1
|
Sapudom J, Alatoom A, Tipay PS, Teo JC. Matrix stiffening from collagen fibril density and alignment modulates YAP-mediated T-cell immune suppression. Biomaterials 2025; 315:122900. [PMID: 39461060 DOI: 10.1016/j.biomaterials.2024.122900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/10/2024] [Revised: 09/16/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
T-cells are essential components of the immune system, adapting their behavior in response to the mechanical environments they encounter within the body. In pathological conditions like cancer, the extracellular matrix (ECM) often becomes stiffer due to increased density and alignment of collagen fibrils, which can have a significant impact on T-cell function. In this study, we explored how these ECM properties-density and fibrillar alignment-affect T-cell behavior using three-dimensional (3D) collagen matrices that mimic these conditions. Our results show that increased matrix stiffness, whether due to higher density or alignment, significantly suppresses T-cell activation, reduces cytokine production, and limits proliferation, largely through enhanced YAP signaling. Individually, matrix alignment appears to lower actin levels in activated T-cells and changes migration behavior in both resting and activated T-cells, an effect not observed in matrices with randomly oriented fibrils. Notably, inhibiting YAP signaling was able to restore T-cell activation and improve immune responses, suggesting a potential strategy to boost the effectiveness of immunotherapy in stiff ECM environments. Overall, this study provides new insights into how ECM characteristics influence T-cell function, offering potential avenues for overcoming ECM-induced immunosuppression in diseases such as cancer.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Aseel Alatoom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates; Department of Biomedical and Mechanical Engineering, Tandon School of Engineering, New York University, USA
| | | | - Jeremy Cm Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates; Department of Biomedical and Mechanical Engineering, Tandon School of Engineering, New York University, USA.
| |
Collapse
|
2
|
Wychowaniec JK, Bektas EI, Vernengo AJ, Muerner M, Airoldi M, Tipay PS, Sapudom J, Teo J, Eglin D, D'Este M. Effect of molecular weight of tyramine-modified hyaluronan on polarization state of THP-1 and peripheral blood mononuclear cells-derived macrophages. BIOMATERIALS ADVANCES 2025; 169:214166. [PMID: 39823943 DOI: 10.1016/j.bioadv.2024.214166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 10/19/2023] [Revised: 10/31/2024] [Accepted: 12/27/2024] [Indexed: 01/20/2025]
Abstract
The immunomodulatory properties of hyaluronan and its derivatives are key to their use in medicine and tissue engineering. In this work we evaluated the capability of soluble tyramine-modified hyaluronan (THA) synthesized from hyaluronan of two molecular weights (low Mw = 280 kDa and high Mw = 1640 kDa) for polarization of THP-1 and peripheral blood mononuclear cells (PBMCs)-derived macrophages (MΦs). We demonstrate the polarization effects of the supplemented THA by flow cytometry and bead-based multiplex immunoassay for the THP-1 derived MΦs and by semi-automated image analysis from confocal microscopy, immunofluorescent staining utilizing CD68 and CD206 surface markers, RT-qPCR gene expression analysis, as well as using the enzyme-linked immunosorbent assay (ELISA) for PBMCs-derived MΦs. Our data indicate that supplementation with LMW THA drives changes in THP-1 derived MΦs towards a pro-inflammatory M1-like phenotype, whereas supplementation with the HMW THA leads to a more mixed profile with some features of both M1 and M2 phenotypes, suggesting either a heterogeneous population or a transitional state. For cells directly sourced from human patients, PMBCs-derived MΦs, results exhibit a higher degree of variability, pointing out a differential regulation of factors including IL-10 and CD206 between the two cell sources. While human primary cells add to the clinical relevance, donor diversity introduces wider variability in the dataset, preventing drawing strong conclusions. Nevertheless, the MΦs profiles observed in THP-1 derived cells for treatments with LMW and HMW THA are generally consistent with what might be expected for the treatment with non-modified hyaluronans of respective molecular weights, confirming the known association holds true for the chemically tyramine-modified hyaluronan. We stipulate that these responses will provide basis for more accurate in vivo representation and translational immunomodulatory guidance for the use of THA-based biomaterials to a wider biomaterials and tissue engineering communities.
Collapse
Affiliation(s)
| | - Ezgi Irem Bektas
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| | - Andrea J Vernengo
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| | - Marcia Muerner
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland; ETH Zürich, Rämistrasse 101, Zürich 8092, Switzerland
| | - Marielle Airoldi
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| | | | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - David Eglin
- Mines Saint-Étienne, Univ Jean Monnet, INSERM, U1059 Sainbiose, Saint-Étienne, France
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| |
Collapse
|
3
|
Jiang J, Tian Y, Wu X, Zeng M, Wu C, Wei D, Luo H, Sun J, Ding J, Fan H. Temperature and light dual-responsive hydrogels for anti-inflammation and wound repair monitoring. J Mater Chem B 2025; 13:2855-2870. [PMID: 39882768 DOI: 10.1039/d4tb02555e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/31/2025]
Abstract
Wound healing is a complex and dynamic biological process that requires meticulous management to ensure optimal outcomes. Traditional wound dressings, such as gauze and bandages, although commonly used, often fall short in their frequent need for replacement, lack of real-time monitoring and absence of anti-inflammatory and antibacterial properties, which can lead to increased risk of infection and delayed healing. Here, we address these limitations by introducing an innovative hydrogel dressing, named PHDNN6, to combine wireless Bluetooth temperature monitoring and light-triggered nitric oxide (NO) release to enhance wound healing and management. The PHDNN6 hydrogel is based on a poly(N-isopropylacrylamide) (PNIPAM) matrix, integrated with methacrylated and dopamine-grafted hyaluronic acid (HA-MA-DA), which allows the dressing to be highly responsive to changes in wound temperature, enabling continuous and real-time monitoring of the wound microenvironment wirelessly. Besides, PHDNN6 is embedded with photothermal polydopamine nanoparticles (PDA NPs) that are loaded with a NO donor, N,N'-di-sec-butyl-N,N'-dinitroso-1,4-phenylenediamine (BNN6). When exposed to near-infrared (NIR) laser irradiation, these PDA@BNN6 nanoparticles release NO to provide potent antibacterial and anti-inflammatory effects. The integration of continuous wireless temperature monitoring with NO release within a single hydrogel dressing represents a significant advancement in clinical wound care. This dual-functional platform not only provides real-time diagnostic capabilities but also offers therapeutic interventions to manage wound infections and promote tissue regeneration. Our research highlights the potential of PHDNN6 to revolutionize wound management by offering a comprehensive solution that addresses both the diagnostic and therapeutic needs in wound healing.
Collapse
Affiliation(s)
- Ji Jiang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Yuan Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Xiaoyang Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Mingze Zeng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Chengheng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610065, P. R. China
| | - Dan Wei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Hongrong Luo
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Jing Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Jie Ding
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610065, P. R. China.
| |
Collapse
|
4
|
Wiesli MG, Huber MW, Weisse B, Zboray R, Kiderlen S, González‐Vázquez A, Maniura‐Weber K, Rottmar M, Lackington WA. Immunomodulation Using BMP-7 and IL-10 to Enhance the Mineralization Capacity of Bone Progenitor Cells in a Fracture Hematoma-Like Environment. Adv Healthc Mater 2025; 14:e2400077. [PMID: 38599586 PMCID: PMC11834375 DOI: 10.1002/adhm.202400077] [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] [Academic Contribution Register] [Received: 01/08/2024] [Revised: 03/19/2024] [Indexed: 04/12/2024]
Abstract
Following biomaterial implantation, a failure to resolve inflammation during the formation of a fracture hematoma can significantly limit the biomaterial's ability to facilitate bone regeneration. This study aims to combine the immunomodulatory and osteogenic effects of BMP-7 and IL-10 with the regenerative capacity of collagen-hydroxyapatite (CHA) scaffolds to enhance in vitro mineralization in a hematoma-like environment. Incubation of CHA scaffolds with human whole blood leads to rapid adsorption of fibrinogen, significant stiffening of the scaffold, and the formation of a hematoma-like environment characterized by a limited capacity to support the infiltration of human bone progenitor cells, a significant upregulation of inflammatory cytokines and acute phase proteins, and significantly reduced osteoconductivity. CHA scaffolds functionalized with BMP-7 and IL-10 significantly downregulate the production of key inflammatory cytokines, including IL-6, IL-8, and leptin, creating a more permissive environment for mineralization, ultimately enhancing the biomaterial's osteoconductivity. In conclusion, targeting the onset of inflammation in the early phase of bone healing using BMP-7 and IL-10 functionalized CHA scaffolds is a promising approach to effectively downregulate inflammatory processes, while fostering a more permissive environment for bone regeneration.
Collapse
Affiliation(s)
- Matthias Guido Wiesli
- Laboratory for BiointerfacesEmpa – Swiss Federal Laboratories for Materials Science and TechnologySt. Gallen9014Switzerland
| | - Matthias Werner Huber
- Laboratory for BiointerfacesEmpa – Swiss Federal Laboratories for Materials Science and TechnologySt. Gallen9014Switzerland
| | - Bernhard Weisse
- Laboratory for Mechanical Systems EngineeringEmpaDübendorf8600Switzerland
| | - Robert Zboray
- Center of X‐ray AnalyticsEmpaDübendorf8600Switzerland
| | | | - Arlyng González‐Vázquez
- Tissue Engineering Research GroupRoyal College of Surgeons in IrelandUniversity of Medicine and Health SciencesDublin 2Ireland
| | - Katharina Maniura‐Weber
- Laboratory for BiointerfacesEmpa – Swiss Federal Laboratories for Materials Science and TechnologySt. Gallen9014Switzerland
| | - Markus Rottmar
- Laboratory for BiointerfacesEmpa – Swiss Federal Laboratories for Materials Science and TechnologySt. Gallen9014Switzerland
| | - William Arthur Lackington
- Laboratory for BiointerfacesEmpa – Swiss Federal Laboratories for Materials Science and TechnologySt. Gallen9014Switzerland
| |
Collapse
|
5
|
Le G, Wen R, Fang H, Huang Z, Wang Y, Luo H. Exosomal miR-122 derived from M2 macrophages induces osteogenic differentiation of bone marrow mesenchymal stem cells in the treatment of alcoholic osteonecrosis of the femoral head. J Orthop Surg Res 2025; 20:107. [PMID: 39881350 PMCID: PMC11776149 DOI: 10.1186/s13018-025-05515-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/02/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Alcoholic osteonecrosis of the femoral head (AIONFH) is caused by long-term heavy drinking, which leads to abnormal alcohol and lipid metabolism, resulting in femoral head tissue damage, and then pathological necrosis of femoral head tissue. If not treated in time in clinical practice, it will seriously affect the quality of life of patients and even require hip replacement to treat alcoholic femoral head necrosis. This study will confirm whether M2 macrophage exosome (M2-Exo) miR-122 mediates alcohol-induced BMSCs osteogenic differentiation, ultimately leading to the inhibition of femoral head necrosis. M2 macrophages were identified by flow cytometry, and the isolated exosomes were characterized by transmission electron microscopy (TEM) and Nanoparticle Tracking Analysis (NTA). Next, miR-122 was overexpressed by transfecting miR-122 mimic, and the expression of miR-122 in M2 macrophages and their exosomes was evaluated. Subsequently, the effect of exosomal miR-122 on the osteogenic differentiation ability of BMSCs was detected, including cell proliferation, expression of osteogenic-related genes (RUNX2, BMP2, OPN, ALP), and calcium nodule formation. Finally, the therapeutic effect of M2-Exo was analyzed in a rat model of AIONFH, and bone repair and pathological damage were evaluated by Micro-CT, RT-qPCR, HE, Masson staining, and immunohistochemistry (COL I). The results showed that M2 macrophages were successfully polarized, with an average M2-Exo particle size of 156.4 nm and a concentration of 3.2E + 12 particles/mL. The expression of miR-122 in M2 macrophages is significantly higher than that in M0 macrophages, and miR-122 mimic can increase the content of miR-122 in M2-Exo. miR-122 in M2-Exo can promote osteogenic differentiation of rat bone marrow BMSCs, enhance cell viability, and increase the expression of osteogenesis-related genes. After being applied to the AIONFH rat model, the injection of M2-exo and miR-122 mimics significantly improved the repair effect of articular cartilage, alleviated pathological changes, and promoted the regeneration of bone tissue. M2-macrophage-derived exosomal miR-122 induces osteogenic differentiation of bone mesenchymal stem cells in treating AIONFH.
Collapse
Affiliation(s)
- Guoping Le
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China
| | - Riyou Wen
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China
| | - Huaixi Fang
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China
| | - Zhifa Huang
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China
| | - Yong Wang
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China
| | - Hanwen Luo
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China.
| |
Collapse
|
6
|
Bahr FS, Müller FE, Kasten M, Benen N, Sieve I, Scherr M, Falk CS, Hilfiker-Kleiner D, Ricke-Hoch M, Ponimaskin E. Serotonin receptor 5-HT7 modulates inflammatory-associated functions of macrophages. Cell Mol Life Sci 2025; 82:51. [PMID: 39833622 PMCID: PMC11747067 DOI: 10.1007/s00018-024-05570-z] [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] [Academic Contribution Register] [Received: 06/14/2024] [Revised: 12/16/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025]
Abstract
The hormone and neurotransmitter serotonin regulates numerous physiological functions within the central nervous system and in the periphery upon binding to specific receptors. In the periphery, the serotonin receptor 7 (5-HT7R) is expressed on different immune cells including monocytes and macrophages. To investigate the impact of 5-HT7R-mediated signaling on macrophage properties, we used human THP-1 cells and differentiated them into pro-inflammatory M1- and anti-inflammatory M2-like macrophages. Pharmacological 5-HT7R activation with the specific agonist LP-211 especially modulates morphology of M1-like macrophages by increasing the number of rounded cells. Furthermore, 5-HT7R stimulation results in significantly reduced phagocytic and migratory ability of M1-like macrophages. Noteworthy, LP-211 treatment leads to changes in secretory properties of all macrophage types with the highest effects obtained for M0- and M2c-like macrophages. Finally, the importance of 5-HT7R for regulation of phagocytosis was confirmed in human primary CD14+ cells. These results indicate that 5-HT7R activation selectively impairs basic functions of macrophages and might thus be a new access point for the modulation of macrophage responses in the future treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Frauke S Bahr
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | | | - Martina Kasten
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Nils Benen
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Irina Sieve
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Centre for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| | - Michaela Scherr
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Christine S Falk
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, DZIF, TTU-IICH, Hannover-Braunschweig Site, Hannover, Germany
| | - Denise Hilfiker-Kleiner
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Medical Faculty of the Philipps-University Marburg, Department of Cardiovascular Complications of Oncologic Therapies, Marburg, Germany
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
7
|
Isali I, McClellan P, Wong TR, Hijaz S, Fletcher DR, Liu G, Bonfield TL, Anderson JM, Hijaz A, Akkus O. Differential effects of macrophage subtype-specific cytokines on fibroblast proliferation and endothelial cell function in co-culture system. J Biomed Mater Res A 2025; 113:e37799. [PMID: 39295242 DOI: 10.1002/jbm.a.37799] [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] [Academic Contribution Register] [Received: 06/21/2024] [Revised: 08/25/2024] [Accepted: 09/06/2024] [Indexed: 09/21/2024]
Abstract
Macrophages are involved in several critical activities associated with tissue repair and regeneration. Current approaches in regenerative medicine are focusing on leveraging the innate immune response to accelerate tissue regeneration and improve long-term healing outcomes. Of particular interest in this regard are the currently known, four main M2 macrophage subtypes: M2interleukin (IL)-4,IL-13, M2IC, M2IL-10, M2non-selective adenosine receptor agonists (NECA) (M2IL-4,IL-13 → M2NECA). In this study, rat bone marrow-derived macrophages (M0) were polarized to each of the four subtypes M2IL-4,IL-13 → M2NECA and cultured for 72 h in vitro. Luminex assay results highlighted increased production of tissue inhibitor of metalloproteinases-1 (TIMP-1) for M2IL-4,IL-13, higher amounts of transforming growth factor-beta 1 (TGF-β1) for M2IL-10, and elevated vascular endothelial growth factor A (VEGF-A) from M2NECA. Co-culture experiments performed with M2IL-10 macrophages and L929 fibroblasts highlighted the increased production of soluble collagen within the media as well as higher amounts of collagen in the extracellular matrix. Human umbilical vein endothelial cells (HUVECs) were co-cultured with M2NECA macrophages, which demonstrated an increase in intercellular adhesion molecule (ICAM) and platelet endothelial cell adhesion molecule (PECAM), as well as increased formation of endothelial tubes. The findings of this study emphasize a critical demand for further characterization and analyses of distinct M2 subtypes and careful selection of specific macrophage populations for regeneration of specific tissue types. The current, broad classification of "M2" may be sufficient in many general tissue engineering applications, but, as conditions are constantly in flux within the microenvironment in vivo, a higher degree of specificity and control over the initial M2 subtype could result in more consistent long-term outcomes where macrophages are utilized as part of an overall regenerative strategy.
Collapse
Affiliation(s)
- Ilaha Isali
- Department of Urology, Weill Cornell Medicine, New York, New York, USA
| | - Phillip McClellan
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Thomas R Wong
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sara Hijaz
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, USA
| | - David R Fletcher
- Case Western Reserve University, Department of Genetics and Genome Sciences, Cleveland, Ohio, USA
| | - Guiming Liu
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, Ohio, USA
| | - Tracey L Bonfield
- Case Western Reserve University, Department of Genetics and Genome Sciences, Cleveland, Ohio, USA
| | - James M Anderson
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Adonis Hijaz
- Department of Urology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ozan Akkus
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Orthopedics, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Chaparro V, Leroux LP, Lebourg A, Chagneau S, Graber TE, Alain T, Jaramillo M. Leukemia inhibitory factor drives transcriptional programs that promote lipid accumulation and M2 polarization in macrophages. J Leukoc Biol 2024; 117:qiae178. [PMID: 39178293 DOI: 10.1093/jleuko/qiae178] [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] [Academic Contribution Register] [Received: 07/12/2024] [Revised: 07/24/2024] [Accepted: 08/22/2024] [Indexed: 08/25/2024] Open
Abstract
Leukemia inhibitory factor, a member of the interleukin-6 cytokine family, plays a central role in homeostasis and disease. Interestingly, some of the pleiotropic effects of leukemia inhibitory factor have been attributed to the modulation of macrophage functions although the molecular underpinnings have not been explored at a genome-wide scale. Herein, we investigated leukemia inhibitory factor-driven transcriptional changes in murine bone marrow-derived macrophages by RNA sequencing. In silico analyses revealed a selective and time-dependent remodeling of macrophage gene expression programs associated with lipid metabolism and cell activation. Accordingly, a subset of leukemia inhibitory factor-upregulated transcripts related to cholesterol metabolism and lipid internalization was validated by real-time quantitative polymerase chain reaction. This was accompanied by a leukemia inhibitory factor-enhanced capacity for lipid accumulation in macrophages upon incubation with oxidized low-density lipoprotein. Mechanistically, leukemia inhibitory factor triggered the phosphorylation (Y705 and S727) and nuclear translocation of the transcription factor STAT3 in bone marrow-derived macrophages. Consistent with this, ingenuity pathway analysis identified STAT3 as an upstream regulator of a subset of transcripts, including Il4ra, in leukemia inhibitory factor-treated macrophages. Notably, leukemia inhibitory factor priming enhanced bone marrow-derived macrophage responses to interleukin-4-mediated M2 polarization (i.e. increased arginase activity and accumulation of transcripts encoding for M2 markers). Conversely, leukemia inhibitory factor stimulation had no significant effect in bone marrow-derived macrophage responses to M1-polarizing stimuli (interferon-γ and lipopolysaccharide). Thus, our study provides insight into the transcriptional landscape of leukemia inhibitory factor-treated macrophages, shedding light on its role in lipid metabolism and M2 polarization responses. A better understanding of the regulatory mechanisms governing leukemia inhibitory factor-driven changes might help informing novel therapeutic approaches aiming to reprogram macrophage phenotypes in diseased states (e.g. cancer, atherosclerosis, and infection).
Collapse
Affiliation(s)
- Visnu Chaparro
- Institut National de la Recherche Scientifique (INRS) - Centre Armand-Frappier Santé Biotechnologie (CAFSB), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Louis-Philippe Leroux
- Institut National de la Recherche Scientifique (INRS) - Centre Armand-Frappier Santé Biotechnologie (CAFSB), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Aurore Lebourg
- Institut National de la Recherche Scientifique (INRS) - Centre Armand-Frappier Santé Biotechnologie (CAFSB), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Sophie Chagneau
- Institut National de la Recherche Scientifique (INRS) - Centre Armand-Frappier Santé Biotechnologie (CAFSB), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Tyson E Graber
- Children's Hospital of Eastern Ontario Research Institute, 401 Smith Rd. Ottawa, ON K1H 8L1, Canada
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, 401 Smith Rd. Ottawa, ON K1H 8L1, Canada
- Department of Biochemistry, Microbiology and Immunology, 75 Laurier Ave E. University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Maritza Jaramillo
- Institut National de la Recherche Scientifique (INRS) - Centre Armand-Frappier Santé Biotechnologie (CAFSB), 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| |
Collapse
|
9
|
Hosseininejad-Chafi M, Eftekhari Z, Oghalaie A, Behdani M, Sotoudeh N, Kazemi-Lomedasht F. Nanobodies as innovative immune checkpoint modulators: advancing cancer immunotherapy. Med Oncol 2024; 42:36. [PMID: 39719469 DOI: 10.1007/s12032-024-02588-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/22/2024] [Accepted: 12/14/2024] [Indexed: 12/26/2024]
Abstract
The immune system relies on a delicate balance between attacking harmful pathogens and preserving the body's own tissues, a balance maintained by immune checkpoints. These checkpoints play a critical role in preventing autoimmune diseases by restraining excessive immune responses while allowing the immune system to recognize and destroy abnormal cells, such as tumors. In recent years, immune checkpoint inhibitors (ICIs) have become central to cancer therapy, enabling the immune system to target and eliminate cancer cells that evade detection. Traditional antibodies, such as IgGs, have been widely used in immune therapies but are limited by their size and complexity. Nanobodies (Nbs), derived from camelid heavy-chain-only antibodies, offer a promising alternative. These small, stable antibody fragments retain the antigen-binding specificity of traditional antibodies but have enhanced solubility and the ability to target otherwise inaccessible epitopes. This review explores the use of Nbs as ICIs, emphasizing their potential in cancer immunotherapy and other immune-related treatments. Their unique structural properties and small size make Nbs highly effective tools for modulating immune responses, representing a novel approach in the evolving landscape of checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Mohammad Hosseininejad-Chafi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Nazli Sotoudeh
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran.
| |
Collapse
|
10
|
Yang X, Li J, Xu C, Zhang G, Che X, Yang J. Potential mechanisms of rheumatoid arthritis therapy: Focus on macrophage polarization. Int Immunopharmacol 2024; 142:113058. [PMID: 39236455 DOI: 10.1016/j.intimp.2024.113058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/18/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that affects multiple organs and systems in the human body, often leading to disability. Its pathogenesis is complex, and the long-term use of traditional anti-rheumatic drugs frequently results in severe toxic side effects. Therefore, the search for a safer and more effective antirheumatic drug is extremely important for the treatment of RA. As important immune cells in the body, macrophages are polarized. Under pathological conditions, macrophages undergo proliferation and are recruited to diseased tissues upon stimulation. In the local microenvironment, they polarize into different types of macrophages in response to specific factors and perform unique functions and roles. Previous studies have shown that there is a link between macrophage polarization and RA, indicating that certain active ingredients can ameliorate RA symptoms through macrophage polarization. Notably, Traditional Chinese medicine (TCM) monomer component and compounds demonstrate a particular advantage in this process. Building upon this insight, we reviewed and analyzed recent studies to offer valuable and meaningful insights and directions for the development and application of anti-rheumatic drugs.
Collapse
Affiliation(s)
- Xinyu Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinling Li
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengchao Xu
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinzhen Che
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiguo Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
11
|
Cheng Y, Liu L, Ye Y, He Y, Hu W, Ke H, Guo ZY, Shao G. Roles of macrophages in lupus nephritis. Front Pharmacol 2024; 15:1477708. [PMID: 39611168 PMCID: PMC11602334 DOI: 10.3389/fphar.2024.1477708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/08/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
LN is a serious complication of systemic lupus erythematosus (SLE), affecting up to 60% of patients with SLE and may lead to end-stage renal disease (ESRD). Macrophages play multifaceted roles in the pathogenesis of LN, including clearance of immune complexes, antigen presentation, regulation of inflammation, and tissue repair. Macrophages are abundant in the glomeruli and tubulointerstitium of LN patients and are positively correlated with serum creatinine levels and the severity of renal pathology. It has been shown that the infiltration of macrophages is closely associated with several clinical indicators, such as serum creatinine and complement C3 levels, anti-dsDNA antibody titers, Austin score, interstitial fibrosis and renal tubular atrophy. Moreover, cytokines expressed by macrophages were upregulated at LN onset and downregulated after remission, suggesting that macrophages may serve as markers of LN pathogenesis and remission. Therapies targeting macrophages have been shown to alleviate LN. There are two main types of macrophages in the kidney: kidney-resident macrophages (KRMs) and monocyte-derived macrophages (MDMs). KRMs and MDMs play different pathological roles in LN, with KRMs promoting leukocyte recruitment at sites of inflammation by expressing monocyte chemokines, while MDMs may exacerbate autoimmune responses by presenting immune complex antigens. Macrophages exhibit high plasticity and can differentiate into various phenotypes in response to distinct environmental stimuli. M1 (proinflammatory) macrophages are linked to the progression of active SLE, whereas the M2 (anti-inflammatory) phenotype is observed during the remission phase of LN. The polarization of macrophages in LN can be manipulated through multiple pathways, such as the modulation of signaling cascades including TLR 2/1, S1P, ERS, metabolic reprogramming, and HMGB1. This paper provides a comprehensive overview of the role of macrophages in the progression of lupus nephritis (LN), and elucidates how these cells and their secretory products function as indicators and therapeutic targets for the disease in the context of diagnosis and treatment of LN.
Collapse
Affiliation(s)
- Yaqian Cheng
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, China
| | - Lulu Liu
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yufei Ye
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yingxue He
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, China
| | - Wenwen Hu
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, China
| | - Haiyan Ke
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, China
| | - Zhi-Yong Guo
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guojian Shao
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
12
|
Phuphanitcharoenkun S, Louis F, Sowa Y, Uchida K, Katsuyama M, Waditee-Sirisattha R, Kageyama H, Matsusaki M, Palaga T. Characterization of macrophages associated with human skin models exposed to UV radiation. Commun Biol 2024; 7:1284. [PMID: 39379484 PMCID: PMC11461876 DOI: 10.1038/s42003-024-06975-z] [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] [Academic Contribution Register] [Received: 03/06/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
Skin macrophages play important roles in the response to external stimuli. Human skin equivalents (HSEs) incorporating the human monocytic cell line THP-1 were fabricated to generate immunocompetent human skin models. These HSEs were used to investigate the influence of the skin microenvironment and ultraviolet A (UVA) on macrophages. Transcriptomic analysis revealed that THP-1 cells in HSEs were enriched in extracellular matrix interaction hallmark but downregulated in DNA replication hallmark. Upon UVA exposure, immunocompetent HSEs presented epidermal distortion and increased DNA double-strand breaks (DSBs). The genes associated with oxidative stress and the inflammatory response were significantly upregulated in THP-1 cells. When the photoprotective agent mycosporine-2-glycine from cyanobacteria was applied to HSEs, the incidence of UVA-induced DSBs was significantly lower, and inflammatory and UV response hallmarks were downregulated in THP-1 cells. Taken together, these results suggest that immunocompetent HSEs can be used to investigate the responses of skin-resident macrophages to external stimuli.
Collapse
Affiliation(s)
- Suphanun Phuphanitcharoenkun
- Graduate Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Materials and Bio-interfaces, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, 565-0871, Japan
| | - Yoshihiro Sowa
- Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
- Department of Plastic Surgery, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Kentaro Uchida
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation Kadoma, Osaka, 571-8686, Japan
| | - Misa Katsuyama
- Materials Solution Department, Product Analysis Center, Panasonic Holdings Corporation Kadoma, Osaka, 571-8686, Japan
| | | | - Hakuto Kageyama
- Graduate School of Environmental and Human Sciences, Meijo University, Nagoya, Aichi, 468-8502, Japan
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, 565-0871, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, 565-0871, Japan
| | - Tanapat Palaga
- Center of Excellence in Materials and Bio-interfaces, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
13
|
Wang J, Wu Q, Wang X, Liu H, Chen M, Xu L, Zhang Z, Li K, Li W, Zhong J. Targeting Macrophage Phenotypes and Metabolism as Novel Therapeutic Approaches in Atherosclerosis and Related Cardiovascular Diseases. Curr Atheroscler Rep 2024; 26:573-588. [PMID: 39133247 PMCID: PMC11392985 DOI: 10.1007/s11883-024-01229-z] [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] [Academic Contribution Register] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
PURPOSE OF THE REVIEW Macrophage accumulation and activation function as hallmarks of atherosclerosis and have complex and intricate dynamics throughout all components and stages of atherosclerotic plaques. In this review, we focus on the regulatory roles and underlying mechanisms of macrophage phenotypes and metabolism in atherosclerosis. We highlight the diverse range of macrophage phenotypes present in atherosclerosis and their potential roles in progression and regression of atherosclerotic plaque. Furthermore, we discuss the challenges and opportunities in developing therapeutic strategies for preventing and treating atherosclerotic cardiovascular disease. RECENT FINDINGS Dysregulation of macrophage polarization between the proinflammatory M1 and anti-inflammatory M2 phenotypealters the immuno-inflammatory response during atherosclerosis progression, leading to plaque initiation, growth, and ultimately rupture. Altered metabolism of macrophage is a key feature for their function and the subsequent progression of atherosclerotic cardiovascular disease. The immunometabolism of macrophage has been implicated to macrophage activation and metabolic rewiring of macrophages within atherosclerotic lesions, thereby shifting altered macrophage immune-effector and tissue-reparative function. Targeting macrophage phenotypes and metabolism are potential therapeutic strategies in the prevention and treatment of atherosclerosis and atherosclerotic cardiovascular diseases. Understanding the precise function and metabolism of specific macrophage subsets and their contributions to the composition and growth of atherosclerotic plaques could reveal novel strategies to delay or halt development of atherosclerotic cardiovascular diseases and their associated pathophysiological consequences. Identifying biological stimuli capable of modulating macrophage phenotypes and metabolism may lead to the development of innovative therapeutic approaches for treating patients with atherosclerosis and coronary artery diseases.
Collapse
Affiliation(s)
- Juan Wang
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Qiang Wu
- Senior Department of Cardiology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
- Journal of Geriatric Cardiology Editorial Office, Chinese PLA General Hospital, Beijing, China
| | - Xinyu Wang
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hongbin Liu
- Department of Cardiology, the Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Mulei Chen
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Li Xu
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ze Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Kuibao Li
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Weiming Li
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Jiuchang Zhong
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
14
|
Xue JD, Gao J, Tang AF, Feng C. Shaping the immune landscape: Multidimensional environmental stimuli refine macrophage polarization and foster revolutionary approaches in tissue regeneration. Heliyon 2024; 10:e37192. [PMID: 39296009 PMCID: PMC11408064 DOI: 10.1016/j.heliyon.2024.e37192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/10/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
In immunology, the role of macrophages extends far beyond their traditional classification as mere phagocytes; they emerge as pivotal architects of the immune response, with their function being significantly influenced by multidimensional environmental stimuli. This review investigates the nuanced mechanisms by which diverse external signals ranging from chemical cues to physical stress orchestrate macrophage polarization, a process that is crucial for the modulation of immune responses. By transitioning between pro-inflammatory (M1) and anti-inflammatory (M2) states, macrophages exhibit remarkable plasticity, enabling them to adapt to and influence their surroundings effectively. The exploration of macrophage polarization provides a compelling narrative on how these cells can be manipulated to foster an immune environment conducive to tissue repair and regeneration. Highlighting cutting-edge research, this review presents innovative strategies that leverage the dynamic interplay between macrophages and their environment, proposing novel therapeutic avenues that harness the potential of macrophages in regenerative medicine. Moreover, this review critically evaluates the current challenges and future prospects of translating macrophage-centered strategies from the laboratory to clinical applications.
Collapse
Affiliation(s)
- Jing-Dong Xue
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jing Gao
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ai-Fang Tang
- Department of Geratology, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Chao Feng
- Department of Reproductive Medicine, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China
| |
Collapse
|
15
|
Torabi S, Zarrabi M, Shekari F, Poorkazem H, Lotfinia M, Bencina S, Gramignoli R, Hassan M, Najimi M, Vosough M. Wharton's Jelly mesenchymal stem cell-derived extracellular vesicles induce liver fibrosis-resolving phenotype in alternatively activated macrophages. J Cell Mol Med 2024; 28:e18507. [PMID: 39288445 PMCID: PMC11407755 DOI: 10.1111/jcmm.18507] [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] [Academic Contribution Register] [Received: 12/05/2023] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 09/19/2024] Open
Abstract
The potential of extracellular vesicles (EVs) isolated from mesenchymal stromal cells in guiding macrophages toward anti-inflammatory immunophenotypes, has been reported in several studies. In our study, we provided experimental evidence of a distinctive effect played by Wharton Jelly mesenchymal stromal cell-derived EVs (WJ-EVs) on human macrophages. We particularly analyzed their anti-inflammatory effects on macrophages by evaluating their interactions with stellate cells, and their protective role in liver fibrosis. A three-step gradient method was used to isolate monocytes from umbilical cord blood (UCB). Two subpopulations of WJ-EVs were isolated by high-speed (20,000 g) and differential ultracentrifugation (110,000 g). Further to their characterization, they were designated as EV20K and EV110K and incubated at different concentrations with UCB-derived monocytes for 7 days. Their anti-fibrotic effect was assessed by studying the differentiation and functional levels of generated macrophages and their potential to modulate the survival and activity of LX2 stellate cells. The EV20K triggers the polarization of UCB-derived monocytes towards a peculiar M2-like functional phenotype more effectively than the M-CSF positive control. The EV20K treated macrophages were characterized by a higher expression of scavenger receptors, increased phagocytic capacity and production level of interleukin-10 and transforming growth factor-β. Conditioned medium from those polarized macrophages attenuated the proliferation, contractility and activation of LX2 stellate cells. Our data show that EV20K derived from WJ-MSCs induces activated macrophages to suppress immune responses and potentially play a protective role in the pathogenesis of liver fibrosis by directly inhibiting HSC's activation.
Collapse
Affiliation(s)
- Shukoofeh Torabi
- Department of Regenerative Medicine, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in MedicineMazandaran University of Medical SciencesSariIran
| | - Morteza Zarrabi
- Department of Regenerative Medicine, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Hedie Poorkazem
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Majid Lotfinia
- Physiology Research CenterKashan University of Medical SciencesKashanIran
| | - Stefan Bencina
- Department of Laboratory Medicine, Division of PathologyKarolinska InstitutetStockholmSweden
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of PathologyKarolinska InstitutetStockholmSweden
- UOSD Cell FactoryIRCCS Istituto Giannina GasliniGenoaItaly
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine and Karolinska University HospitalKarolinska InstituteStockholmSweden
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell TherapyInstitute of Experimental and Clinical Research (IREC), UCLouvainBrusselsBelgium
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Experimental Cancer Medicine, Institution for Laboratory Medicine and Karolinska University HospitalKarolinska InstituteStockholmSweden
| |
Collapse
|
16
|
Farrell LA, O’Rourke MB, Padula MP, Souza-Fonseca-Guimaraes F, Caramori G, Wark PAB, Dharmage SC, Hansbro PM. The Current Molecular and Cellular Landscape of Chronic Obstructive Pulmonary Disease (COPD): A Review of Therapies and Efforts towards Personalized Treatment. Proteomes 2024; 12:23. [PMID: 39189263 PMCID: PMC11348234 DOI: 10.3390/proteomes12030023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/28/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) ranks as the third leading cause of global illness and mortality. It is commonly triggered by exposure to respiratory irritants like cigarette smoke or biofuel pollutants. This multifaceted condition manifests through an array of symptoms and lung irregularities, characterized by chronic inflammation and reduced lung function. Present therapies primarily rely on maintenance medications to alleviate symptoms, but fall short in impeding disease advancement. COPD's diverse nature, influenced by various phenotypes, complicates diagnosis, necessitating precise molecular characterization. Omics-driven methodologies, including biomarker identification and therapeutic target exploration, offer a promising avenue for addressing COPD's complexity. This analysis underscores the critical necessity of improving molecular profiling to deepen our comprehension of COPD and identify potential therapeutic targets. Moreover, it advocates for tailoring treatment strategies to individual phenotypes. Through comprehensive exploration-based molecular characterization and the adoption of personalized methodologies, innovative treatments may emerge that are capable of altering the trajectory of COPD, instilling optimism for efficacious disease-modifying interventions.
Collapse
Affiliation(s)
- Luke A. Farrell
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Centre for Inflammation, Ultimo, NSW 2007, Australia;
| | - Matthew B. O’Rourke
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Centre for Inflammation, Ultimo, NSW 2007, Australia;
| | - Matthew P. Padula
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | | | - Gaetano Caramori
- Pulmonology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
| | - Peter A. B. Wark
- School of Translational Medicine, Monash University, Melbourne, VIC 3000, Australia;
| | - Shymali C. Dharmage
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Melbourne, VIC 3000, Australia;
| | - Phillip M. Hansbro
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Centre for Inflammation, Ultimo, NSW 2007, Australia;
| |
Collapse
|
17
|
Marrone L, Romano S, Albanese M, Giordano S, Morello A, Cimmino M, Di Giacomo V, Malasomma C, Romano MF, Corcione N. Tirofiban prevents the effects of SARS-CoV-2 spike protein on macrophage activation and endothelial cell death. Heliyon 2024; 10:e35341. [PMID: 39170431 PMCID: PMC11336627 DOI: 10.1016/j.heliyon.2024.e35341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/22/2023] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
SARS-CoV-2 viral-derived particles have been proposed to have a causal role in tissue inflammation. Macrophage is the culprit cell in the pathogenesis of destructive inflammatory response to the SARS-CoV-2 virus. We investigated whether the spike protein might play a role in perturbing the physiological process of resolution of inflammation. Using an in vitro model of M2 polarized macrophages, we found that recombinant spike protein produced typical M1 morphological features in these alternative differentiated cells. In the presence of spike, M2-macrophages lose their elongated morphology, become rounded and acquire a strong capability to stimulate lymphocyte activation and proliferation. Moreover, in M2 macrophages, spike activated the signal transducer and activator-1 (STAT1) the pivotal mediator of pro-inflammatory macrophages. We observed STAT1 activation also in endothelial cells cultured with recombinant spike, accompanied by Bax upregulation and cell death. Blockade of beta3 integrin with the RGD mimetic tirofiban reverted the spike-induced costimulatory effects on M2 macrophages. Also, tirofiban counteracted STAT1 and Bax activation in endothelial cells cultured with spike and reduced endothelial cell death. In conclusion, we found that some proinflammatory effects of the spike protein can involve the integrin pathway and provide elements supporting use of RGD mimetics against SARS-Cov-2.
Collapse
Affiliation(s)
- Laura Marrone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Michele Albanese
- Cardiovascular Interventions Unit, Pineta Grande Hospital, Castel Volturno, Italy
- Hemodynamics Unit, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| | - Salvatore Giordano
- Division of Cardiology, Department of Medical and Surgical Sciences, “Magna Graecia” University, Catanzaro, Italy
| | - Alberto Morello
- Cardiovascular Interventions Unit, Pineta Grande Hospital, Castel Volturno, Italy
- Hemodynamics Unit, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| | - Michele Cimmino
- Cardiovascular Interventions Unit, Pineta Grande Hospital, Castel Volturno, Italy
- Hemodynamics Unit, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| | - Valeria Di Giacomo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Chiara Malasomma
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Nicola Corcione
- Cardiovascular Interventions Unit, Pineta Grande Hospital, Castel Volturno, Italy
- Hemodynamics Unit, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| |
Collapse
|
18
|
Kong L, Sun P, Pan X, Xiao C, Song B, Song Z. Glycerol monolaurate regulates apoptosis and inflammation by suppressing lipopolysaccharide-induced ROS production and NF-κB activation in avian macrophages. Poult Sci 2024; 103:103870. [PMID: 38851181 PMCID: PMC11208948 DOI: 10.1016/j.psj.2024.103870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/03/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 06/10/2024] Open
Abstract
Macrophages play a crucial role in both innate and adaptive immunity. However, their abnormal activation can lead to undesirable inflammatory reactions. This study aimed to investigate the effects of glycerol monolaurate (GML), a natural monoester known for its anti-inflammatory and immunoregulatory properties, on avian macrophages using the HD11 cell line. The results indicated that a concentration of 10 μg/mL of GML enhanced the phagocytic activity of HD11 cells (P < 0.05) without affecting cell viability (P > 0.05). GML decreased the expression of M1 macrophage polarization markers, such as CD86 and TNF-α genes (P < 0.05), while increasing the expression of M2 macrophage polarization markers, such as TGF-β1 and IL-10 genes (P < 0.05). GML suppressed ROS production, apoptosis, and the expression of proinflammatory genes (IL-1β and IL-6) induced by LPS (P < 0.05). GML also promoted the expression of TGF-β1 and IL-10 (P < 0.05), both in the presence and absence of LPS exposure. Moreover, GML suppressed the gene expression of TLR4 and NF-κB p65 induced by LPS (P < 0.05), as well as the phosphorylation of NF-κB p65 (P < 0.05). In conclusion, GML exhibited regulatory effects on the polarized state of avian macrophages and demonstrated significant anti-apoptotic and anti-inflammatory properties by suppressing intracellular ROS and the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Linglian Kong
- Office of Assessment, Jining Polytechnic, Jining, Shandong 272037, China; Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Peng Sun
- Laboratory of Chemistry of Natural Molecules, Gembloux Agro-Bio Tech, University of Liège, Gembloux 5030, Belgium
| | - Xue Pan
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Chuanpi Xiao
- Laboratory of Chemistry of Natural Molecules, Gembloux Agro-Bio Tech, University of Liège, Gembloux 5030, Belgium
| | - Bochen Song
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Zhigang Song
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China.
| |
Collapse
|
19
|
Streutker EM, Devamoglu U, Vonk MC, Verdurmen WPR, Le Gac S. Fibrosis-on-Chip: A Guide to Recapitulate the Essential Features of Fibrotic Disease. Adv Healthc Mater 2024; 13:e2303991. [PMID: 38536053 DOI: 10.1002/adhm.202303991] [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] [Academic Contribution Register] [Received: 11/14/2023] [Revised: 03/15/2024] [Indexed: 05/05/2024]
Abstract
Fibrosis, which is primarily marked by excessive extracellular matrix (ECM) deposition, is a pathophysiological process associated with many disorders, which ultimately leads to organ dysfunction and poor patient outcomes. Despite the high prevalence of fibrosis, currently there exist few therapeutic options, and importantly, there is a paucity of in vitro models to accurately study fibrosis. This review discusses the multifaceted nature of fibrosis from the viewpoint of developing organ-on-chip (OoC) disease models, focusing on five key features: the ECM component, inflammation, mechanical cues, hypoxia, and vascularization. The potential of OoC technology is explored for better modeling these features in the context of studying fibrotic diseases and the interplay between various key features is emphasized. This paper reviews how organ-specific fibrotic diseases are modeled in OoC platforms, which elements are included in these existing models, and the avenues for novel research directions are highlighted. Finally, this review concludes with a perspective on how to address the current gap with respect to the inclusion of multiple features to yield more sophisticated and relevant models of fibrotic diseases in an OoC format.
Collapse
Affiliation(s)
- Emma M Streutker
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Utku Devamoglu
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Wouter P R Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| |
Collapse
|
20
|
Hwang S, Cho JM, Yoon YJ, Seo S, Hong Y, Lim JY. Retroductal dexamethasone administration promotes the recovery from obstructive and inflammatory salivary gland dysfunction. Front Immunol 2024; 15:1418703. [PMID: 39044831 PMCID: PMC11263033 DOI: 10.3389/fimmu.2024.1418703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/17/2024] [Accepted: 06/12/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction Salivary gland dysfunction, often resulting from salivary gland obstruction-induced inflammation, is a prevalent condition. Corticosteroid, known for its anti-inflammatory and immunomodulatory properties, is commonly prescribed in clinics. This study investigates the therapeutic implications and potential side effects of dexamethasone on obstructive sialadenitis recovery using duct ligation mice and salivary gland organoid models. Methods Functional and pathological changes were assessed after administering dexamethasone to the duct following deligation 2 weeks after maintaining ligation of the mouse submandibular duct. Additionally, lipopolysaccharide- and tumor necrosis factor-induced salivary gland organoid inflammation models were established to investigate the effects and underlying mechanisms of action of dexamethasone. Results Dexamethasone administration facilitated SG function restoration, by increasing salivary gland weight and saliva volume while reducing saliva lag time. Histological evaluation revealed, reduced acinar cell atrophy and fibrosis with dexamethasone treatment. Additionally, dexamethasone suppressed pro-inflammatory cytokines IL-1β and TNF expression. In a model of inflammation in salivary gland organoids induced by inflammatory substances, dexamethasone restored acinar markers such as AQP5 gene expression levels, while inhibiting pro-inflammatory cytokines TNF and IL6, as well as chemokines CCL2, CXCL5, and CXCL12 induction. Macrophages cultured in inflammatory substance-treated media from salivary gland organoid cultures exhibited pro-inflammatory polarization. However, treatment with dexamethasone shifted them towards an anti-inflammatory phenotype by reducing M1 markers (Tnf, Il6, Il1b, and Cd86) and elevating M2 markers (Ym1, Il10, Cd163, and Klf4). However, high-dose or prolonged dexamethasone treatment induced acino-ductal metaplasia and had side effects in both in vivo and in vitro models. Conclusions Our findings suggest the effectiveness of corticosteroids in treating obstructive sialadenitis-induced salivary gland dysfunction by regulating pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Seungyeon Hwang
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Min Cho
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yeo-Jun Yoon
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sunyoung Seo
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yongpyo Hong
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Yol Lim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
21
|
González-Callejo P, García-Astrain C, Herrero-Ruiz A, Henriksen-Lacey M, Seras-Franzoso J, Abasolo I, Liz-Marzán LM. 3D Bioprinted Tumor-Stroma Models of Triple-Negative Breast Cancer Stem Cells for Preclinical Targeted Therapy Evaluation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:27151-27163. [PMID: 38764168 PMCID: PMC11145592 DOI: 10.1021/acsami.4c04135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024]
Abstract
Breast cancer stem cells (CSCs) play a pivotal role in therapy resistance and tumor relapse, emphasizing the need for reliable in vitro models that recapitulate the complexity of the CSC tumor microenvironment to accelerate drug discovery. We present a bioprinted breast CSC tumor-stroma model incorporating triple-negative breast CSCs (TNB-CSCs) and stromal cells (human breast fibroblasts), within a breast-derived decellularized extracellular matrix bioink. Comparison of molecular signatures in this model with different clinical subtypes of bioprinted tumor-stroma models unveils a unique molecular profile for artificial CSC tumor models. We additionally demonstrate that the model can recapitulate the invasive potential of TNB-CSC. Surface-enhanced Raman scattering imaging allowed us to monitor the invasive potential of tumor cells in deep z-axis planes, thereby overcoming the depth-imaging limitations of confocal fluorescence microscopy. As a proof-of-concept application, we conducted high-throughput drug testing analysis to assess the efficacy of CSC-targeted therapy in combination with conventional chemotherapeutic compounds. The results highlight the usefulness of tumor-stroma models as a promising drug-screening platform, providing insights into therapeutic efficacy against CSC populations resistant to conventional therapies.
Collapse
Affiliation(s)
| | - Clara García-Astrain
- CIC
biomaGUNE, Basque Research
and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Barcelona 08035, Spain
| | - Ada Herrero-Ruiz
- CIC
biomaGUNE, Basque Research
and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Barcelona 08035, Spain
| | - Malou Henriksen-Lacey
- CIC
biomaGUNE, Basque Research
and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Barcelona 08035, Spain
| | - Joaquín Seras-Franzoso
- Centro
de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Barcelona 08035, Spain
- Clinical
Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d’Hebron
Research Institute (VHIR), Vall d’Hebron
University Hospital, Barcelona 08035, Spain
- Department
of Genetics and Microbiology, Universitat
Autònoma de Barcelona (UAB), Bellaterra 08193, Spain
| | - Ibane Abasolo
- Centro
de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Barcelona 08035, Spain
- Clinical
Biochemistry, Drug Delivery and Therapy Group (CB-DDT), Vall d’Hebron
Research Institute (VHIR), Vall d’Hebron
University Hospital, Barcelona 08035, Spain
- Clinical
Biochemistry Service, Vall d’Hebron
University Hospital, Barcelona 08035, Spain
| | - Luis M. Liz-Marzán
- CIC
biomaGUNE, Basque Research
and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Barcelona 08035, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao 48009, Spain
| |
Collapse
|
22
|
Marques E, Kramer R, Ryan DG. Multifaceted mitochondria in innate immunity. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:6. [PMID: 38812744 PMCID: PMC11129950 DOI: 10.1038/s44324-024-00008-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Academic Contribution Register] [Received: 03/01/2024] [Accepted: 04/14/2024] [Indexed: 05/31/2024]
Abstract
The ability of mitochondria to transform the energy we obtain from food into cell phosphorylation potential has long been appreciated. However, recent decades have seen an evolution in our understanding of mitochondria, highlighting their significance as key signal-transducing organelles with essential roles in immunity that extend beyond their bioenergetic function. Importantly, mitochondria retain bacterial motifs as a remnant of their endosymbiotic origin that are recognised by innate immune cells to trigger inflammation and participate in anti-microbial defence. This review aims to explore how mitochondrial physiology, spanning from oxidative phosphorylation (OxPhos) to signalling of mitochondrial nucleic acids, metabolites, and lipids, influences the effector functions of phagocytes. These myriad effector functions include macrophage polarisation, efferocytosis, anti-bactericidal activity, antigen presentation, immune signalling, and cytokine regulation. Strict regulation of these processes is critical for organismal homeostasis that when disrupted may cause injury or contribute to disease. Thus, the expanding body of literature, which continues to highlight the central role of mitochondria in the innate immune system, may provide insights for the development of the next generation of therapies for inflammatory diseases.
Collapse
Affiliation(s)
- Eloïse Marques
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Robbin Kramer
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Dylan G. Ryan
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
23
|
Rippon MG, Daly K, Rogers AA, Westgate S. Safety and effectiveness of an antiseptic wound cleansing and irrigation solution containing polyhexamethylene biguanide. J Wound Care 2024; 33:324-334. [PMID: 38683774 DOI: 10.12968/jowc.2024.33.5.324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 05/02/2024]
Abstract
OBJECTIVE There is currently a wide range of cleansing and irrigation solutions available for wounds, many of which contain antimicrobial agents. The aim of this study was to assess the safety of HydroClean Solution (HARTMANN, Germany), a polyhexamethylene biguanide (PHMB)-containing irrigation solution, in a standard cytotoxicity assay, and to assess its effect in a three-dimensional (3D) full-thickness model of human skin. METHOD A number of commercially available wound cleansing and irrigation solutions, including the PHMB-containing irrigation solution, were tested in a cytotoxicity assay using L929 mouse fibroblasts (ISO 10993-5:2009). The PHMB-containing irrigation solution was then assessed in an in vitro human keratinocyte-fibroblast 3D full-thickness wounded skin model to determine its effect on wound healing over six days. The effect of the PHMB-containing irrigation solution on tissue viability was measured using a lactate dehydrogenase (LDH) assay, and proinflammatory effects were measured using an interleukin-6 (IL-6) production assay. RESULTS The PHMB-containing irrigation solution was shown to be equivalent to other commercially available cleansing and irrigation solutions when tested in the L929 fibroblast cytotoxicity assay. When assessed in the in vitro 3D human full-thickness wound healing model, the PHMB-containing irrigation solution treatment resulted in no difference in levels of LDH or IL-6 when compared with levels produced in control Dulbecco's phosphate-buffered saline cultures. There was, however, a pronounced tissue thickening of the skin model in the periwound region. CONCLUSION The experimental data presented in this study support the conclusion that the PHMB-containing irrigation solution has a safety profile similar to other commercially available cleansing and irrigation solutions. Evidence also suggests that the PHMB-containing irrigation solution does not affect tissue viability or proinflammatory cytokine production, as evidenced by LDH levels or the production of IL-6 in a 3D human full-thickness wound healing model. The PHMB-containing irrigation solution stimulated new tissue growth in the periwound region of the skin model.
Collapse
Affiliation(s)
- Mark G Rippon
- Huddersfield University, Huddersfield, UK
- Daneriver Consultancy Ltd, Holmes Chapel, Cheshire, UK
| | | | | | | |
Collapse
|
24
|
Lin DW, Yang TM, Ho C, Shih YH, Lin CL, Hsu YC. Targeting Macrophages: Therapeutic Approaches in Diabetic Kidney Disease. Int J Mol Sci 2024; 25:4350. [PMID: 38673935 PMCID: PMC11050450 DOI: 10.3390/ijms25084350] [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] [Academic Contribution Register] [Received: 03/17/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetes is not solely a metabolic disorder but also involves inflammatory processes. The immune response it incites is a primary contributor to damage in target organs. Research indicates that during the initial phases of diabetic nephropathy, macrophages infiltrate the kidneys alongside lymphocytes, initiating a cascade of inflammatory reactions. The interplay between macrophages and other renal cells is pivotal in the advancement of kidney disease within a hyperglycemic milieu. While M1 macrophages react to the inflammatory stimuli induced by elevated glucose levels early in the disease progression, their subsequent transition to M2 macrophages, which possess anti-inflammatory and tissue repair properties, also contributes to fibrosis in the later stages of nephropathy by transforming into myofibroblasts. Comprehending the diverse functions of macrophages in diabetic kidney disease and regulating their activity could offer therapeutic benefits for managing this condition.
Collapse
Affiliation(s)
- Da-Wei Lin
- Department of Internal Medicine, St. Martin De Porres Hospital, Chiayi City 60069, Taiwan;
| | - Tsung-Ming Yang
- Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi County 61363, Taiwan;
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33303, Taiwan;
| | - Cheng Ho
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital, Chiayi County 61363, Taiwan;
| | - Ya-Hsueh Shih
- Departments of Nephrology, Chang Gung Memorial Hospital, Chiayi County 61363, Taiwan;
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi County 61363, Taiwan
| | - Chun-Liang Lin
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33303, Taiwan;
- Departments of Nephrology, Chang Gung Memorial Hospital, Chiayi County 61363, Taiwan;
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi County 61363, Taiwan
- Kidney Research Center, Chang Gung Memorial Hospital, Taipei 10507, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Yung-Chien Hsu
- Departments of Nephrology, Chang Gung Memorial Hospital, Chiayi County 61363, Taiwan;
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi County 61363, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33303, Taiwan
| |
Collapse
|
25
|
Giri PS, Rath SN. Macrophage Polarization Dynamics in Biomaterials: Implications for in Vitro Wound Healing. ACS APPLIED BIO MATERIALS 2024; 7:2413-2422. [PMID: 38536097 DOI: 10.1021/acsabm.4c00066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 04/16/2024]
Abstract
The interaction between biomaterials and the immune system plays a pivotal role in determining the success or failure of implantable devices. Macrophages, as key orchestrators of immune responses, exhibit diverse reactions that influence tissue integration or lead to implant failure. This study focuses on unraveling the intricate relationship between macrophage phenotypes and biomaterials, specifically hydrogels, by employing THP-1 cells as a model. Through a comprehensive investigation using polysaccharide, polymer, and protein-based hydrogels, our research sheds light on how the properties of hydrogels influence macrophage polarization. Phenotypic observations, biochemical assays, surface marker expression, and gene expression profiles collectively demonstrate the differential macrophage polarization abilities of polysaccharide-, polymer-, and protein-based hydrogels. Moreover, our indirect coculture studies reveal that hydrogels fostering M2 polarization exhibit exceptional wound-healing capabilities. These findings highlight the crucial role of the hydrogel microenvironment in adjusting macrophage polarization, offering a fresh avenue for refining biomaterials to bolster advantageous immune responses and improve tissue integration. This research contributes valuable insights for designing biomaterials with tailored properties that can guide macrophage behavior, ultimately improving the overall success of implantable devices.
Collapse
Affiliation(s)
- Pravin Shankar Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284 Telangana, India
| | - Subha Narayan Rath
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, 502284 Telangana, India
| |
Collapse
|
26
|
Dirand Z, Maraux M, Tissot M, Chatelain B, Supp D, Viennet C, Perruche S, Rolin G. Macrophage phenotype is determinant for fibrosis development in keloid disease. Matrix Biol 2024; 128:79-92. [PMID: 38485100 DOI: 10.1016/j.matbio.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/16/2023] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 04/02/2024]
Abstract
Keloid refers to a fibroproliferative disorder characterized by an accumulation of extracellular matrix (ECM) components at the dermis level, overgrowth beyond initial wound, and formation of tumor-like nodule areas. Treating keloid is still an unmet clinical need and the lack of an efficient therapy is clearly related to limited knowledge about keloid etiology, despite the growing interest of the scientific community in this pathology. In past decades, keloids were often studied in vitro through the sole prism of fibroblasts considered as the major effector of ECM deposition. Nevertheless, development of keloids results from cross-interactions of keloid fibroblasts (KFs) and their surrounding microenvironment, including immune cells such as macrophages. Our study aimed to evaluate the effect of M1 and M2 monocyte-derived macrophages on KFs in vitro. We focused on the effects of the macrophage secretome on fibrosis-related criteria in KFs, including proliferation, migration, differentiation, and ECM synthesis. First, we demonstrated that M2-like macrophages enhanced the fibrogenic profile of KFs in culture. Then, we surprisingly founded that M1-like macrophages can have an anti-fibrogenic effect on KFs, even in a pro-fibrotic environment. These results demonstrate, for the first time, that M1 and M2 macrophage subsets differentially impact the fibrotic fate of KFs in vitro, and suggest that restoring the M1/M2 balance to favor M1 in keloids could be an efficient therapeutic lever to prevent or treat keloid fibrosis.
Collapse
Affiliation(s)
- Zélie Dirand
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France
| | - Mélissa Maraux
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France
| | - Marion Tissot
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; DImaCell Imaging Resource Center, 25000 Besançon, France
| | - Brice Chatelain
- Service de Chirurgie Maxillo-faciale, Stomatologie et Odontologie Hospitalière, CHU Besançon, 25000 Besançon, France
| | - Dorothy Supp
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Scientific Staff, Shriners Children's Ohio, Dayton, Ohio, USA
| | - Céline Viennet
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; DImaCell Imaging Resource Center, 25000 Besançon, France
| | - Sylvain Perruche
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; MED'INN'Pharma 25000 Besançon, France
| | - Gwenaël Rolin
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; DImaCell Imaging Resource Center, 25000 Besançon, France; INSERM CIC-1431, CHU Besançon, 25000 Besançon, France.
| |
Collapse
|
27
|
Chen YF. Temporal Single-Cell Sequencing Analysis Reveals That GPNMB-Expressing Macrophages Potentiate Muscle Regeneration. RESEARCH SQUARE 2024:rs.3.rs-4108866. [PMID: 38585871 PMCID: PMC10996783 DOI: 10.21203/rs.3.rs-4108866/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 04/09/2024]
Abstract
Macrophages play a crucial role in coordinating the skeletal muscle repair response, but their phenotypic diversity and the transition of specialized subsets to resolution-phase macrophages remain poorly understood. To address this issue, we induced injury and performed single-cell RNA sequencing on individual cells in skeletal muscle at different time points. Our analysis revealed a distinct macrophage subset that expressed high levels of Gpnmb and that coexpressed critical factors involved in macrophage-mediated muscle regeneration, including Igf1, Mertk, and Nr1h3. Gpnmb gene knockout inhibited macrophage-mediated efferocytosis and impaired skeletal muscle regeneration. Functional studies demonstrated that GPNMB acts directly on muscle cells in vitro and improves muscle regeneration in vivo. These findings provide a comprehensive transcriptomic atlas of macrophages during muscle injury, highlighting the key role of the GPNMB macrophage subset in regenerative processes. Targeting GPNMB signaling in macrophages could have therapeutic potential for restoring skeletal muscle integrity and homeostasis.
Collapse
Affiliation(s)
- Yu-Fan Chen
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taiwan
| |
Collapse
|
28
|
Quartey BC, Sapudom J, ElGindi M, Alatoom A, Teo J. Matrix-Bound Hyaluronan Molecular Weight as a Regulator of Dendritic Cell Immune Potency. Adv Healthc Mater 2024; 13:e2303125. [PMID: 38104242 DOI: 10.1002/adhm.202303125] [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] [Academic Contribution Register] [Received: 09/17/2023] [Revised: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Hyaluronic acid (HA) is a glycosaminoglycan in the extracellular matrix with immunoregulatory properties depending on its molecular weight (MW). However, the impact of matrix-bound HA on dendritic cells (DCs) remains unclear due to varying distribution of HA MW under different physiological conditions. To investigate DCs in defined biosystems, 3D collagen matrices modified with HA of specific MW with similar microstructure and HA levels are used. It is found that HA MW influences cytokine binding to matrix, suggesting modulation of cytokine availability by the different HA MWs. These studies on DC immune potency reveal that low MW HA (8-15 kDa) enhances immature DC differentiation and antigen uptake, while medium (MMW-HA; 500-750 kDa) and high MW HA (HMW-HA; 1250-1500 kDa) increase cytokine secretion in mature DCs. The effect on DC phenotype and cytokine secretion by different MWs of HA is independent of CD44. However, blocking the CD44 receptor reveals its potential role in regulating acute inflammation through increased secretion of CCL2, CXCL8, and IL-6. Additionally, MMW- and HMW-HA matrices reduce migratory capacity of DCs, dependent on CD44. Overall, these findings provide insights into MW-dependent effects of matrix-bound HA on DCs, opening avenues for the design of DC-modulating materials to enhance DC-based therapy.
Collapse
Affiliation(s)
- Brian Chesney Quartey
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Mei ElGindi
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Aseel Alatoom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
- Department of Mechanical Engineering, Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, 11201, USA
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
- Department of Mechanical Engineering, Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, 11201, USA
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| |
Collapse
|
29
|
Toita R, Kitamura M, Tsuchiya A, Kang J, Kasahara S. Releasable, Immune-Instructive, Bioinspired Multilayer Coating Resists Implant-Induced Fibrosis while Accelerating Tissue Repair. Adv Healthc Mater 2024; 13:e2302611. [PMID: 38095751 PMCID: PMC11468989 DOI: 10.1002/adhm.202302611] [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] [Academic Contribution Register] [Received: 08/10/2023] [Indexed: 12/21/2023]
Abstract
Implantable biomaterials trigger foreign body reactions (FBRs), which reduces the functional life of medical devices and prevents effective tissue regeneration. Although existing therapeutic approaches can circumvent collagen-rich fibrotic encapsulation secondary to FBRs, they disrupt native tissue repair. Herein, a new surface engineering strategy in which an apoptotic-mimetic, immunomodulatory, phosphatidylserine liposome (PSL) is released from an implant coating to induce the formation of a macrophage phenotype that mitigates FBRs and improves tissue healing is described. PSL-multilayers constructed on implant surfaces via the layer-by-layer method release PSLs over a 1-month period. In rat muscles, poly(etheretherketone) (PEEK), a nondegradable polymer implant model, induces FBRs with dense fibrotic scarring under an aberrant cellular profile that recruits high levels of inflammatory infiltrates, foreign body giant cells (FBGCs), scar-forming myofibroblasts, and inflammatory M1-like macrophages but negligible amounts of anti-inflammatory M2-like phenotypes. However, the PSL-multilayer coating markedly diminishes these detrimental signatures by shifting the macrophage phenotype. Unlike other therapeutics, PSL-multilayered coatings also stimulate muscle regeneration. This study demonstrates that PSL-multilayered coatings are effective in eliminating FBRs and promoting regeneration, hence offering potent and broad applications for implantable biomaterials.
Collapse
Affiliation(s)
- Riki Toita
- Biomedical Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)1‐8‐31 MidorigaokaIkedaOsaka563‐8577Japan
- AIST‐Osaka University Advanced Photonics and Biosensing Open Innovation LaboratoryAIST2‐1 YamadaokaSuitaOsaka565‐0871Japan
| | - Masahiro Kitamura
- Niterra Co., Ltd.2808 IwasakiKomakiAichi485–8510Japan
- NGK Spark Plug‐AIST Healthcare Materials Cooperative Research Laboratory2266–98 AnagahoraShimoshidami, Moriyama‐kuNagoyaAichi463–8560Japan
| | - Akira Tsuchiya
- Department of BiomaterialsFaculty of Dental ScienceKyushu University3‐1‐1 MaidashiHigashi‐kuFukuoka812–8582Japan
| | - Jeong‐Hun Kang
- Division of Biopharmaceutics and PharmacokineticsNational Cerebral and Cardiovascular Center Research Institute6‐1 Shinmachi, KishibeSuitaOsaka564–8565Japan
| | | |
Collapse
|
30
|
Lin H, Zhang M, Hu M, Zhang Y, Jiang W, Tang W, Ouyang Y, Jiang L, Mi Y, Chen Z, He P, Zhao G, Ouyang X. Emerging applications of single-cell profiling in precision medicine of atherosclerosis. J Transl Med 2024; 22:97. [PMID: 38263066 PMCID: PMC10804726 DOI: 10.1186/s12967-023-04629-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/12/2023] [Accepted: 10/14/2023] [Indexed: 01/25/2024] Open
Abstract
Atherosclerosis is a chronic, progressive, inflammatory disease that occurs in the arterial wall. Despite recent advancements in treatment aimed at improving efficacy and prolonging survival, atherosclerosis remains largely incurable. In this review, we discuss emerging single-cell sequencing techniques and their novel insights into atherosclerosis. We provide examples of single-cell profiling studies that reveal phenotypic characteristics of atherosclerosis plaques, blood, liver, and the intestinal tract. Additionally, we highlight the potential clinical applications of single-cell analysis and propose that combining this approach with other techniques can facilitate early diagnosis and treatment, leading to more accurate medical interventions.
Collapse
Affiliation(s)
- Huiling Lin
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China
- Department of Physiology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Ming Zhang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, 511518, Guangdong, China
| | - Mi Hu
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China
| | - Yangkai Zhang
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China
| | - WeiWei Jiang
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wanying Tang
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China
| | - Yuxin Ouyang
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China
| | - Liping Jiang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yali Mi
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, 511518, Guangdong, China
| | - Zhi Chen
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Pingping He
- Department of Nursing, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China.
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, 511518, Guangdong, China.
| | - Xinping Ouyang
- Department of Physiology, Medical College, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, University of South China, Hengyang, 421001, Hunan, China.
- Department of Physiology, School of Medicine, Hunan Normal University, Changsha, 410081, Hunan, China.
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, 410081, Hunan, Changsha, China.
- The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, School of Medicine, Hunan Normal University, 410081, Hunan, Changsha, China.
| |
Collapse
|
31
|
Luo G, Zhou Z, Cao Z, Huang C, Li C, Li X, Deng C, Wu P, Yang Z, Tang J, Qing L. M2 macrophage-derived exosomes induce angiogenesis and increase skin flap survival through HIF1AN/HIF-1α/VEGFA control. Arch Biochem Biophys 2024; 751:109822. [PMID: 38030054 DOI: 10.1016/j.abb.2023.109822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/27/2023] [Revised: 10/24/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Skin flap transplantation is a routine strategy in plastic and reconstructive surgery for skin-soft tissue defects. Recent research has shown that M2 macrophages have the potential for pro-angiogenesis during tissue healing. METHODS In our research, we extracted the exosomes from M2 macrophages(M2-exo) and applied the exosomes in the model of skin flap transplantation. The flap survival area was measured, and the choke vessels were assessed by morphological observation. Hematoxylin and eosin (H&E) staining and Immunohistochemistry were applied to assess the neovascularization. The effect of M2-exo on the function of Human umbilical vein endothelial cells (HUVECs) was also investigated. We also administrated 2-methoxyestradiol (2-ME2, an inhibitor of HIF-1α) to explore the underlying mechanism. We tested the effects of M2-Exo on the proliferation of HUVECs through CCK8 assay and EdU staining assay. RESULTS The survival area and number of micro-vessels in the skin flaps were increased in the M2-exo group. Besides, the dilation rate of choke vessels was also enhanced in the M2-exo group. Additionally, compared with the control group, M2-exo could accelerate the proliferation, migration and tube formation of HUVECs in vitro. Furthermore, the expression of the pro-angiogenesis factors, HIF-1α and VEGFA, were overexpressed with the treatment of the M2-exo. The expression of HIF1AN protein level was decreased in the M2-exo group. Finally, treatment with HIF-1α inhibitor reverses the pro-survival effect of M2-exo on skin flaps by interfering with the HIF1AN/HIF-1α/VEGFA signaling pathway. CONCLUSION This study showed that M2-exosomes promote skin flap survival by enhancing angiogenesis, with HIF1AN/HIF-1α/VEGFA playing a crucial role in this process.
Collapse
Affiliation(s)
- Gaojie Luo
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zekun Zhou
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zheming Cao
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Chengxiong Huang
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Cheng Li
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxiao Li
- Department of Pathology, Changsha Medical University, Changsha, China
| | - Chao Deng
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Panfeng Wu
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenni Yang
- Hunan University of Medicine, Huaihua, China
| | - Juyu Tang
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.
| | - Liming Qing
- Department of Orthopedics, Hand and Microsurgery, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
32
|
Tolentino MJ, Tolentino AJ, Tolentino EM, Krishnan A, Genead MA. Sialic Acid Mimetic Microglial Sialic Acid-Binding Immunoglobulin-like Lectin Agonism: Potential to Restore Retinal Homeostasis and Regain Visual Function in Age-Related Macular Degeneration. Pharmaceuticals (Basel) 2023; 16:1735. [PMID: 38139861 PMCID: PMC10747662 DOI: 10.3390/ph16121735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/07/2023] [Revised: 11/29/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Age-related macular degeneration (AMD), a leading cause of visual loss and dysfunction worldwide, is a disease initiated by genetic polymorphisms that impair the negative regulation of complement. Proteomic investigation points to altered glycosylation and loss of Siglec-mediated glyco-immune checkpoint parainflammatory and inflammatory homeostasis as the main determinant for the vision impairing complications of macular degeneration. The effect of altered glycosylation on microglial maintained retinal para-inflammatory homeostasis and eventual recruitment and polarization of peripheral blood monocyte-derived macrophages (PBMDMs) into the retina can explain the phenotypic variability seen in this clinically heterogenous disease. Restoring glyco-immune checkpoint control with a sialic acid mimetic agonist targeting microglial/macrophage Siglecs to regain retinal para-inflammatory and inflammatory homeostasis is a promising therapeutic that could halt the progression of and improve visual function in all stages of macular degeneration.
Collapse
Affiliation(s)
- Michael J. Tolentino
- Department of Ophthalmology, University of Central Florida College of Medicine, Orlando, FL 32827, USA
- Department of Ophthalmology, Orlando College of Osteopathic Medicine, Orlando, FL 34787, USA
- Aviceda Therapeutics, Cambridge, MA 02142, USA; (A.K.); (M.A.G.)
| | - Andrew J. Tolentino
- Department of Biology, University of California Berkeley, Berkeley, CA 94720, USA;
| | | | - Anitha Krishnan
- Aviceda Therapeutics, Cambridge, MA 02142, USA; (A.K.); (M.A.G.)
| | | |
Collapse
|
33
|
Dyck C, Isaac KV, Edelstein-Keshet L. Models for Implant-Induced Capsular Contracture Post Breast Cancer Surgery. Bull Math Biol 2023; 86:7. [PMID: 38091110 PMCID: PMC10719149 DOI: 10.1007/s11538-023-01236-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/12/2023] [Accepted: 11/09/2023] [Indexed: 12/17/2023]
Abstract
Capsular contracture is a painful deformation of scar-tissue that may form around an implant in post-breast cancer reconstruction or cosmetic surgery. Inflammation due to surgical trauma or contamination in the tissue around the implant could account for recruitment of immune cells, and transdifferentiation of resident fibroblasts into cells that deposit abnormally thick collagen. Here we examine this hypothesis using a mathematical model for interacting macrophages, fibroblasts, myofibroblasts, and collagen. Our model demonstrates that cellular response can, together with inflammatory cell recruitment, account for prognoses.
Collapse
Affiliation(s)
- Cheryl Dyck
- Insight Math Unincorporated, Port Moody, BC, Canada
| | - Kathryn V Isaac
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Leah Edelstein-Keshet
- Department of Mathematics, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada.
| |
Collapse
|
34
|
Sapudom J, Karaman S, Quartey BC, Mohamed WKE, Mahtani N, Garcia-Sabaté A, Teo J. Collagen Fibril Orientation Instructs Fibroblast Differentiation Via Cell Contractility. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301353. [PMID: 37249413 PMCID: PMC10401101 DOI: 10.1002/advs.202301353] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Indexed: 05/31/2023]
Abstract
Collagen alignment is one of the key microarchitectural signatures of many pathological conditions, including scarring and fibrosis. Investigating how collagen alignment modulates cellular functions will pave the way for understanding tissue scarring and regeneration and new therapeutic strategies. However, current approaches for the fabrication of three-dimensional (3D) aligned collagen matrices are low-throughput and require special devices. To overcome these limitations, a simple approach to reconstitute homogeneous 3D collagen matrices with adjustable degree of fibril alignment using 3D printed inclined surfaces is developed. By characterizing the mechanical properties of reconstituted matrices, it is found that the elastic modulus of collagen matrices is enhanced with an increase in the alignment degree. The reconstituted matrices are used to study fibroblast behavior to reveal the progression of scar formation where a gradual enhancement of collagen alignment can be observed. It is found that matrices with aligned fibrils trigger fibroblast differentiation into myofibroblasts via cell contractility, while collagen stiffening through a crosslinker does not. The results suggest the impact of collagen fibril organization on the regulation of fibroblast differentiation. Overall, this approach to reconstitute 3D collagen matrices with fibril alignment opens opportunities for biomimetic pathological-relevant tissue in vitro, which can be applied for other biomedical research.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Shaza Karaman
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Brian Chesney Quartey
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Walaa Kamal Eldin Mohamed
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Nick Mahtani
- School of Engineering, Ecole Polytechnique Federale de Lausanne, Lausanne, 1015, Switzerland
| | - Anna Garcia-Sabaté
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
- Department of Mechanical and Biomedical Engineering, Tandon School of Engineering, New York University, New York, 11201, USA
| |
Collapse
|
35
|
ElGindi M, Sapudom J, Garcia Sabate A, Chesney Quartey B, Alatoom A, Al-Sayegh M, Li R, Chen W, Teo J. Effects of an aged tissue niche on the immune potency of dendritic cells using simulated microgravity. NPJ AGING 2023; 9:14. [PMID: 37393393 DOI: 10.1038/s41514-023-00111-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/15/2023] [Accepted: 05/18/2023] [Indexed: 07/03/2023]
Abstract
Microgravity accelerates the aging of various physiological systems, and it is well acknowledged that aged individuals and astronauts both have increased susceptibility to infections and poor response to vaccination. Immunologically, dendritic cells (DCs) are the key players in linking innate and adaptive immune responses. Their distinct and optimized differentiation and maturation phases play a critical role in presenting antigens and mounting effective lymphocyte responses for long-term immunity. Despite their importance, no studies to date have effectively investigated the effects of microgravity on DCs in their native microenvironment, which is primarily located within tissues. Here, we address a significantly outstanding research gap by examining the effects of simulated microgravity via a random positioning machine on both immature and mature DCs cultured in biomimetic collagen hydrogels, a surrogate for tissue matrices. Furthermore, we explored the effects of loose and dense tissues via differences in collagen concentration. Under these various environmental conditions, the DC phenotype was characterized using surface markers, cytokines, function, and transcriptomic profiles. Our data indicate that aged or loose tissue and exposure to RPM-induced simulated microgravity both independently alter the immunogenicity of immature and mature DCs. Interestingly, cells cultured in denser matrices experience fewer effects of simulated microgravity at the transcriptome level. Our findings are a step forward to better facilitate healthier future space travel and enhance our understanding of the aging immune system on Earth.
Collapse
Affiliation(s)
- Mei ElGindi
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Anna Garcia Sabate
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Brian Chesney Quartey
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Aseel Alatoom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Rui Li
- Department of Biomedical Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Weiqiang Chen
- Department of Biomedical Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
- Department of Mechanical and Aerospace Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates.
- Department of Biomedical Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA.
- Department of Mechanical and Aerospace Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA.
| |
Collapse
|
36
|
de Paula AP, de Lima JD, Bastos TSB, Czaikovski AP, dos Santos Luz RB, Yuasa BS, Smanioto CCS, Robert AW, Braga TT. Decellularized Extracellular Matrix: The Role of This Complex Biomaterial in Regeneration. ACS OMEGA 2023; 8:22256-22267. [PMID: 37396215 PMCID: PMC10308580 DOI: 10.1021/acsomega.2c06216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 10/04/2022] [Accepted: 01/12/2023] [Indexed: 07/04/2023]
Abstract
Organ transplantation is understood as a technique where an organ from a donor patient is transferred to a recipient patient. This practice gained strength in the 20th century and ensured advances in areas of knowledge such as immunology and tissue engineering. The main problems that comprise the practice of transplants involve the demand for viable organs and immunological aspects related to organ rejection. In this review, we address advances in tissue engineering for reversing the current challenges of transplants, focusing on the possible use of decellularized tissues in tissue engineering. We address the interaction of acellular tissues with immune cells, especially macrophages and stem cells, due to their potential use in regenerative medicine. Our goal is to exhibit data that demonstrate the use of decellularized tissues as alternative biomaterials that can be applied clinically as partial or complete organ substitutes.
Collapse
Affiliation(s)
| | - Jordana Dinorá de Lima
- Department
of Pathology, Federal University of Parana, Curitiba, Parana 80060-000, Brazil
| | | | | | | | - Bruna Sadae Yuasa
- Department
of Pathology, Federal University of Parana, Curitiba, Parana 80060-000, Brazil
| | | | - Anny Waloski Robert
- Stem
Cells Basic Biology Laboratory, Carlos Chagas
Institute − FIOCRUZ/PR, Curitiba, Parana 81350-010, Brazil
| | - Tárcio Teodoro Braga
- Department
of Pathology, Federal University of Parana, Curitiba, Parana 80060-000, Brazil
- Graduate
Program in Biosciences and Biotechnology, Institute Carlos Chagas, Fiocruz, Parana 81310-020, Brazil
| |
Collapse
|
37
|
Zhang W, Wu Q, Hao S, Chen S. The hallmark and crosstalk of immune cells after intracerebral hemorrhage: Immunotherapy perspectives. Front Neurosci 2023; 16:1117999. [PMID: 36711145 PMCID: PMC9877537 DOI: 10.3389/fnins.2022.1117999] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/07/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is one of the most dangerous types of strokes with a high morbidity and mortality rate. Currently, the treatment of ICH is not well developed, mainly because its mechanisms are still unclear. Inflammation is one of the main types of secondary injury after ICH and catalyzes the adverse consequences of ICH. A large number of immune cells are involved in neuroinflammation, such as microglia, astrocytes, oligodendrocytes, lymphocytes, macrophages, and neutrophils. Nevertheless, the characteristics and crosstalk of immune cells have not been fully elucidated. In this review, we endeavor to delve into the respective characteristics of immune cells and their interactions in neuroimmune inflammation, and further elucidate favorable immunotherapeutic approaches regarding ICH, and finally present an outlook.
Collapse
Affiliation(s)
- Wenqing Zhang
- School of Medicine, Chongqing University, Chongqing, China,Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Qingyuan Wu
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China,*Correspondence: Shilei Hao,
| | - Shengli Chen
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China,Shengli Chen,
| |
Collapse
|
38
|
Mamilos A, Winter L, Schmitt VH, Barsch F, Grevenstein D, Wagner W, Babel M, Keller K, Schmitt C, Gürtler F, Schreml S, Niedermair T, Rupp M, Alt V, Brochhausen C. Macrophages: From Simple Phagocyte to an Integrative Regulatory Cell for Inflammation and Tissue Regeneration-A Review of the Literature. Cells 2023; 12:276. [PMID: 36672212 PMCID: PMC9856654 DOI: 10.3390/cells12020276] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/29/2022] [Revised: 12/29/2022] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
The understanding of macrophages and their pathophysiological role has dramatically changed within the last decades. Macrophages represent a very interesting cell type with regard to biomaterial-based tissue engineering and regeneration. In this context, macrophages play a crucial role in the biocompatibility and degradation of implanted biomaterials. Furthermore, a better understanding of the functionality of macrophages opens perspectives for potential guidance and modulation to turn inflammation into regeneration. Such knowledge may help to improve not only the biocompatibility of scaffold materials but also the integration, maturation, and preservation of scaffold-cell constructs or induce regeneration. Nowadays, macrophages are classified into two subpopulations, the classically activated macrophages (M1 macrophages) with pro-inflammatory properties and the alternatively activated macrophages (M2 macrophages) with anti-inflammatory properties. The present narrative review gives an overview of the different functions of macrophages and summarizes the recent state of knowledge regarding different types of macrophages and their functions, with special emphasis on tissue engineering and tissue regeneration.
Collapse
Affiliation(s)
- Andreas Mamilos
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Lina Winter
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Volker H. Schmitt
- Department of Cardiology, University Medical Centre, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, 55131 Mainz, Germany
| | - Friedrich Barsch
- Medical Center, Faculty of Medicine, Institute for Exercise and Occupational Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - David Grevenstein
- Clinic and Polyclinic for Orthopedics and Trauma Surgery, University Hospital of Cologne, 50937 Cologne, Germany
| | - Willi Wagner
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Translational Lung Research Centre Heidelberg (TLRC), German Lung Research Centre (DZL), 69120 Heidelberg, Germany
| | - Maximilian Babel
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Karsten Keller
- Department of Cardiology, University Medical Centre, Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Department of Sports Medicine, Medical Clinic VII, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Christine Schmitt
- Department of Internal Medicine, St. Vincenz and Elisabeth Hospital of Mainz (KKM), 55131 Mainz, Germany
| | - Florian Gürtler
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Stephan Schreml
- Department of Dermatology, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Tanja Niedermair
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Markus Rupp
- Department for Trauma Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Volker Alt
- Department for Trauma Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
- Central Biobank Regensburg, University and University Hospital Regensburg, 93053 Regensburg, Germany
- Institute of Pathology, University Medical Centre Mannheim, Ruprecht-Karls-University Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
39
|
Zhang Y, Cao J, Jian M, Zhou Z, Anwar N, Xiao L, Ma Y, Zhang D, Zhang J, Wang X. Fabrication of Interleukin-4 Encapsulated Bioactive Microdroplets for Regulating Inflammation and Promoting Osteogenesis. Int J Nanomedicine 2023; 18:2019-2035. [PMID: 37155503 PMCID: PMC10122853 DOI: 10.2147/ijn.s397359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/20/2022] [Accepted: 04/11/2023] [Indexed: 05/10/2023] Open
Abstract
Background Despite the inherent regenerative ability of bone, large bone defect regeneration remains a major clinical challenge for orthopedic surgery. Therapeutic strategies medicated by M2 phenotypic macrophages or M2 macrophage inducer have been widely used to promote tissue remodeling. In this study, ultrasound-responsive bioactive microdroplets (MDs) encapsulated with bioactive molecule interleukin-4 (IL4, hereafter designated MDs-IL4) were fabricated to regulate macrophage polarization and potentiate the osteogenic differentiation of human mesenchymal stem cells (hBMSCs). Materials and Methods The MTT assay, live and dead staining, and phalloidin/DAPI dual staining were used to evaluate biocompatibility in vitro. H&E staining was used to evaluate biocompatibility in vivo. Inflammatory macrophages were further induced via lipopolysaccharide (LPS) stimulation to mimic the pro-inflammatory condition. The immunoregulatory role of the MDs-IL4 was tested via macrophage phenotypic marker gene expression, pro-inflammatory cytokine level, cell morphological analysis, and immunofluorescence staining, etc. The immune-osteogenic response of hBMSCs via macrophages and hBMSCs interactions was further investigated in vitro. Results The bioactive MDs-IL4 scaffold showed good cytocompatibility in RAW 264.7 macrophages and hBMSCs. The results confirmed that the bioactive MDs-IL4 scaffold could reduce inflammatory phenotypic macrophages, as evidenced by changing in morphological features, reduction in pro-inflammatory marker gene expression, increase of M2 phenotypic marker genes, and inhibition of pro-inflammatory cytokine secretion. Additionally, our results indicate that the bioactive MDs-IL4 could significantly enhance the osteogenic differentiation of hBMSCs via its potential immunomodulatory properties. Conclusion Our results demonstrate that the bioactive MDs-IL4 scaffold could be used as novel carrier system for other pro-osteogenic molecules, thus having potential applications in bone tissue regeneration.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Hygiene Toxicology, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
| | - Jin Cao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, People’s Republic of China
| | - Minghui Jian
- Department of Hygiene Toxicology, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
| | - Zhixiao Zhou
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, People’s Republic of China
| | - Nadia Anwar
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, People’s Republic of China
| | - Lan Xiao
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, 4059, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, 4059, Australia
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, People’s Republic of China
| | - Dingmei Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, People’s Republic of China
| | - Jun Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, People’s Republic of China
| | - Xin Wang
- Department of Hygiene Toxicology, Zunyi Medical University, Zunyi, Guizhou, 563000, People’s Republic of China
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, People’s Republic of China
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, 4059, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, QLD, 4059, Australia
- Correspondence: Xin Wang, Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, People’s Republic of China, Tel +86 136 3928 8558, Fax +86-851-2860 8903, Email
| |
Collapse
|
40
|
Cialdai F, Risaliti C, Monici M. Role of fibroblasts in wound healing and tissue remodeling on Earth and in space. Front Bioeng Biotechnol 2022; 10:958381. [PMID: 36267456 PMCID: PMC9578548 DOI: 10.3389/fbioe.2022.958381] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/31/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
Wound healing (WH) and the role fibroblasts play in the process, as well as healing impairment and fibroblast dysfunction, have been thoroughly reviewed by other authors. We treat these topics briefly, with the only aim of contextualizing the true focus of this review, namely, the microgravity-induced changes in fibroblast functions involved in WH. Microgravity is a condition typical of spaceflight. Studying its possible effects on fibroblasts and WH is useful not only for the safety of astronauts who will face future interplanetary space missions, but also to help improve the management of WH impairment on Earth. The interesting similarity between microgravity-induced alterations of fibroblast behavior and fibroblast dysfunction in WH impairment on Earth is highlighted. The possibility of using microgravity-exposed fibroblasts and WH in space as models of healing impairment on Earth is suggested. The gaps in knowledge on fibroblast functions in WH are analyzed. The contribution that studies on fibroblast behavior in weightlessness can make to fill these gaps and, consequently, improve therapeutic strategies is considered.
Collapse
|
41
|
ElGindi M, Sapudom J, Laws P, Garcia-Sabaté A, Daqaq MF, Teo J. 3D microenvironment attenuates simulated microgravity-mediated changes in T cell transcriptome. Cell Mol Life Sci 2022; 79:508. [PMID: 36063234 PMCID: PMC11803002 DOI: 10.1007/s00018-022-04531-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/20/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 11/03/2022]
Abstract
Human space travel and exploration are of interest to both the industrial and scientific community. However, there are many adverse effects of spaceflight on human physiology. In particular, there is a lack of understanding of the extent to which microgravity affects the immune system. T cells, key players of the adaptive immune system and long-term immunity, are present not only in blood circulation but also reside within the tissue. As of yet, studies investigating the effects of microgravity on T cells are limited to peripheral blood or traditional 2D cell culture that recapitulates circulating blood. To better mimic interstitial tissue, 3D cell culture has been well established for physiologically and pathologically relevant models. In this work, we utilize 2D cell culture and 3D collagen matrices to gain an understanding of how simulated microgravity, using a random positioning machine, affects both circulating and tissue-resident T cells. T cells were studied in both resting and activated stages. We found that 3D cell culture attenuates the effects of simulated microgravity on the T cells transcriptome and nuclear irregularities compared to 2D cell culture. Interestingly, simulated microgravity appears to have less effect on activated T cells compared to those in the resting stage. Overall, our work provides novel insights into the effects of simulated microgravity on circulating and tissue-resident T cells which could provide benefits for the health of space travellers.
Collapse
Affiliation(s)
- Mei ElGindi
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Praveen Laws
- Laboratory of Applied Nonlinear Dynamics, Division of Engineering, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Anna Garcia-Sabaté
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Mohammed F Daqaq
- Laboratory of Applied Nonlinear Dynamics, Division of Engineering, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates.
- Department of Mechanical and Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA.
| |
Collapse
|
42
|
Kaushal A, Zhang Y, Ballantyne LL, Fitzpatrick LE. The extended effect of adsorbed damage-associated molecular patterns and Toll-like receptor 2 signaling on macrophage-material interactions. Front Bioeng Biotechnol 2022; 10:959512. [PMID: 36091432 PMCID: PMC9458975 DOI: 10.3389/fbioe.2022.959512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/01/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022] Open
Abstract
Implanted biomaterials elicit an immune-mediated foreign body reaction (FBR) that results in the fibrous encapsulation of the implant and can critically impact the performance of some implants. Consequently, understanding the molecular mechanisms that underpin cell-materials interactions that initiate biomaterial-induced inflammation and fibrosis is critical to improving the performance of biomaterial implants negatively impacted by the FBR. Damage-associated molecular patterns (DAMPs) are endogenous mediators of inflammation that are released upon tissue injury and induce sterile inflammation via Toll-like receptors (TLRs). However, the prevalence of DAMPs within the adsorbed protein layer on material surfaces and their role mediating cell-material interactions is unclear. Previously, our group demonstrated that molecules in fibroblast lysates adsorbed to various biomaterials and induced a potent TLR2-dependent inflammatory response in macrophages at 24 h. In this study, we examined the extended response of RAW-Blue reporter macrophages on lysate or serum-adsorbed Teflon™ AF surfaces to understand the potential role of adsorbed DAMPs in macrophage-material interactions at later time points. Lysate-conditioned surfaces maintained increased nuclear factor kappa B (NF-κB) and activator protein 1 (AP-1) transcription factor activity and increased expression Regulated upon Activation, Normal T Cell Expressed and Presumably Secreted (RANTES/CCL5) at 72 h and 120 h, compared to FBS-conditioned surfaces. In contrast, monocyte chemoattractant protein 1 (MCP-1/CCL2) was only elevated at 72 h in lysate conditions. Transforming growth factor beta 1 (TGF-β1) secretion was significantly increased on lysate-conditioned surfaces, while conditioned media from macrophages on lysate-conditioned surfaces induced alpha smooth muscle actin (αSMA) expression in 3T3 fibroblasts. TLR2 neutralizing antibody treatment significantly decreased NF-κB/AP-1 activity and attenuated TGF-β1 expression at both time points, and MCP-1 and RANTES at 72 h. Finally, multinucleated cells were observed on lysate-conditioned surfaces at 72 h, indicating adsorbed DAMPs induced a fusion permissive environment for adherent macrophages. This study demonstrates that adsorbed DAMPs continue to influence macrophage-material responses beyond the initial 24-h period and maintain a pro-inflammatory and fibrotic response that models aspects of the early FBR. Furthermore, the transient inhibition of TLR2 continued to exert an effect at these later time points, suggesting TLR2 may be a target for therapeutic interventions in FBR.
Collapse
Affiliation(s)
- Anuj Kaushal
- Department of Chemical Engineering, Queen’s University, Kingston, ON, Canada
| | - Yuxi Zhang
- Department of Chemical Engineering, Queen’s University, Kingston, ON, Canada
| | - Laurel L. Ballantyne
- Department of Chemical Engineering, Queen’s University, Kingston, ON, Canada
- The Centre for Health Innovation, Queen’s University and the Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Lindsay E. Fitzpatrick
- Department of Chemical Engineering, Queen’s University, Kingston, ON, Canada
- The Centre for Health Innovation, Queen’s University and the Kingston Health Sciences Centre, Kingston, ON, Canada
- *Correspondence: Lindsay E. Fitzpatrick,
| |
Collapse
|
43
|
Inflammation-mediated matrix remodeling of extracellular matrix-mimicking biomaterials in tissue engineering and regenerative medicine. Acta Biomater 2022; 151:106-117. [PMID: 35970482 DOI: 10.1016/j.actbio.2022.08.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/27/2022] [Revised: 06/30/2022] [Accepted: 08/08/2022] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM)-mimicking biomaterials are considered effective tissue-engineered scaffolds for regenerative medicine because of their biocompatibility, biodegradability, and bioactivity. ECM-mimicking biomaterials preserve natural microstructures and matrix-related bioactive components and undergo continuous matrix remodeling upon transplantation. The interaction between host immune cells and transplanted ECM-mimicking biomaterials has attracted considerable attention in recent years. Transplantation of biomaterials may initiate injuries and early pro-inflammation reactions characterized by infiltration of neutrophils and M1 macrophages. Pro-inflammation reactions may lead to degradation of the transplanted biomaterial and drive the matrix into a fetal-like state. ECM degradation leads to the release of matrix-related bioactive components that act as signals for cell migration, proliferation, and differentiation. In late stages, pro-inflammatory cells fade away, and anti-inflammatory cells emerge, which involves macrophage polarization to the M2 phenotype and leukocyte activation to T helper 2 (Th2) cells. These anti-inflammatory cells interact with each other to facilitate matrix deposition and tissue reconstruction. Deposited ECM molecules serve as vital components of the mature tissue and influence tissue homeostasis. However, dysregulation of matrix remodeling results in several pathological conditions, such as aggressive inflammation, difficult healing, and non-functional fibrosis. In this review, we summarize the characteristics of inflammatory responses in matrix remodeling after transplantation of ECM-mimicking biomaterials. Additionally, we discuss the intrinsic linkages between matrix remodeling and tissue regeneration. STATEMENT OF SIGNIFICANCE: Extracellular matrix (ECM)-mimicking biomaterials are effectively used as scaffolds in tissue engineering and regenerative medicine. However, dysregulation of matrix remodeling can cause various pathological conditions. Here, the review describes the characteristics of inflammatory responses in matrix remodeling after transplantation of ECM-mimicking biomaterials. Additionally, we discuss the intrinsic linkages between matrix remodeling and tissue regeneration. We believe that understanding host immune responses to matrix remodeling of transplanted biomaterials is important for directing effective tissue regeneration of ECM-mimicking biomaterials. Considering the close relationship between immune response and matrix remodeling results, we highlight the need for studies of the effects of clinical characteristics on matrix remodeling of transplanted biomaterials.
Collapse
|
44
|
Song Q, Zhang Y, Zhou M, Xu Y, Zhang Q, Wu L, Liu S, Zhang M, Zhang L, Wu Z, Peng W, Liu X, Zhao C. The Culture Dish Surface Influences the Phenotype and Dissociation Strategy in Distinct Mouse Macrophage Populations. Front Immunol 2022; 13:920232. [PMID: 35874686 PMCID: PMC9299442 DOI: 10.3389/fimmu.2022.920232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/14/2022] [Accepted: 06/10/2022] [Indexed: 12/04/2022] Open
Abstract
The nature of the culture dish surface and the technique used to detach adherent cells could very likely influence the cell viability and cell membrane protein integrity of harvested macrophages. Several previous studies assessed the detachment efficacies of enzymatic and non-enzymatic methods for harvesting the single cell suspensions of macrophages, but a comprehensive study assessing different dissociation methods and culture conditions for detaching functionally different macrophage populations has not yet been reported. In this study, via the well-established GM-CSF and M-CSF differentiated bone marrow derived macrophage models (GM-BMDMs and M-BMDMs), we compared four commonly used enzymatic (trypsin and accutase) and non-enzymatic (PBS and EDTA) dissociation methods along with necessary mechanical detaching steps (scraping and pipetting) to evaluate the viable cell recovery and cell surface marker integrality of GM-BMDMs and M-BMDMs cultured on standard cell culture dish (TC dish), or on culture dish (noTC dish) that was not conditioned to enhance adherence. The data showed that accutase yielded a better recovery of viable cells comparing with PBS and EDTA, especially for tightly adherent GM-BMDMs on TC dishes, with a relatively higher level of detected cell membrane marker F4/80 than trypsin. An additional gradient centrifugation-based dead cell removal approach could increase the proportion of viable cells for TC cultured GM-BMDMs after accutase dissociation. Furthermore, transcriptome analysis was performed to evaluate the putative influence of culture dishes. At steady state, BMDMs cultured on noTC dishes exhibited more proinflammatory gene expression signatures (e.g. IL6, CXCL2 and ILlβ) and functions (e.g. TNF and IL17 signaling pathways). Similar inflammatory responses were observed upon LPS challenge regardless of culture conditions and differentiation factors. However, in LPS treated samples, the difference of gene expression patterns, signaling pathways and molecular functions between TC and noTC cultured BMDMs were largely dependent on the types of growth factors (M-CSF and GM-CSF). This observation might provide valuable information for in vitro macrophage studies.
Collapse
Affiliation(s)
- Qiaoling Song
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Innovation Platform of Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yazhuo Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Mingming Zhou
- Innovation Platform of Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yuting Xu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Qianyue Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Lihong Wu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shan Liu
- Innovation Platform of Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Minghui Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Lei Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Innovation Platform of Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Zhihua Wu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Weixun Peng
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Xutao Liu
- Samueli School of Engineering, University of California Los Angeles, Los Angeles, CA, United States
| | - Chenyang Zhao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Innovation Platform of Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
45
|
Nascimento MT, Cordeiro RSO, Abreu C, Santos CP, Peixoto F, Duarte GA, Cardoso T, de Oliveira CI, Carvalho E, Carvalho LP. Pioglitazone, a Peroxisome Proliferator-Activated Receptor-γ Agonist, Downregulates the Inflammatory Response in Cutaneous Leishmaniasis Patients Without Interfering in Leishmania braziliensis Killing by Monocytes. Front Cell Infect Microbiol 2022; 12:884237. [PMID: 35909958 PMCID: PMC9329526 DOI: 10.3389/fcimb.2022.884237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/25/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
Patients with cutaneous leishmaniasis (CL) due to Leishmania braziliensis infection have an exacerbated inflammatory response associated with tissue damage and ulcer development. An increase in the rate of patients who fail therapy with pentavalent antimony has been documented. An adjuvant therapy with an anti-inflammatory drug with the potential of Leishmania killing would benefit CL patients. The aim of the present study was to investigate the contribution of peroxisome proliferator-activated receptor-γ (PPAR-γ) activation by pioglitazone in the regulation of the inflammatory response and L. braziliensis killing by monocytes. Pioglitazone is an oral drug used in the treatment of diabetes, and its main mechanism of action is through the activation of PPAR-γ, which is expressed in many cell types of the immune response. We found that activation of PPAR-γ by pioglitazone decreases the inflammatory response in CL patients without affecting L. braziliensis killing by monocytes. Our data suggest that pioglitazone may serve as an adjunctive treatment for CL caused by L. braziliensis.
Collapse
Affiliation(s)
- Maurício T. Nascimento
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Ravena S. O. Cordeiro
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Cayo Abreu
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Camila P. Santos
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Fábio Peixoto
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Gabriela A. Duarte
- Laboratório de Enfermidades Infecciosas Transmitidas por Vetores, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
| | - Thiago Cardoso
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Camila I. de Oliveira
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
- Laboratório de Enfermidades Infecciosas Transmitidas por Vetores, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Ministério de Ciências e Tecnologia, Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Edgar M. Carvalho
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
- Ministério de Ciências e Tecnologia, Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| | - Lucas P. Carvalho
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Brazil
- Serviço de Imunologia, Complexo Hospitalar Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
- Ministério de Ciências e Tecnologia, Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, Brazil
| |
Collapse
|
46
|
Carraro E, Rossi L, Maghin E, Canton M, Piccoli M. 3D in vitro Models of Pathological Skeletal Muscle: Which Cells and Scaffolds to Elect? Front Bioeng Biotechnol 2022; 10:941623. [PMID: 35898644 PMCID: PMC9313593 DOI: 10.3389/fbioe.2022.941623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2022] [Accepted: 06/21/2022] [Indexed: 12/29/2022] Open
Abstract
Skeletal muscle is a fundamental tissue of the human body with great plasticity and adaptation to diseases and injuries. Recreating this tissue in vitro helps not only to deepen its functionality, but also to simulate pathophysiological processes. In this review we discuss the generation of human skeletal muscle three-dimensional (3D) models obtained through tissue engineering approaches. First, we present an overview of the most severe myopathies and the two key players involved: the variety of cells composing skeletal muscle tissue and the different components of its extracellular matrix. Then, we discuss the peculiar characteristics among diverse in vitro models with a specific focus on cell sources, scaffold composition and formulations, and fabrication techniques. To conclude, we highlight the efficacy of 3D models in mimicking patient-specific myopathies, deepening muscle disease mechanisms or investigating possible therapeutic effects.
Collapse
Affiliation(s)
- Eugenia Carraro
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lucia Rossi
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Edoardo Maghin
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Marcella Canton
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Martina Piccoli
- Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- *Correspondence: Martina Piccoli,
| |
Collapse
|
47
|
Zhang Q, Burrell JC, Zeng J, Motiwala FI, Shi S, Cullen DK, Le AD. Implantation of a nerve protector embedded with human GMSC-derived Schwann-like cells accelerates regeneration of crush-injured rat sciatic nerves. Stem Cell Res Ther 2022; 13:263. [PMID: 35725660 PMCID: PMC9208168 DOI: 10.1186/s13287-022-02947-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/22/2022] [Accepted: 06/08/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Peripheral nerve injuries (PNIs) remain one of the great clinical challenges because of their considerable long-term disability potential. Postnatal neural crest-derived multipotent stem cells, including gingiva-derived mesenchymal stem cells (GMSCs), represent a promising source of seed cells for tissue engineering and regenerative therapy of various disorders, including PNIs. Here, we generated GMSC-repopulated nerve protectors and evaluated their therapeutic effects in a crush injury model of rat sciatic nerves. METHODS GMSCs were mixed in methacrylated collagen and cultured for 48 h, allowing the conversion of GMSCs into Schwann-like cells (GiSCs). The phenotype of GiSCs was verified by fluorescence studies on the expression of Schwann cell markers. GMSCs encapsulated in the methacrylated 3D-collagen hydrogel were co-cultured with THP-1-derived macrophages, and the secretion of anti-inflammatory cytokine IL-10 or inflammatory cytokines TNF-α and IL-1β in the supernatant was determined by ELISA. In addition, GMSCs mixed in the methacrylated collagen were filled into a nerve protector made from the decellularized small intestine submucosal extracellular matrix (SIS-ECM) and cultured for 24 h, allowing the generation of functionalized nerve protectors repopulated with GiSCs. We implanted the nerve protector to wrap the injury site of rat sciatic nerves and performed functional and histological assessments 4 weeks post-surgery. RESULTS GMSCs encapsulated in the methacrylated 3D-collagen hydrogel were directly converted into Schwann-like cells (GiSCs) characterized by the expression of S-100β, p75NTR, BDNF, and GDNF. In vitro, co-culture of GMSCs encapsulated in the 3D-collagen hydrogel with macrophages remarkably increased the secretion of IL-10, an anti-inflammatory cytokine characteristic of pro-regenerative (M2) macrophages, but robustly reduced LPS-stimulated secretion of TNF-1α and IL-1β, two cytokines characteristic of pro-inflammatory (M1) macrophages. In addition, our results indicate that implantation of functionalized nerve protectors repopulated with GiSCs significantly accelerated functional recovery and axonal regeneration of crush-injured rat sciatic nerves accompanied by increased infiltration of pro-regenerative (M2) macrophages while a decreased infiltration of pro-inflammatory (M1) macrophages. CONCLUSIONS Collectively, these findings suggest that Schwann-like cells converted from GMSCs represent a promising source of supportive cells for regenerative therapy of PNI through their dual functions, neurotrophic effects, and immunomodulation of pro-inflammatory (M1)/pro-regenerative (M2) macrophages.
Collapse
Affiliation(s)
- Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA, 19104, USA.
| | - Justin C. Burrell
- grid.25879.310000 0004 1936 8972Department of Neurosurgery, Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA USA ,grid.410355.60000 0004 0420 350XCenter for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104 USA
| | - Jincheng Zeng
- grid.25879.310000 0004 1936 8972Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA 19104 USA ,grid.410560.60000 0004 1760 3078Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan, 523808 China
| | - Faizan I. Motiwala
- grid.25879.310000 0004 1936 8972Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA 19104 USA
| | - Shihong Shi
- grid.25879.310000 0004 1936 8972Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA 19104 USA
| | - D. Kacy Cullen
- grid.25879.310000 0004 1936 8972Department of Neurosurgery, Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA USA ,grid.410355.60000 0004 0420 350XCenter for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104 USA
| | - Anh D. Le
- grid.25879.310000 0004 1936 8972Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA 19104 USA ,grid.411115.10000 0004 0435 0884Department of Oral and Maxillofacial Surgery, Perelman Center for Advanced Medicine, Penn Medicine Hospital of the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104 USA
| |
Collapse
|
48
|
Damiani V, Cufaro MC, Fucito M, Dufrusine B, Rossi C, Del Boccio P, Federici L, Turco MC, Sallese M, Pieragostino D, De Laurenzi V. Proteomics Approach Highlights Early Changes in Human Fibroblasts-Pancreatic Ductal Adenocarcinoma Cells Crosstalk. Cells 2022; 11:1160. [PMID: 35406724 PMCID: PMC8997741 DOI: 10.3390/cells11071160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/18/2022] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 02/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer mortality worldwide. Non-specific symptoms, lack of biomarkers in the early stages, and drug resistance due to the presence of a dense fibrous stroma all contribute to the poor outcome of this disease. The extracellular matrix secreted by activated fibroblasts contributes to the desmoplastic tumor microenvironment formation. Given the importance of fibroblast activation in PDAC pathology, it is critical to recognize the mechanisms involved in the transformation of normal fibroblasts in the early stages of tumorigenesis. To this aim, we first identified the proteins released from the pancreatic cancer cell line MIA-PaCa2 by proteomic analysis of their conditioned medium (CM). Second, normal fibroblasts were treated with MIA-PaCa2 CM for 24 h and 48 h and their proteostatic changes were detected by proteomics. Pathway analysis indicated that treated fibroblasts undergo changes compatible with the activation of migration, vasculogenesis, cellular homeostasis and metabolism of amino acids and reduced apoptosis. These biological activities are possibly regulated by ITGB3 and TGFB1/2 followed by SMAD3, STAT3 and BAG3 activation. In conclusion, this study sheds light on the crosstalk between PDAC cells and associated fibroblasts. Data are available via ProteomeXchange with identifier PXD030974.
Collapse
Affiliation(s)
- Verena Damiani
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (V.D.); (M.F.); (B.D.); (C.R.); (L.F.); (M.S.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
| | - Maria Concetta Cufaro
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Maurine Fucito
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (V.D.); (M.F.); (B.D.); (C.R.); (L.F.); (M.S.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
| | - Beatrice Dufrusine
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (V.D.); (M.F.); (B.D.); (C.R.); (L.F.); (M.S.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
| | - Claudia Rossi
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (V.D.); (M.F.); (B.D.); (C.R.); (L.F.); (M.S.); (V.D.L.)
- Department of Psychological, Health and Territory Sciences, School of Medicine and Health Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Piero Del Boccio
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Luca Federici
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (V.D.); (M.F.); (B.D.); (C.R.); (L.F.); (M.S.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
| | - Maria Caterina Turco
- Department of Medicine, Surgery and Dentistry Schola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy;
- R&D Division, BIOUNIVERSA s.r.l., 84081 Baronissi, Italy
| | - Michele Sallese
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (V.D.); (M.F.); (B.D.); (C.R.); (L.F.); (M.S.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
| | - Damiana Pieragostino
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (V.D.); (M.F.); (B.D.); (C.R.); (L.F.); (M.S.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (V.D.); (M.F.); (B.D.); (C.R.); (L.F.); (M.S.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.C.C.); (P.D.B.)
| |
Collapse
|
49
|
Guo Y, Tsai HI, Zhang L, Zhu H. Mitochondrial DNA on Tumor-Associated Macrophages Polarization and Immunity. Cancers (Basel) 2022; 14:1452. [PMID: 35326602 PMCID: PMC8946090 DOI: 10.3390/cancers14061452] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2022] [Revised: 02/26/2022] [Accepted: 03/09/2022] [Indexed: 12/04/2022] Open
Abstract
As the richest immune cells in most tumor microenvironments (TMEs), tumor-associated macrophages (TAMs) play an important role in tumor development and treatment sensitivity. The phenotypes and functions of TAMs vary according to their sources and tumor progression. Different TAM phenotypes display distinct behaviors in terms of tumor immunity and are regulated by intracellular and exogenous molecules. Additionally, dysfunctional and oxidatively stressed mitochondrial-derived mitochondrial DNA (mtDNA) plays an important role in remodeling the phenotypes and functions of TAMs. This article reviews the interactions between mtDNA and TAMs in the TME and further discusses the influence of their performance on tumor genesis and development.
Collapse
Affiliation(s)
- Yaxin Guo
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Hsiang-i Tsai
- Laboratory of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Lirong Zhang
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Haitao Zhu
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
- Laboratory of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| |
Collapse
|