidal (and therefore defensive) cells endowed with a rich offensive armamentarium through which, if frustrated in their attempts to engulf and phagocytose biofilms, they can trigger the destruction of periprosthetic bone. This study speculates on how neutrophils interact with biofilms in the dramatic scenario of implant infections, also considering the implications of this interaction in view of the design of new therapeutic strategies and functionalized biomaterials, to help neutrophils in their arduous task of managing biofilms.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Stefano Ravaioli
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Paolo Dolzani
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
| | - Lucio Montanaro
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy;
| | - Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| |
Collapse
|
23
|
Goral A, Sledz M, Manda-Handzlik A, Cieloch A, Wojciechowska A, Lachota M, Mroczek A, Demkow U, Zagozdzon R, Matusik K, Wachowska M, Muchowicz A. Regulatory T cells contribute to the immunosuppressive phenotype of neutrophils in a mouse model of chronic lymphocytic leukemia. Exp Hematol Oncol 2023; 12:89. [PMID: 37817276 PMCID: PMC10563345 DOI: 10.1186/s40164-023-00452-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 09/29/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Impaired neutrophil activity is an important issue in chronic lymphocytic leukemia (CLL), as it contributes to a dysfunctional immune response leading to life-threatening infections in patients. Some features typical of CLL neutrophils, e.g., the B-cell-supportive secretion profile, have already been described. However, most of these studies were performed on cells isolated from peripheral blood. It is still unclear which molecular factors and cell types are involved in shaping neutrophil function and phenotype in the CLL microenvironment. Since regulatory T cells (Treg) play an important role in CLL progression and influence the activity of neutrophils, we investigated the crosstalk between Treg and neutrophils in the spleen using a murine model of CLL. METHODS In this work, we used an Eµ-TCL1 mouse model of human CLL. For our in vivo and ex vivo experiments, we inoculated wild-type mice with TCL1 leukemic cells isolated from Eµ-TCL1 transgenic mice and then monitored disease progression by detecting leukemic cells in peripheral blood. We analyzed both the phenotype and activity of neutrophils isolated from the spleens of TCL1 leukemia-bearing mice. To investigate the interrelation between Treg and neutrophils in the leukemia microenvironment, we performed experiments using TCL1-injected DEREG mice with Treg depletion or RAG2KO mice with adoptively transferred TCL1 cells alone or together with Treg. RESULTS The obtained results underline the plasticity of the neutrophil phenotype, observed under the influence of leukemic cells alone and depending on the presence of Treg. In particular, Treg affect the expression of CD62L and IL-4 receptor in neutrophils, both of which are crucial for the function of these cells. Additionally, we show that Treg depletion and IL-10 neutralization induce changes in the leukemia microenvironment, partially restoring the "healthy" phenotype of neutrophils. CONCLUSIONS Altogether, the results indicate that the crosstalk between Treg and neutrophils in CLL may play an important role in CLL progression by interfering with the immune response.
Collapse
Affiliation(s)
- Agnieszka Goral
- Department of Immunology, Medical University of Warsaw, Warsaw, 02-097, Poland
| | - Marta Sledz
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, 02-097, Poland
- Doctoral School, Medical University of Warsaw, Warsaw, 02-091, Poland
| | - Aneta Manda-Handzlik
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, 02-091, Poland
| | - Adrianna Cieloch
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, 02-091, Poland
| | - Alicja Wojciechowska
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, 02-097, Poland
| | - Mieszko Lachota
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, 02-097, Poland
- Department of Ophthalmology, Children's Memorial Health Institute, Warsaw, 04-730, Poland
| | - Agnieszka Mroczek
- Doctoral School, Medical University of Warsaw, Warsaw, 02-091, Poland
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, 02-091, Poland
| | - Urszula Demkow
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, 02-091, Poland
| | - Radoslaw Zagozdzon
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, 02-097, Poland
| | - Katarzyna Matusik
- Doctoral School, Medical University of Warsaw, Warsaw, 02-091, Poland
| | - Malgorzata Wachowska
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, 02-091, Poland
| | - Angelika Muchowicz
- Department of Immunology, Medical University of Warsaw, Warsaw, 02-097, Poland.
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, 02-097, Poland.
| |
Collapse
|
24
|
Luo J, Wang Z, Tang C, Yin Z, Huang J, Ruan D, Fei Y, Wang C, Mo X, Li J, Zhang J, Fang C, Li J, Chen X, Shen W. Animal model for tendinopathy. J Orthop Translat 2023; 42:43-56. [PMID: 37637777 PMCID: PMC10450357 DOI: 10.1016/j.jot.2023.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/18/2023] [Accepted: 06/30/2023] [Indexed: 08/29/2023] Open
Abstract
Background Tendinopathy is a common motor system disease that leads to pain and reduced function. Despite its prevalence, our mechanistic understanding is incomplete, leading to limited efficacy of treatment options. Animal models contribute significantly to our understanding of tendinopathy and some therapeutic options. However, the inadequacies of animal models are also evident, largely due to differences in anatomical structure and the complexity of human tendinopathy. Different animal models reproduce different aspects of human tendinopathy and are therefore suitable for different scenarios. This review aims to summarize the existing animal models of tendinopathy and to determine the situations in which each model is appropriate for use, including exploring disease mechanisms and evaluating therapeutic effects. Methods We reviewed relevant literature in the PubMed database from January 2000 to December 2022 using the specific terms ((tendinopathy) OR (tendinitis)) AND (model) AND ((mice) OR (rat) OR (rabbit) OR (lapin) OR (dog) OR (canine) OR (sheep) OR (goat) OR (horse) OR (equine) OR (pig) OR (swine) OR (primate)). This review summarized different methods for establishing animal models of tendinopathy and classified them according to the pathogenesis they simulate. We then discussed the advantages and disadvantages of each model, and based on this, identified the situations in which each model was suitable for application. Results For studies that aim to study the pathophysiology of tendinopathy, naturally occurring models, treadmill models, subacromial impingement models and metabolic models are ideal. They are closest to the natural process of tendinopathy in humans. For studies that aim to evaluate the efficacy of possible treatments, the selection should be made according to the pathogenesis simulated by the modeling method. Existing tendinopathy models can be classified into six types according to the pathogenesis they simulate: extracellular matrix synthesis-decomposition imbalance, inflammation, oxidative stress, metabolic disorder, traumatism and mechanical load. Conclusions The critical factor affecting the translational value of research results is whether the selected model is matched with the research purpose. There is no single optimal model for inducing tendinopathy, and researchers must select the model that is most appropriate for the study they are conducting. The translational potential of this article The critical factor affecting the translational value of research results is whether the animal model used is compatible with the research purpose. This paper provides a rationale and practical guide for the establishment and selection of animal models of tendinopathy, which is helpful to improve the clinical transformation ability of existing models and develop new models.
Collapse
Affiliation(s)
- Junchao Luo
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Zetao Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Chenqi Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| | - Zi Yin
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jiayun Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Dengfeng Ruan
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Yang Fei
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Canlong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Xianan Mo
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jiajin Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
| | - Jun Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Department of Orthopedics, Longquan People's Hospital, Zhejiang, 323799, China
| | - Cailian Fang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
| | - Jianyou Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Department of Orthopedics, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Zhejiang University Huzhou Hospital, 313000, Huzhou, Zhejiang, China
| | - Xiao Chen
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, 310058, Hangzhou City, Zhejiang Province, China
- Sports Medicine Institute of Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, 315825, Hangzhou, Zhejiang, China
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Sikora JP, Karawani J, Sobczak J. Neutrophils and the Systemic Inflammatory Response Syndrome (SIRS). Int J Mol Sci 2023; 24:13469. [PMID: 37686271 PMCID: PMC10488036 DOI: 10.3390/ijms241713469] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
We are not entirely able to understand, assess, and modulate the functioning of the immune system in clinical situations that lead to a systemic inflammatory response. In the search for diagnostic and treatment strategies (which are still far from perfect), it became very important to study the pathogenesis and participation of endogenous inflammation mediators. This study attempts to more precisely establish the role of neutrophils in individual phenomena occurring during an inflammatory and anti-inflammatory reaction, taking into account their cidal, immunoregulatory, and reparative abilities. Pro- and anticoagulatory properties of endothelium in systemic inflammatory response syndrome (SIRS) are emphasised, along with the resulting clinical implications (the application of immunotherapy using mesenchymal stem/stromal cells (MSCs) or IL-6 antagonists in sepsis and COVID-19 treatment, among others). Special attention is paid to reactive oxygen species (ROS), produced by neutrophils activated during "respiratory burst" in the course of SIRS; the protective and pathogenic role of these endogenous mediators is highlighted. Moreover, clinically useful biomarkers of SIRS (neutrophil extracellular traps, cell-free DNA, DAMP, TREMs, NGAL, miRNA, selected cytokines, ROS, and recognised markers of endothelial damage from the group of adhesins by means of immunohistochemical techniques) related to the neutrophils are presented, and their role in the diagnosing and forecasting of sepsis, burn disease, and COVID-19 is emphasised. Finally, examples of immunomodulation of sepsis and antioxidative thermal injury therapy are presented.
Collapse
Affiliation(s)
- Janusz P. Sikora
- Department of Paediatric Emergency Medicine, 2nd Chair of Paediatrics, Central Clinical Hospital, Medical University of Łódź, ul. Sporna 36/50, 91-738 Łódź, Poland;
| | - Jakub Karawani
- Faculty of Medicine, Lazarski University, ul. Świeradowska 43, 02-662 Warsaw, Poland;
| | - Jarosław Sobczak
- Department of Paediatric Emergency Medicine, 2nd Chair of Paediatrics, Central Clinical Hospital, Medical University of Łódź, ul. Sporna 36/50, 91-738 Łódź, Poland;
- Department of Management and Logistics in Healthcare, Medical University of Łódź, ul. Lindleya 6, 90-131 Łódź, Poland
| |
Collapse
|
26
|
Main EN, Cruz TM, Bowlin GL. Mitochondria as a therapeutic: a potential new frontier in driving the shift from tissue repair to regeneration. Regen Biomater 2023; 10:rbad070. [PMID: 37663015 PMCID: PMC10468651 DOI: 10.1093/rb/rbad070] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/12/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023] Open
Abstract
Fibrosis, or scar tissue development, is associated with numerous pathologies and is often considered a worst-case scenario in terms of wound healing or the implantation of a biomaterial. All that remains is a disorganized, densely packed and poorly vascularized bundle of connective tissue, which was once functional tissue. This creates a significant obstacle to the restoration of tissue function or integration with any biomaterial. Therefore, it is of paramount importance in tissue engineering and regenerative medicine to emphasize regeneration, the successful recovery of native tissue function, as opposed to repair, the replacement of the native tissue (often with scar tissue). A technique dubbed 'mitochondrial transplantation' is a burgeoning field of research that shows promise in in vitro, in vivo and various clinical applications in preventing cell death, reducing inflammation, restoring cell metabolism and proper oxidative balance, among other reported benefits. However, there is currently a lack of research regarding the potential for mitochondrial therapies within tissue engineering and regenerative biomaterials. Thus, this review explores these promising findings and outlines the potential for mitochondrial transplantation-based therapies as a new frontier of scientific research with respect to driving regeneration in wound healing and host-biomaterial interactions, the current successes of mitochondrial transplantation that warrant this potential and the critical questions and remaining obstacles that remain in the field.
Collapse
Affiliation(s)
- Evan N Main
- Department of Biomedical Engineering, University of Memphis, 330 Engineering Technology Building, Memphis, TN 38152, USA
| | - Thaiz M Cruz
- Department of Biomedical Engineering, University of Memphis, 330 Engineering Technology Building, Memphis, TN 38152, USA
| | - Gary L Bowlin
- Department of Biomedical Engineering, University of Memphis, 330 Engineering Technology Building, Memphis, TN 38152, USA
| |
Collapse
|
27
|
Almeida FS, Vanderley SER, Comberlang FC, Andrade AGD, Cavalcante-Silva LHA, Silva EDS, Palmeira PHDS, Amaral IPGD, Keesen TSL. Leishmaniasis: Immune Cells Crosstalk in Macrophage Polarization. Trop Med Infect Dis 2023; 8:tropicalmed8050276. [PMID: 37235324 DOI: 10.3390/tropicalmed8050276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Leishmaniasis is a complex infectious parasitic disease caused by protozoa of the genus Leishmania, belonging to a group of neglected tropical diseases. It establishes significant global health challenges, particularly in socio-economically disadvantaged regions. Macrophages, as innate immune cells, play a crucial role in initiating the inflammatory response against the pathogens responsible for this disease. Macrophage polarization, the process of differentiating macrophages into pro-inflammatory (M1) or anti-inflammatory (M2) phenotypes, is essential for the immune response in leishmaniasis. The M1 phenotype is associated with resistance to Leishmania infection, while the M2 phenotype is predominant in susceptible environments. Notably, various immune cells, including T cells, play a significant role in modulating macrophage polarization by releasing cytokines that influence macrophage maturation and function. Furthermore, other immune cells can also impact macrophage polarization in a T-cell-independent manner. Therefore, this review comprehensively examines macrophage polarization's role in leishmaniasis and other immune cells' potential involvement in this intricate process.
Collapse
Affiliation(s)
- Fernanda Silva Almeida
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil
| | - Shayenne Eduarda Ramos Vanderley
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil
| | - Fernando Cézar Comberlang
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil
| | - Arthur Gomes de Andrade
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil
| | - Luiz Henrique Agra Cavalcante-Silva
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil
| | - Edson Dos Santos Silva
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil
| | - Pedro Henrique de Sousa Palmeira
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil
| | - Ian P G do Amaral
- Laboratory of Biochemistry, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil
| | - Tatjana S L Keesen
- Immunology of Infectious Diseases Laboratory, Department of Cellular and Molecular Biology, Federal University of Paraiba, João Pessoa 58051-900, PB, Brazil
| |
Collapse
|
28
|
Prognostic Biomarkers Identification in Esophageal Cancer Based on WGCNA and Single-Cell Analysis. DISEASE MARKERS 2022; 2022:6595778. [PMID: 36569220 PMCID: PMC9788878 DOI: 10.1155/2022/6595778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 11/11/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022]
Abstract
Background Esophageal cancer is one of the most common cancers worldwide. Dysregulation of genes plays an important role in cancer. In this study, we aimed to investigate the prognostic biomarkers in esophageal cancer based on comprehensive bioinformatics analysis including WGCNA and single cell analysis. Methods RNA sequencing data of esophageal cancer was downloaded from GSE75241 dataset in the GEO database. We also selected esophageal cancer patients from public databases (Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA)). WGCNA was used to construct a scale-free coexpression network of genes. Multifactor Cox analysis model was constructed as the prognostic model in esophageal cancer. Furthermore, single-cell gene analysis was used to discover the mechanism of hub genes in esophageal cancer. Results WGCNA discovered 182 genes for further analysis. Among 182 genes, four genes including ANGPT2, VCAN, MS4A4A, and FOS had significant prognostic value in esophageal cancer. In single cell analysis, seven types of cells subsets were distinguished including T cells, B cells, NK cells, monocytes, macrophages, DCs, neutrophils. The expression of four hub genes (ANGPT2, VCAN, MS4A4A, and FOS) in inflammatory cell subsets was evaluated, respectively. Hub genes were correlated with inflammatory cells in esophageal cancer. In addition, the subgroups of specific inflammatory cells such as macrophages, monocytes, and DCs were analyzed to identify the function of hub genes, either. Hub genes were correlated with differentiation of inflammatory cells including monocytes, macrophages, and DCs in tumor environment. Conclusions We identified specific hub genes correlated with prognosis of esophageal cancer. These hub genes play critical roles by regulating inflammatory cells status in esophageal cancer.
Collapse
|
29
|
Bourgon C, Albin AS, Ando-Grard O, Da Costa B, Domain R, Korkmaz B, Klonjkowski B, Le Poder S, Meunier N. Neutrophils play a major role in the destruction of the olfactory epithelium during SARS-CoV-2 infection in hamsters. Cell Mol Life Sci 2022; 79:616. [PMID: 36460750 PMCID: PMC9734468 DOI: 10.1007/s00018-022-04643-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 11/02/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022]
Abstract
The loss of smell (anosmia) related to SARS-CoV-2 infection is one of the most common symptoms of COVID-19. Olfaction starts in the olfactory epithelium mainly composed of olfactory sensory neurons surrounded by supporting cells called sustentacular cells. It is now clear that the loss of smell is related to the massive infection by SARS-CoV-2 of the sustentacular cells in the olfactory epithelium leading to its desquamation. However, the molecular mechanism behind the destabilization of the olfactory epithelium is less clear. Using golden Syrian hamsters infected with an early circulating SARS-CoV-2 strain harboring the D614G mutation in the spike protein; we show here that rather than being related to a first wave of apoptosis as proposed in previous studies, the innate immune cells play a major role in the destruction of the olfactory epithelium. We observed that while apoptosis remains at a low level in the damaged area of the infected epithelium, the latter is invaded by Iba1+ cells, neutrophils and macrophages. By depleting the neutrophil population or blocking the activity of neutrophil elastase-like proteinases, we could reduce the damage induced by the SARS-CoV-2 infection. Surprisingly, the impairment of neutrophil activity led to a decrease in SARS-CoV-2 infection levels in the olfactory epithelium. Our results indicate a counterproductive role of neutrophils leading to the release of infected cells in the lumen of the nasal cavity and thereby enhanced spreading of the virus in the early phase of the SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Clara Bourgon
- Unité de Virologie et Immunologie Moléculaires (UR892), INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Audrey St Albin
- Unité de Virologie et Immunologie Moléculaires (UR892), INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Ophélie Ando-Grard
- Unité de Virologie et Immunologie Moléculaires (UR892), INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Bruno Da Costa
- Unité de Virologie et Immunologie Moléculaires (UR892), INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Roxane Domain
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, 37032, Tours, France
| | - Brice Korkmaz
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, 37032, Tours, France
| | - Bernard Klonjkowski
- UMR 1161 Virologie, INRAE-ENVA-ANSES, École Nationale Vétérinaire d'Alfort, Maisons-Alfort, 94704, Paris, France
| | - Sophie Le Poder
- UMR 1161 Virologie, INRAE-ENVA-ANSES, École Nationale Vétérinaire d'Alfort, Maisons-Alfort, 94704, Paris, France
| | - Nicolas Meunier
- Unité de Virologie et Immunologie Moléculaires (UR892), INRAE, Université Paris-Saclay, Jouy-en-Josas, France.
| |
Collapse
|
30
|
Ren H, Xia X, Dai X, Dai Y. The role of neuroplastin65 in macrophage against E. coli infection in mice. Mol Immunol 2022; 150:78-89. [PMID: 36007354 DOI: 10.1016/j.molimm.2022.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/20/2022] [Accepted: 08/04/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Innate immune response constitutes the first line of defense against pathogens. Inflammatory responses involve close contact between different populations of cells. These adhesive interactions mediate migration of cells to sites of infection leading the effective action of cells within the lesions. Cell adhesion molecules are critical to controlling immune response mediating cell adhesion or chemotaxis, as well as coordinating actin-based cell motility during phagocytosis and chemotaxis. Recently, a newly discovered neuroplastin (Np) adhesion molecule is found to play an important role in the nervous system. However, there is limited information on Np functions in immune response. To understand how Np is involved in innate immune response, a mouse model of intraperitoneal infection was established to investigate the effect of Np on macrophage-mediated clearance of E. coli infection and its possible molecular mechanisms. METHODS Specific deficiency mice with Nptn gene controlling Np65 isoform were employed in this study. The expression levels of mRNA and proteins were detected by qPCR and western blot, or evaluated by flow cytometry. The expression level of NO and ROS were measured with their specific indicators. Cell cycle and apoptosis were detected by specific detection kits. Acid phosphatase activity was measured by flow cytometry after labelling with LysoRed fluorescent probe. Bone marrow derived macrophages (BMDMs) were isolated from bone marrow of mice hind legs. Cell proliferation was detected by CCK8 assay. Cell migration was measured by wound healing assay or transwell assay. RESULTS The lethal dose of E. coli infection in Np65-/- mice dropped to the half of lethal dose in WT mice. The bacterial load in the spleen, kidney and liver from Np65-/- mice were significantly higher than that from WT mice, which were due to the dramatic reduction of NO and ROS production in phagocytes from Np65-/- mice. Np65 gene deficiency remarkably impaired phagocytosis and function of lysosome in macrophage. Furthermore, Np65 molecule was involved in maturation and proliferation, even in migration and chemotaxis of BMDM in vitro. CONCLUSION This study for the first time demonstrates that Np is involved in multi-function of phagocytes during bacterial infection, proposing that Np adhesion molecule plays a critical role in clearing pathogen infection in innate immunity.
Collapse
Affiliation(s)
- Huan Ren
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, and Department of Immunology and Microbiology, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, China
| | - Xiaoxue Xia
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, and Department of Immunology and Microbiology, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, China
| | - Xueting Dai
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, and Department of Immunology and Microbiology, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, China
| | - Yalei Dai
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, and Department of Immunology and Microbiology, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, China.
| |
Collapse
|
31
|
Turner CT, Pawluk M, Bolsoni J, Zeglinski MR, Shen Y, Zhao H, Ponomarev T, Richardson KC, West CR, Papp A, Granville DJ. Sulfaphenazole reduces thermal and pressure injury severity through rapid restoration of tissue perfusion. Sci Rep 2022; 12:12622. [PMID: 35871073 PMCID: PMC9308818 DOI: 10.1038/s41598-022-16512-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/11/2022] [Indexed: 11/20/2022] Open
Abstract
Pressure injuries, also known as pressure ulcers, are regions of localized damage to the skin and/or underlying tissue. Repeated rounds of ischemia-reperfusion (I/R) have a major causative role for tissue damage in pressure injury. Ischemia prevents oxygen/nutrient supply, and restoration of blood flow induces a burst of reactive oxygen species that damages blood vessels, surrounding tissues and can halt blood flow return. Minimizing the consequences of repeated I/R is expected to provide a protective effect against pressure injury. Sulfaphenazole (SP), an off patent sulfonamide antibiotic, is a potent CYP 2C6 and CYP 2C9 inhibitor, functioning to decrease post-ischemic vascular dysfunction and increase blood flow. The therapeutic effect of SP on pressure injury was therefore investigated in apolipoprotein E knockout mice, a model of aging susceptible to ischemic injury, which were subjected to repeated rounds of I/R-induced skin injury. SP reduced overall severity, improved wound closure and increased wound tensile strength compared to vehicle-treated controls. Saliently, SP restored tissue perfusion in and around the wound rapidly to pre-injury levels, decreased tissue hypoxia, and reduced both inflammation and fibrosis. SP also demonstrated bactericidal activity through enhanced M1 macrophage activity. The efficacy of SP in reducing thermal injury severity was also demonstrated. SP is therefore a potential therapeutic option for pressure injury and other ischemic skin injuries.
Collapse
Affiliation(s)
- Christopher T. Turner
- grid.17091.3e0000 0001 2288 9830International Collaboration on Repair Discoveries (ICORD) Centre, Blusson Spinal Cord Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Rm 4470, 818 West 10th Ave., Vancouver, BC V5Z 1M9 Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Megan Pawluk
- grid.17091.3e0000 0001 2288 9830International Collaboration on Repair Discoveries (ICORD) Centre, Blusson Spinal Cord Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Rm 4470, 818 West 10th Ave., Vancouver, BC V5Z 1M9 Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Juliana Bolsoni
- grid.17091.3e0000 0001 2288 9830International Collaboration on Repair Discoveries (ICORD) Centre, Blusson Spinal Cord Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Rm 4470, 818 West 10th Ave., Vancouver, BC V5Z 1M9 Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Matthew R. Zeglinski
- grid.17091.3e0000 0001 2288 9830International Collaboration on Repair Discoveries (ICORD) Centre, Blusson Spinal Cord Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Rm 4470, 818 West 10th Ave., Vancouver, BC V5Z 1M9 Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Yue Shen
- grid.17091.3e0000 0001 2288 9830International Collaboration on Repair Discoveries (ICORD) Centre, Blusson Spinal Cord Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Rm 4470, 818 West 10th Ave., Vancouver, BC V5Z 1M9 Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Hongyan Zhao
- grid.17091.3e0000 0001 2288 9830International Collaboration on Repair Discoveries (ICORD) Centre, Blusson Spinal Cord Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Rm 4470, 818 West 10th Ave., Vancouver, BC V5Z 1M9 Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Tatjana Ponomarev
- grid.17091.3e0000 0001 2288 9830Centre for Heart Lung Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC Canada
| | - Katlyn C. Richardson
- grid.17091.3e0000 0001 2288 9830International Collaboration on Repair Discoveries (ICORD) Centre, Blusson Spinal Cord Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Rm 4470, 818 West 10th Ave., Vancouver, BC V5Z 1M9 Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Christopher R. West
- grid.17091.3e0000 0001 2288 9830International Collaboration on Repair Discoveries (ICORD) Centre, Blusson Spinal Cord Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Rm 4470, 818 West 10th Ave., Vancouver, BC V5Z 1M9 Canada ,grid.17091.3e0000 0001 2288 9830Department of Cell and Physiological Sciences, University of British Columbia, Vancouver, BC Canada
| | - Anthony Papp
- grid.17091.3e0000 0001 2288 9830Department of Surgery, University of British Columbia, Vancouver, BC Canada
| | - David J. Granville
- grid.17091.3e0000 0001 2288 9830International Collaboration on Repair Discoveries (ICORD) Centre, Blusson Spinal Cord Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Rm 4470, 818 West 10th Ave., Vancouver, BC V5Z 1M9 Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada ,grid.417243.70000 0004 0384 4428British Columbia Professional Firefighters’ Burn and Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, BC Canada
| |
Collapse
|
32
|
Liu X, Dou G, Li Z, Wang X, Jin R, Liu Y, Kuang H, Huang X, Yang X, Yang X, Liu S, Wu M, Guo H, Ding F, Xu H, Liu S, Jin Y, Xuan K. Hybrid Biomaterial Initiates Refractory Wound Healing via Inducing Transiently Heightened Inflammatory Responses. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105650. [PMID: 35603963 PMCID: PMC9313498 DOI: 10.1002/advs.202105650] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/15/2022] [Indexed: 05/22/2023]
Abstract
Inflammation plays a crucial role in triggering regeneration, while inadequate or chronic inflammation hinders the regenerative process, resulting in refractory wounds. Inspired by the ideal regeneration mode in lower vertebrates and the human oral mucosa, realigning dysregulated inflammation to a heightened and acute response provides a promising option for refractory wound therapy. Neutrophils play important roles in inflammation initiation and resolution. Here, a hybrid biomaterial is used to stimulate transiently heightened inflammatory responses by precise tempospatial regulation of neutrophil recruitment and apoptosis. The hybrid biomaterial (Gel@fMLP/SiO2 -FasL) is constructed by loading of formyl-met-leu-phe (fMLP) and FasL-conjugated silica nanoparticles (SiO2 -FasL) into a pH-responsive hydrogel matrix. This composition enables burst release of fMLP to rapidly recruit neutrophils for heightened inflammation initiation. After neutrophils act to produce acids, the pH-responsive hydrogel degrades to expose SiO2 -FasL, which induces activated neutrophils apoptosis via FasL-Fas signaling triggering timely inflammation resolution. Apoptotic neutrophils are subsequently cleared by macrophages, and this efferocytosis activates key signalings to promote macrophage anti-inflammatory phenotypic transformation to drive regeneration. Ultimately, Gel@fMLP/SiO2 -FasL successfully promotes tissue regeneration by manipulating inflammation in critical-sized calvarial bone defects and diabetic cutaneous wound models. This work provides a new strategy for refractory wound therapy via inducing transiently heightened inflammatory responses.
Collapse
Affiliation(s)
- Xuemei Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral DiseasesDepartment of Preventive DentistrySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
- Department of Pediatric DentistrySchool and Hospital of StomatologyChina Medical UniversityLiaoning Provincial Key Laboratory of Oral DiseasesShenyang110002China
| | - Geng Dou
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Zihan Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral DiseasesDepartment of Preventive DentistrySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Xiangdong Wang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Ronghua Jin
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation and College of PharmacyGuangxi Medical UniversityNanning530021China
| | - Yao Liu
- Department of Pediatric DentistrySchool and Hospital of StomatologyChina Medical UniversityLiaoning Provincial Key Laboratory of Oral DiseasesShenyang110002China
| | - Huijuan Kuang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Xiaoyao Huang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral DiseasesDepartment of Preventive DentistrySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Xiaoxue Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral DiseasesDepartment of Preventive DentistrySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Xiaoshan Yang
- Stomatology HospitalSouthern Medical UniversityGuangzhou510280China
| | - Siying Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Meiling Wu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral DiseasesDepartment of Preventive DentistrySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Hao Guo
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral DiseasesDepartment of Preventive DentistrySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Feng Ding
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Haokun Xu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Yan Jin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral DiseasesCenter for Tissue EngineeringSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Kun Xuan
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral DiseasesDepartment of Preventive DentistrySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| |
Collapse
|
33
|
Shang L, Shao J, Ge S. Immunomodulatory Properties: The Accelerant of Hydroxyapatite-Based Materials for Bone Regeneration. Tissue Eng Part C Methods 2022; 28:377-392. [PMID: 35196904 DOI: 10.1089/ten.tec.2022.00111112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The immunoinflammatory response is the prerequisite step for wound healing and tissue regeneration, and the immunomodulatory effects of biomaterials have attracted increasing attention. Hydroxyapatite [Ca10(PO4)6(OH)2] (HAp), a common calcium phosphate ceramic, due to its structural and functional similarity to the inorganic constituent of natural bones, has been developed for different application purposes such as bone substitutes, tissue engineering scaffolds, and implant coatings. Recently, the interaction between HAp-based materials and the immune system (various immune cells), and the immunomodulatory effects of HAp-based materials on bone tissue regeneration have been explored extensively. Macrophages-mediated regenerative effect by HAp stimulation occupies the mainstream status of immunomodulatory strategies. The immunomodulation of HAp can be manipulated by tuning the physical, chemical, and biological cues such as surface functionalization (physical or chemical modifications), structural and textural characteristics (size, shape, and surface topography), and the incorporation of bioactive substances (cytokines, rare-earth elements, and bioactive ions). Therefore, HAp ceramic materials can contribute to bone regeneration by creating a favorable osteoimmune microenvironment, which would provide a more comprehensive theoretical basis for their further clinical applications. Considering the rapidly developed HAp-based materials as well as their excellent biological performances in the field of regenerative medicine, this review discusses the recent advances concerning the immunomodulatory methods for HAp-based biomaterials and their roles in bone tissue regeneration.
Collapse
Affiliation(s)
- Lingling Shang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Jinlong Shao
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Shaohua Ge
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
34
|
Arenas-Hernandez M, Romero R, Gershater M, Tao L, Xu Y, Garcia-Flores V, Pusod E, Miller D, Galaz J, Motomura K, Schwenkel G, Para R, Gomez-Lopez N. Specific innate immune cells uptake fetal antigen and display homeostatic phenotypes in the maternal circulation. J Leukoc Biol 2022; 111:519-538. [PMID: 34889468 PMCID: PMC8881318 DOI: 10.1002/jlb.5hi0321-179rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 10/25/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
Pregnancy represents a period when the mother undergoes significant immunological changes to promote tolerance of the fetal semi-allograft. Such tolerance results from the exposure of the maternal immune system to fetal antigens (Ags), a process that has been widely investigated at the maternal-fetal interface and in the adjacent draining lymph nodes. However, the peripheral mechanisms of maternal-fetal crosstalk are poorly understood. Herein, we hypothesized that specific innate immune cells interact with fetal Ags in the maternal circulation. To test this hypothesis, a mouse model was utilized in which transgenic male mice expressing the chicken ovalbumin (OVA) Ag under the beta-actin promoter were allogeneically mated with wild-type females to allow for tracking of the fetal Ag. Fetal Ag-carrying Ly6G+ and F4/80+ cells were identified in the maternal circulation, where they were more abundant in the second half of pregnancy. Such innate immune cells displayed unique phenotypes: while Ly6G+ cells expressed high levels of MHC-II and CD80 together with low levels of pro-inflammatory cytokines, F4/80+ cells up-regulated the expression of CD86 as well as the anti-inflammatory cytokines IL-10 and TGF-β. In vitro studies using allogeneic GFP+ placental particles revealed that maternal peripheral Ly6G+ and F4/80+ cells phagocytose fetal Ags in mid and late murine pregnancy. Importantly, cytotrophoblast-derived particles were also engulfed in vitro by CD15+ and CD14+ cells from women in the second and third trimester, providing translational evidence that this process also occurs in humans. Collectively, this study demonstrates novel interactions between specific maternal circulating innate immune cells and fetal Ags, thereby shedding light on the systemic mechanisms of maternal-fetal crosstalk.
Collapse
Affiliation(s)
- Marcia Arenas-Hernandez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
- Detroit Medical Center, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, FL, USA
| | - Meyer Gershater
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Li Tao
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Errile Pusod
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kenichiro Motomura
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - George Schwenkel
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert Para
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U. S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
35
|
Doolan R, Bouchery T. Hookworm infections: Reappraising the evidence for a role of Neutrophils in light of NETosis. Parasite Immunol 2022; 44:e12911. [PMID: 35124825 PMCID: PMC9285577 DOI: 10.1111/pim.12911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/27/2022] [Accepted: 02/03/2022] [Indexed: 11/26/2022]
Abstract
In Hookworm infection, neutrophils have long had the image of the villain, being recruited to the site of larval migration because of damage but participating themselves in tissue injury. With recent developments in neutrophil biology, there is an increasing body of evidence for the role of neutrophils as effector cells in hookworm immunity. In particular, their ability to release extracellular traps, or neutrophil extracellular traps (NETs), confer neutrophils a larvicidal activity. Here, we review recent evidence in this nascent field and discuss the avenue for future research on NETs/hookworm interactions.
Collapse
Affiliation(s)
- Rory Doolan
- Hookworm Immunobiology Laboratory Department of Medical Parasitology & Infection Biology Swiss Tropical and Public Health Institute Socinstrasse 57 4051 CH Basel Switzerland
| | - Tiffany Bouchery
- Hookworm Immunobiology Laboratory Department of Medical Parasitology & Infection Biology Swiss Tropical and Public Health Institute Socinstrasse 57 4051 CH Basel Switzerland
| |
Collapse
|
36
|
Herrero-Cervera A, Soehnlein O, Kenne E. Neutrophils in chronic inflammatory diseases. Cell Mol Immunol 2022; 19:177-191. [PMID: 35039631 PMCID: PMC8803838 DOI: 10.1038/s41423-021-00832-3] [Citation(s) in RCA: 338] [Impact Index Per Article: 112.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation is a component of many disease conditions that affect a large group of individuals worldwide. Chronic inflammation is characterized by persistent, low-grade inflammation and is increased in the aging population. Neutrophils are normally the first responders to acute inflammation and contribute to the resolution of inflammation. However, in chronic inflammation, the role of neutrophils is less well understood and has been described as either beneficial or detrimental, causing tissue damage and enhancing the immune response. Emerging evidence suggests that neutrophils are important players in several chronic diseases, such as atherosclerosis, diabetes mellitus, nonalcoholic fatty liver disease and autoimmune disorders. This review will highlight the interaction of neutrophils with other cells in the context of chronic inflammation, the contribution of neutrophils to selected chronic inflammatory diseases, and possible future therapeutic strategies.
Collapse
Affiliation(s)
- Andrea Herrero-Cervera
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Westfälische Wilhelms-Universität Münster, Münster, Germany.
| | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Westfälische Wilhelms-Universität Münster, Münster, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ellinor Kenne
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Neutrophil Extracellular Traps Facilitate A549 Cell Invasion and Migration in a Macrophage-Maintained Inflammatory Microenvironment. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8316525. [PMID: 35036439 PMCID: PMC8758275 DOI: 10.1155/2022/8316525] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/24/2021] [Accepted: 12/13/2021] [Indexed: 12/23/2022]
Abstract
Introduction The biological functions of neutrophil extracellular traps (NETs) in tumorigenesis have drawn an increasing amount of attention. This study explored the relationship between NETs and the inflammatory microenvironment in lung cancer cell invasion and metastasis. Methods NETs were quantified using myeloperoxidase (MPO–DNA) and immunofluorescence staining. Cytokine levels were measured using ELISA kits. THP-1 and A549 cells were used for in vitro experiments. Transwell and Matrigel assays were used to assess the invasion and migration abilities of the cells. Results Neutrophil infiltration and NET formation were observed in the lung cancer tissues. Compared with healthy controls, the level of MPO–DNA complexes in lung cancer patients increased remarkably and was positively correlated with peripheral blood neutrophil counts, smoking status, and poor prognosis. Increased circulating NET levels were also positively correlated with the levels of inflammatory cytokines, including IL-1β, IL-6, IL-18, and TNF-α. Neutrophils isolated from patients with lung cancer are more prone to NET release. NETs can promote the invasion and migration ability of THP-1 and A549 cell in coculture systems, while pretreatment with NET inhibitors can effectively reduce NET-induced invasion and metastasis. The ability of NETs to promote invasion and metastasis is partly dependent on macrophages. Conclusion Taken together, our study demonstrated that NETs facilitate A549 cell invasion and migration in a macrophage-maintained inflammatory microenvironment.
Collapse
|
38
|
Zhao XF, Huffman LD, Hafner H, Athaiya M, Finneran MC, Kalinski AL, Kohen R, Flynn C, Passino R, Johnson CN, Kohrman D, Kawaguchi R, Yang LJS, Twiss JL, Geschwind DH, Corfas G, Giger RJ. The injured sciatic nerve atlas (iSNAT), insights into the cellular and molecular basis of neural tissue degeneration and regeneration. eLife 2022; 11:80881. [PMID: 36515985 PMCID: PMC9829412 DOI: 10.7554/elife.80881] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Upon trauma, the adult murine peripheral nervous system (PNS) displays a remarkable degree of spontaneous anatomical and functional regeneration. To explore extrinsic mechanisms of neural repair, we carried out single-cell analysis of naïve mouse sciatic nerve, peripheral blood mononuclear cells, and crushed sciatic nerves at 1 day, 3 days, and 7 days following injury. During the first week, monocytes and macrophages (Mo/Mac) rapidly accumulate in the injured nerve and undergo extensive metabolic reprogramming. Proinflammatory Mo/Mac with a high glycolytic flux dominate the early injury response and rapidly give way to inflammation resolving Mac, programmed toward oxidative phosphorylation. Nerve crush injury causes partial leakiness of the blood-nerve barrier, proliferation of endoneurial and perineurial stromal cells, and entry of opsonizing serum proteins. Micro-dissection of the nerve injury site and distal nerve, followed by single-cell RNA-sequencing, identified distinct immune compartments, triggered by mechanical nerve wounding and Wallerian degeneration, respectively. This finding was independently confirmed with Sarm1-/- mice, in which Wallerian degeneration is greatly delayed. Experiments with chimeric mice showed that wildtype immune cells readily enter the injury site in Sarm1-/- mice, but are sparse in the distal nerve, except for Mo. We used CellChat to explore intercellular communications in the naïve and injured PNS and report on hundreds of ligand-receptor interactions. Our longitudinal analysis represents a new resource for neural tissue regeneration, reveals location- specific immune microenvironments, and reports on large intercellular communication networks. To facilitate mining of scRNAseq datasets, we generated the injured sciatic nerve atlas (iSNAT): https://cdb-rshiny.med.umich.edu/Giger_iSNAT/.
Collapse
Affiliation(s)
- Xiao-Feng Zhao
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States
| | - Lucas D Huffman
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States,Neuroscience Graduate Program, University of Michigan–Ann ArborAnn ArborUnited States
| | - Hannah Hafner
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States
| | - Mitre Athaiya
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States,Neuroscience Graduate Program, University of Michigan–Ann ArborAnn ArborUnited States
| | - Matthew C Finneran
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States,Neuroscience Graduate Program, University of Michigan–Ann ArborAnn ArborUnited States
| | - Ashley L Kalinski
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States
| | - Rafi Kohen
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States,Neuroscience Graduate Program, University of Michigan–Ann ArborAnn ArborUnited States
| | - Corey Flynn
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States
| | - Ryan Passino
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States
| | - Craig N Johnson
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States
| | - David Kohrman
- Kresge Hearing Institute, University of Michigan–Ann ArborAnn ArborUnited States
| | - Riki Kawaguchi
- Departments of Psychiatry and Neurology, University of California, Los AngelesLos AngelesUnited States
| | - Lynda JS Yang
- Department of Neurosurgery, University of Michigan-Ann ArborAnn ArborUnited States
| | - Jeffery L Twiss
- Department of Biological Sciences, University of South CarolinaColumbiaUnited States
| | - Daniel H Geschwind
- Department of Neurology, Program in Neurogenetics, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States,Department of Human Genetics,David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States,Institute of Precision Health, University of California, Los AngelesLos AngelesUnited States
| | - Gabriel Corfas
- Neuroscience Graduate Program, University of Michigan–Ann ArborAnn ArborUnited States,Kresge Hearing Institute, University of Michigan–Ann ArborAnn ArborUnited States,Department of Neurology, University of Michigan–Ann ArborAnn ArborUnited States
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan-Ann ArborAnn ArborUnited States,Neuroscience Graduate Program, University of Michigan–Ann ArborAnn ArborUnited States,Department of Neurology, University of Michigan–Ann ArborAnn ArborUnited States
| |
Collapse
|
39
|
Kuzheleva EA, Fedyunina VA, Garganeeva AA. [Patterns of immunological reactions in the pathogenesis of chronic heart failure: review]. KARDIOLOGIIA 2021; 61:94-104. [PMID: 35057726 DOI: 10.18087/cardio.2021.12.n1598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/01/2021] [Indexed: 06/14/2023]
Abstract
The immune system is essential for maintaining the homeostasis. At present, there is convincing evidence for participation of the immune system in the pathogenesis of cardiovascular pathology, including the final step of cardiovascular continuum, heart failure. Objective difficulties in understanding subtle processes of loss of the normal cardiac structure and function are based on the diversity of pathogenetic factors of development and progression of chronic heart failure (CHF) and the involvement of most organs and body systems. Russian and international scientists actively study issues of immune homeostasis, including the efficacy of current immune therapy. At the same time, available reports are largely uncompiled and reflect isolated parts of the immunopathogenesis of cardiovascular diseases. This review focuses on comprehensive elucidation of major patterns of immune processes in the CHF pathogenesis to form an integral view of the problem under study.
Collapse
Affiliation(s)
- E A Kuzheleva
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - V A Fedyunina
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - A A Garganeeva
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
40
|
Chan L, Karimi N, Morovati S, Alizadeh K, Kakish JE, Vanderkamp S, Fazel F, Napoleoni C, Alizadeh K, Mehrani Y, Minott JA, Bridle BW, Karimi K. The Roles of Neutrophils in Cytokine Storms. Viruses 2021; 13:v13112318. [PMID: 34835125 PMCID: PMC8624379 DOI: 10.3390/v13112318] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 12/16/2022] Open
Abstract
A cytokine storm is an abnormal discharge of soluble mediators following an inappropriate inflammatory response that leads to immunopathological events. Cytokine storms can occur after severe infections as well as in non-infectious situations where inflammatory cytokine responses are initiated, then exaggerated, but fail to return to homeostasis. Neutrophils, macrophages, mast cells, and natural killer cells are among the innate leukocytes that contribute to the pathogenesis of cytokine storms. Neutrophils participate as mediators of inflammation and have roles in promoting homeostatic conditions following pathological inflammation. This review highlights the advances in understanding the mechanisms governing neutrophilic inflammation against viral and bacterial pathogens, in cancers, and in autoimmune diseases, and how neutrophils could influence the development of cytokine storm syndromes. Evidence for the destructive potential of neutrophils in their capacity to contribute to the onset of cytokine storm syndromes is presented across a multitude of clinical scenarios. Further, a variety of potential therapeutic strategies that target neutrophils are discussed in the context of suppressing multiple inflammatory conditions.
Collapse
Affiliation(s)
- Lily Chan
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Negar Karimi
- Department of Clinical Sciences, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad 91779-4897, Iran;
| | - Solmaz Morovati
- Division of Biotechnology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz 71557-13876, Iran;
| | - Kasra Alizadeh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Julia E. Kakish
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Sierra Vanderkamp
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Fatemeh Fazel
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Christina Napoleoni
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Kimia Alizadeh
- Department of Diagnostic Medicine & Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA;
| | - Yeganeh Mehrani
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
- Department of Clinical Sciences, School of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad 91779-4897, Iran;
| | - Jessica A. Minott
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
| | - Byram W. Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
- Correspondence: (B.W.B.); (K.K.); Tel.: +1-519-824-4120 (ext. 54657) (B.W.B.); +1-519-824-4120 (ext. 54668) (K.K.)
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.C.); (J.E.K.); (S.V.); (F.F.); (C.N.); (Y.M.); (J.A.M.)
- Correspondence: (B.W.B.); (K.K.); Tel.: +1-519-824-4120 (ext. 54657) (B.W.B.); +1-519-824-4120 (ext. 54668) (K.K.)
| |
Collapse
|
41
|
CD5L deficiency attenuate acetaminophen-induced liver damage in mice via regulation of JNK and ERK signaling pathway. Cell Death Dis 2021; 7:342. [PMID: 34750342 PMCID: PMC8575892 DOI: 10.1038/s41420-021-00742-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/19/2021] [Accepted: 10/27/2021] [Indexed: 11/09/2022]
Abstract
CD5 molecule like (CD5L), a member of the scavenger receptor cysteine-rich domain superfamily, plays a critical role in immune homeostasis and inflammatory disease. Acetaminophen (APAP) is a safe and effective antipyretic analgesic. However, overdose may cause liver damage or even liver failure. APAP hepatotoxicity is characterized by extensive necrotic cell death and a sterile inflammatory response, in which the role of CD5L remains to be investigated. In this study, we found that the expression of CD5L was increased in the livers of mice after APAP overdose. Furthermore, CD5L deficiency reduced the increase of alanine transaminase (ALT) level, histopathologic lesion area, c-Jun N-terminal kinase (JNK)/extracellular signal-regulated kinase (ERK) phosphorylation level, Transferase-Mediated dUTP Nick End-Labeling positive (TUNEL+) cells proportion, vascular endothelial cell permeability and release of inflammatory cytokines induced by excess APAP. Therefore, our findings reveal that CD5L may be a potential therapeutic target for prevention and treatment of APAP-induced liver injury.
Collapse
|
42
|
Lafuente-Gracia L, Borgiani E, Nasello G, Geris L. Towards in silico Models of the Inflammatory Response in Bone Fracture Healing. Front Bioeng Biotechnol 2021; 9:703725. [PMID: 34660547 PMCID: PMC8514728 DOI: 10.3389/fbioe.2021.703725] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 09/07/2021] [Indexed: 12/21/2022] Open
Abstract
In silico modeling is a powerful strategy to investigate the biological events occurring at tissue, cellular and subcellular level during bone fracture healing. However, most current models do not consider the impact of the inflammatory response on the later stages of bone repair. Indeed, as initiator of the healing process, this early phase can alter the regenerative outcome: if the inflammatory response is too strongly down- or upregulated, the fracture can result in a non-union. This review covers the fundamental information on fracture healing, in silico modeling and experimental validation. It starts with a description of the biology of fracture healing, paying particular attention to the inflammatory phase and its cellular and subcellular components. We then discuss the current state-of-the-art regarding in silico models of the immune response in different tissues as well as the bone regeneration process at the later stages of fracture healing. Combining the aforementioned biological and computational state-of-the-art, continuous, discrete and hybrid modeling technologies are discussed in light of their suitability to capture adequately the multiscale course of the inflammatory phase and its overall role in the healing outcome. Both in the establishment of models as in their validation step, experimental data is required. Hence, this review provides an overview of the different in vitro and in vivo set-ups that can be used to quantify cell- and tissue-scale properties and provide necessary input for model credibility assessment. In conclusion, this review aims to provide hands-on guidance for scientists interested in building in silico models as an additional tool to investigate the critical role of the inflammatory phase in bone regeneration.
Collapse
Affiliation(s)
- Laura Lafuente-Gracia
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Edoardo Borgiani
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA in silico Medicine, University of Liège, Liège, Belgium
| | - Gabriele Nasello
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Liesbet Geris
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus: Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.,Biomechanics Research Unit, GIGA in silico Medicine, University of Liège, Liège, Belgium.,Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
43
|
Jin X, Jian Z, Chen X, Ma Y, Ma H, Liu Y, Gong L, Xiang L, Zhu S, Shu X, Qi S, Li H, Wang K. Short Chain Fatty Acids Prevent Glyoxylate-Induced Calcium Oxalate Stones by GPR43-Dependent Immunomodulatory Mechanism. Front Immunol 2021; 12:729382. [PMID: 34675921 PMCID: PMC8523925 DOI: 10.3389/fimmu.2021.729382] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/20/2021] [Indexed: 02/05/2023] Open
Abstract
Calcium oxalate (CaOx) stones are the most common type of kidney stones and are associated with high recurrence, short chain fatty acids (SCFAs), and inflammation. However, it remains uncertain whether SCFAs affect the formation of CaOx stones through immunomodulation. We first performed mass cytometry (CyTOF) and RNA sequencing on kidney immune cells with glyoxylate-induced CaOx crystals (to elucidate the landscape of the associated immune cell population) and explored the role of SCFAs in renal CaOx stone formation through immunomodulation. We identified 29 distinct immune cell subtypes in kidneys with CaOx crystals, where CX3CR1+CD24- macrophages significantly decreased and GR1+ neutrophils significantly increased. In accordance with the CyTOF data, RNA sequencing showed that most genes involved were related to monocytes and neutrophils. SCFAs reduced kidney CaOx crystals by increasing the frequency of CX3CR1+CD24- macrophages and decreasing GR1+ neutrophil infiltration in kidneys with CaOx crystals, which was dependent on the gut microbiota. GPR43 knockdown by transduction with adeno-associated virus inhibited the alleviation of crystal formation and immunomodulatory effects in the kidney, due to SCFAs. Moreover, CX3CR1+CD24- macrophages regulated GR1+ neutrophils via GPR43. Our results demonstrated a unique trilateral relationship among SCFAs, immune cells, and the kidneys during CaOx formation. These findings suggest that future immunotherapies may be used to prevent kidney stones using SCFAs.
Collapse
Affiliation(s)
- Xi Jin
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Zhongyu Jian
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoting Chen
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yucheng Ma
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Hongwen Ma
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Yu Liu
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Lina Gong
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Liyuan Xiang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Shiyu Zhu
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoling Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shiqian Qi
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| | - Kunjie Wang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Nolan E, Malanchi I. Connecting the dots: Neutrophils at the interface of tissue regeneration and cancer. Semin Immunol 2021; 57:101598. [PMID: 35221216 PMCID: PMC9232712 DOI: 10.1016/j.smim.2022.101598] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/19/2022] [Accepted: 02/08/2022] [Indexed: 11/28/2022]
Abstract
Knowledge about neutrophil biology has exponentially grown over the past decades. A high volume of investigations focusing on the characterization of their initially unappreciated multifaceted functions have grown in parallel with the immunity and the cancer fields. This has led to a significant gain in knowledge about their functions not only in tissue defence against pathogens and the collateral damage their overactivation can cause, but also their role in tissue repair and regeneration especially in the context of sterile injuries. On the other hand, the cancer field has also intensively focused its attention on neutrophil engagement in the many steps of the tumorigenic process. This review aims to draw the readers' attention to the similar functions described for neutrophils in tissue repair and in cancer. By bridging the two fields, we provide support for the hypothesis that the underlying program driving cancer-dependent exploitation of neutrophils is rooted in their physiologic tissue protection functions. In this view, cross-fertilization between the two fields will expedite the discovery of therapeutic interventions based on neutrophil targeting or their manipulation.
Collapse
Affiliation(s)
- Emma Nolan
- Tumour Host Interaction Laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, United Kingdom
| | - Ilaria Malanchi
- Tumour Host Interaction Laboratory, The Francis Crick Institute, 1 Midland Road, NW1 1AT London, United Kingdom.
| |
Collapse
|
45
|
Transcriptional Profile of Cytokines, Regulatory Mediators and TLR in Mesenchymal Stromal Cells after Inflammatory Signaling and Cell-Passaging. Int J Mol Sci 2021; 22:ijms22147309. [PMID: 34298927 PMCID: PMC8306573 DOI: 10.3390/ijms22147309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Adult human subcutaneous adipose tissue (AT) harbors a rich population of mesenchymal stromal cells (MSCs) that are of interest for tissue repair. For this purpose, it is of utmost importance to determine the response of AT-MSCs to proliferative and inflammatory signals within the damaged tissue. We have characterized the transcriptional profile of cytokines, regulatory mediators and Toll-like receptors (TLR) relevant to the response of MSCs. AT-MSCs constitutively present a distinct profile for each gene and differentially responded to inflammation and cell-passaging. Inflammation leads to an upregulation of IL-6, IL-8, IL-1β, TNFα and CCL5 cytokine expression. Inflammation and cell-passaging increased the expression of HGF, IDO1, PTGS1, PTGS2 and TGFβ. The expression of the TLR pattern was differentially modulated with TLR 1, 2, 3, 4, 9 and 10 being increased, whereas TLR 5 and 6 downregulated. Functional enrichment analysis demonstrated a complex interplay between cytokines, TLR and regulatory mediators central for tissue repair. This profiling highlights that following a combination of inflammatory and proliferative signals, the sensitivity and responsive capacity of AT-MSCs may be significantly modified. Understanding these transcriptional changes may help the development of novel therapeutic approaches.
Collapse
|
46
|
Wang X, Cai J, Lin B, Ma M, Tao Y, Zhou Y, Bai L, Jiang W, Zhou R. GPR34-mediated sensing of lysophosphatidylserine released by apoptotic neutrophils activates type 3 innate lymphoid cells to mediate tissue repair. Immunity 2021; 54:1123-1136.e8. [PMID: 34107271 DOI: 10.1016/j.immuni.2021.05.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/25/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023]
Abstract
Neutrophils migrate rapidly to damaged tissue and play critical roles in host defense and tissue homeostasis. Here we investigated the mechanisms whereby neutrophils participate in tissue repair. In an intestinal epithelia injury model, neutrophil depletion exacerbated colitis and associated with reduced interleukin (IL)-22 and limited activation of type 3 innate lymphoid cells (ILC3s). Co-culture with neutrophils activated ILC3s in a manner dependent on neutrophil apoptosis. Metabolomic analyses revealed that lysophosphatidylserine (LysoPS) from apoptotic neutrophils directly stimulated ILC3 activation. ILC3-specific deletion of Gpr34, encoding the LysoPS receptor GPR34, or inhibition of downstream PI3K-AKT or ERK suppressed IL-22 production in response to apoptotic neutrophils. Gpr34-/- mice exhibited compromised ILC3 activation and tissue repair during colon injury, and neutrophil depletion abrogated these defects. GPR34 deficiency in ILC3s limited IL-22 production and tissue repair in vivo in settings of colon and skin injury. Thus, GPR34 is an ILC3-expressed damage-sensing receptor that triggers tissue repair upon recognition of dying neutrophils.
Collapse
Affiliation(s)
- Xiaqiong Wang
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China; Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Juan Cai
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Bolong Lin
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Ming Ma
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Ye Tao
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Yubo Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Li Bai
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Wei Jiang
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China.
| | - Rongbin Zhou
- Department of Geriatrics, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China; Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; CAS Centre for Excellence in Cell and Molecular Biology, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
47
|
Kang Y, Zheng C, Ye J, Song F, Wang X, Liu Y, Tian M, Dong J, Lu S. Effects of advanced glycation end products on neutrophil migration and aggregation in diabetic wounds. Aging (Albany NY) 2021; 13:12143-12159. [PMID: 33902006 PMCID: PMC8109105 DOI: 10.18632/aging.202924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/14/2021] [Indexed: 04/29/2023]
Abstract
Increased accumulation of advanced glycation end products (AGEs) in diabetic skin is closely related to delayed wound healing. Studies have shown that the concentration of AGEs is elevated in the skin tissues and not subcutaneous tissues in refractory diabetic wounds, which suggests there may be a causal relationship between the two. In the present study, in vitro experiments revealed that AGEs activated neutrophils, and the migratory and adhesive functions of neutrophils decreased once AGE levels reached a certain threshold. Different levels of AGE expression differentially affected the function of neutrophils. Messenger RNA (mRNA) sequencing analysis combined with real-time polymerase chain reaction (PCR) showed that poliovirus receptor (PVR/CD155) and CTNND1, which play a role in migration- and adhesion-related signaling pathways, were decreased following AGE stimulation. Consequently, neutrophils cannot effectively stimulate the formation of the inflammatory belt needed to remove necrotic tissues and defend against foreign microorganisms within diabetic chronic wounds. In addition, this phenomenon may be related to the differential accumulation of AGEs in different layers of the skin.
Collapse
Affiliation(s)
- Yutian Kang
- Department of Burn, Shanghai Jiao Tong University Affiliated Ruijin Hospital, Shanghai, China
| | - Chongliang Zheng
- Department of Burn, Shanghai Jiao Tong University Affiliated Ruijin Hospital, Shanghai, China
| | - Junna Ye
- Department of Rheumatology and Immunology, Shanghai Jiao Tong University Affiliated Ruijin Hospital, Shanghai, China
| | - Fei Song
- Department of Burn, Shanghai Jiao Tong University Affiliated Ruijin Hospital, Shanghai, China
| | - Xiqiao Wang
- Department of Burn, Shanghai Jiao Tong University Affiliated Ruijin Hospital, Shanghai, China
| | - Yingkai Liu
- Department of Burn, Shanghai Jiao Tong University Affiliated Ruijin Hospital, Shanghai, China
| | - Ming Tian
- Department of Burn, Shanghai Jiao Tong University Affiliated Ruijin Hospital, Shanghai, China
| | - Jiaoyun Dong
- Department of Burn, Shanghai Jiao Tong University Affiliated Ruijin Hospital, Shanghai, China
| | - Shuliang Lu
- Department of Burn, Shanghai Jiao Tong University Affiliated Ruijin Hospital, Shanghai, China
| |
Collapse
|
48
|
Interplay between ESKAPE Pathogens and Immunity in Skin Infections: An Overview of the Major Determinants of Virulence and Antibiotic Resistance. Pathogens 2021; 10:pathogens10020148. [PMID: 33540588 PMCID: PMC7912840 DOI: 10.3390/pathogens10020148] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/16/2022] Open
Abstract
The skin is the largest organ in the human body, acting as a physical and immunological barrier against pathogenic microorganisms. The cutaneous lesions constitute a gateway for microbial contamination that can lead to chronic wounds and other invasive infections. Chronic wounds are considered as serious public health problems due the related social, psychological and economic consequences. The group of bacteria known as ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter sp.) are among the most prevalent bacteria in cutaneous infections. These pathogens have a high level of incidence in hospital environments and several strains present phenotypes of multidrug resistance. In this review, we discuss some important aspects of skin immunology and the involvement of ESKAPE in wound infections. First, we introduce some fundamental aspects of skin physiology and immunology related to cutaneous infections. Following this, the major virulence factors involved in colonization and tissue damage are highlighted, as well as the most frequently detected antimicrobial resistance genes. ESKAPE pathogens express several virulence determinants that overcome the skin's physical and immunological barriers, enabling them to cause severe wound infections. The high ability these bacteria to acquire resistance is alarming, particularly in the hospital settings where immunocompromised individuals are exposed to these pathogens. Knowledge about the virulence and resistance markers of these species is important in order to develop new strategies to detect and treat their associated infections.
Collapse
|
49
|
Abstract
ABSTRACT Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are caused by an exaggerated inflammatory response arising from a wide variety of pulmonary and systemic insults. Lung tissue is composed of a variety of cell populations, including parenchymal and immune cells. Emerging evidence has revealed that multiple cell populations in the lung work in concert to regulate lung inflammation in response to both direct and indirect stimulations. To date, the question of how different types of pulmonary cells communicate with each other and subsequently regulate or modulate inflammatory cascades remains to be fully addressed. In this review, we provide an overview of current advancements in understanding the role of cell-cell interaction in the development of ALI and depict molecular mechanisms by which cell-cell interactions regulate lung inflammation, focusing on inter-cellular activities and signaling pathways that point to possible therapeutic opportunities for ALI/ARDS.
Collapse
Affiliation(s)
- Huiting Zhou
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Erica K. Fan
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
50
|
Liu K, Wang FS, Xu R. Neutrophils in liver diseases: pathogenesis and therapeutic targets. Cell Mol Immunol 2020; 18:38-44. [PMID: 33159158 PMCID: PMC7852892 DOI: 10.1038/s41423-020-00560-0] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Previously, it was assumed that peripheral neutrophils are a homogeneous population that displays antimicrobial functions. However, recent data have revealed that neutrophils are heterogeneous and are additionally involved in tissue damage and immune regulation. The phenotypic and functional plasticity of neutrophils has been identified in patients with cancer, inflammatory disorders, infections, and other diseases. Currently, neutrophils, with their autocrine, paracrine, and immune modulation functions, have been shown to be involved in liver diseases, including viral hepatitis, nonalcoholic steatohepatitis, alcoholic liver disease, liver fibrosis, cirrhosis, liver failure, and liver cancer. Accordingly, this review summarizes the role of neutrophils in liver diseases.
Collapse
Affiliation(s)
- Kai Liu
- Peking University 302 Clinical Medical School, Beijing, China.,Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Fu-Sheng Wang
- Peking University 302 Clinical Medical School, Beijing, China. .,Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.
| | - Ruonan Xu
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.
| |
Collapse
|