1
|
Bauer TM, Moon J, Shadiow J, Buckley S, Gallagher KA. Mechanisms of Impaired Wound Healing in Type 2 Diabetes: The Role of Epigenetic Factors. Arterioscler Thromb Vasc Biol 2025; 45:632-642. [PMID: 40109262 PMCID: PMC12018132 DOI: 10.1161/atvbaha.124.321446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Despite decades of research, impaired extremity wound healing in type 2 diabetes remains a significant driver of patient morbidity, mortality, and health care costs. Advances in surgical and medical therapies, including the advent of endovascular interventions for peripheral artery disease and topical therapies developed to promote wound healing, have not reduced the frequency of lower leg amputations for nonhealing wounds in type 2 diabetes. This brief report is aimed at reviewing the roles of various cell types in tissue repair and summarizing the known dysfunctions of these cell types in diabetic foot ulcers. Recent advances in our understanding of the epigenetic regulation in immune cells identified to be altered in type 2 diabetes are summarized, and particular attention is paid to the developing research defining the epigenetic regulation of structural cells, including keratinocytes, fibroblasts, and endothelial cells. Gaps in knowledge are highlighted, and potential future directions are suggested based on the current state of the field.
Collapse
Affiliation(s)
- Tyler M. Bauer
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jadie Moon
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - James Shadiow
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Sam Buckley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Katherine A. Gallagher
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
2
|
Song J, Wu Y, Chen Y, Sun X, Zhang Z. Epigenetic regulatory mechanism of macrophage polarization in diabetic wound healing (Review). Mol Med Rep 2025; 31:2. [PMID: 39422035 PMCID: PMC11551531 DOI: 10.3892/mmr.2024.13367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Diabetic wounds represent a significant complication of diabetes and present a substantial challenge to global public health. Macrophages are crucial effector cells that play a pivotal role in the pathogenesis of diabetic wounds, through their polarization into distinct functional phenotypes. The field of epigenetics has emerged as a rapidly advancing research area, as this phenomenon has the potential to markedly affect gene expression, cellular differentiation, tissue development and susceptibility to disease. Understanding epigenetic mechanisms is crucial to further exploring disease pathogenesis. A growing body of scientific evidence has highlighted the pivotal role of epigenetics in the regulation of macrophage phenotypes. Various epigenetic mechanisms, such as DNA methylation, histone modification and non‑coding RNAs, are involved in the modulation of macrophage phenotype differentiation in response to the various environmental stimuli present in diabetic wounds. The present review provided an overview of the various changes that take place in macrophage phenotypes and functions within diabetic wounds and discussed the emerging role of epigenetic modifications in terms of regulating macrophage plasticity in diabetic wounds. It is hoped that this synthesis of information will facilitate the elucidation of diabetic wound pathogenesis and the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Jielin Song
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| | - Yuqing Wu
- The First Clinical Medical College, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yunli Chen
- The First Clinical Medical College, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xu Sun
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| | - Zhaohui Zhang
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300000, P.R. China
| |
Collapse
|
3
|
Kun L, Yunpeng D, Shuyuan F. Mechanism of DT-13 regulating macrophages in diabetic wound healing. Cell Signal 2024; 124:111446. [PMID: 39366531 DOI: 10.1016/j.cellsig.2024.111446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Diabetes is a complex metabolic system disease, and one of the main reasons why it is difficult to heal is that macrophages cannot realize the transition from pro-inflammatory M1 phenotype to anti-inflammatory M2 phenotype. Liriope spicata Lour. is a traditional Chinese medicine. According to the theory of traditional Chinese medicine, Liriope spicata Lour. has nourishing Yin Sheng Jin, moistening lung clear heart, used for lung dryness dry cough, Yin deficiency cough, throat arthralgia throat pain, thirst, internal heat thirst, upset insomnia, intestinal dryness constipation, is the classification of Yin tonifying drugs. Liriope muscari baily saponins C (DT-13) is one of the main active ingredients of Liriope spicata Lour., has significant anti-inflammatory, anti-tumor, and immunomodulatory effects. This thesis aims to explore the role of DT-13 in angiogenesis by regulating the polarization of macrophages, and ultimately play an anti-inflammatory role in regeneration by regulating immune cells. METHODS We conducted in vivo experiments using a diabetic mouse ulcer model to verify the effect of DT-13 in promoting wound healing. Spatial transcriptome sequencing technology was utilized to perform RNA transcript analysis on wound tissues from type II diabetic mice and non-diabetic mice. Subsequently, we used the CCK-8 assay to evaluate the impact of DT-13 on the viability of THP-1 cells (human monocytes). ELISA, immunofluorescence, and Western blot techniques were employed to study the mechanisms by which DT-13 inhibits the sustained inflammation and polarization process of M1 macrophages induced by LPS. The Transwell assay was used to assess the influence of DT-13 treatment on the co-culture of M1 macrophages induced by LPS under high glucose conditions with HUVEC cells. Finally, the chick embryo chorioallantoic membrane (CAM) assay was applied to explore the effects of DT-13 on angiogenesis stimulated by M1 macrophages under high glucose conditions with LPS stimulation. RESULTS We found that DT-13 can promote wound healing in a diabetic ulcer model in mice. Through spatial transcriptome sequencing results, we discovered that type II diabetic mice had higher levels of inflammation at the wound site and abnormal expression of macrophage characteristic proteins. The CCK-8 assay detected that DT-13 at 20 μmol/L had an effect on THP-1 cells. Through Q-PCR, ELISA, immunofluorescence, and Western blot results, we found that the mechanism by which DT-13 exerts anti-inflammatory effects on M1 macrophages with sustained inflammation induced by LPS under high glucose conditions may be through the TLR4-NFKB signaling pathway, and the mechanism for inducing the polarization of M1 macrophages to M2 type may be through the ERK-STAT3 signaling pathway. Interestingly, through the Transwell assay, we found that M1 macrophages induced by LPS under high glucose conditions, after treatment with DT-13 and co-cultured with HUVECs, could increase the migratory ability of HUVEC cells. This indicates that M1 macrophages induced by LPS under high glucose conditions, after treatment with DT-13, can promote angiogenesis. CONCLUSION DT-13 can significantly promote healing wounds in diabetic mice, and its mechanism may be to participate in promoting the formation of blood vessels by changing the polarization of macrophages.
Collapse
Affiliation(s)
- Li Kun
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China.
| | - Diao Yunpeng
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Fan Shuyuan
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China.
| |
Collapse
|
4
|
Franz S, Torregrossa M, Anderegg U, Ertel A, Saalbach A. Dysregulated S100A9 Expression Impairs Matrix Deposition in Chronic Wounds. Int J Mol Sci 2024; 25:9980. [PMID: 39337466 PMCID: PMC11432490 DOI: 10.3390/ijms25189980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Chronic non-healing wounds are characterized by persistent inflammation, excessive matrix-degrading proteolytic activity and compromised extracellular matrix (ECM) synthesis. Previous studies showed that S100A8/A9 are strongly dysregulated in delayed wound healing and impair the proper function of immune cells. Here, we demonstrate an unrecognized pathological function of S100A9 overexpression in wounds with impaired healing that directly affects ECM functions in fibroblasts. S100A9 was analyzed in two different mouse models mimicking the features of the two most prominent types of non-healing wounds in humans. Db/db mice were used as a model for diabetes-associated impaired wound healing. Iron-overloaded mice were used to mimic the conditions of impaired wound healing in chronic venous leg ulcers. The skin wounds of both mouse models are characterized by delayed wound closure, high and sustained expression of pro-inflammatory mediators and a substantially decreased ECM deposition, all together the hallmarks of non-healing wounds in humans. The wounds of both mouse models also present a solid and prolonged expression of S100A8 and S100A9 that coincides with a compromised ECM deposition and that was confirmed in chronic wounds in humans. Mechanistically, we reveal that S100A9 directly affects ECM deposition by shifting the balance of expression of ECM proteins and ECM degrading enzymes in fibroblasts via toll-like-receptor 4-dependent signaling. Consequently, blocking S100A9 during delayed wound healing in db/db mice restores fibroblast ECM functions eliciting increased matrix deposition. Our data indicate that the dysregulation of S100A9 directly contributes to a compromised ECM deposition in chronic wounds and further suggests S100A9 as a promising therapeutic target to improve tissue repair in chronic wounds.
Collapse
Affiliation(s)
| | | | | | | | - Anja Saalbach
- Department of Dermatology, Venereology and Allergology, Max Bürger Research Centre, Medical Faculty, University Leipzig, Johannisallee 30, 04103 Leipzig, Germany; (S.F.); (M.T.); (U.A.); (A.E.)
| |
Collapse
|
5
|
Wolf SJ, Audu CO, Moon JY, Joshi AD, Melvin WJ, Barrett EC, Mangum K, de Jimenez GS, Rocco S, Buckley S, Ahmed Z, Wasikowski R, Kahlenberg JM, Tsoi LC, Gudjonsson JE, Gallagher KA. Diabetic Wound Keratinocytes Induce Macrophage JMJD3-Mediated Nlrp3 Expression via IL-1R Signaling. Diabetes 2024; 73:1462-1472. [PMID: 38869447 PMCID: PMC11333374 DOI: 10.2337/db23-0968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/29/2024] [Indexed: 06/14/2024]
Abstract
Macrophage (Mφ) plasticity is critical for normal wound repair; however, in type 2 diabetic wounds, Mφs persist in a low-grade inflammatory state that prevents the resolution of wound inflammation. Increased NLRP3 inflammasome activity has been shown in diabetic wound Mφs; however, the molecular mechanisms regulating NLRP3 expression and activity are unclear. Here, we identified that diabetic wound keratinocytes induce Nlrp3 gene expression in wound Mφs through IL-1 receptor-mediated signaling, resulting in enhanced inflammasome activation in the presence of pathogen-associated molecular patterns and damage-associated molecular patterns. We found that IL-1α is increased in human and murine wound diabetic keratinocytes compared with nondiabetic controls and directly induces Mφ Nlrp3 expression through IL-1 receptor signaling. Mechanistically, we report that the histone demethylase, JMJD3, is increased in wound Mφs late post-injury and is induced by IL-1α from diabetic wound keratinocytes, resulting in Nlrp3 transcriptional activation through an H3K27me3-mediated mechanism. Using genetically engineered mice deficient in JMJD3 in myeloid cells (Jmjd3f/flyz2Cre+), we demonstrate that JMJD3 controls Mφ-mediated Nlrp3 expression during diabetic wound healing. Thus, our data suggest a role for keratinocyte-mediated IL-1α/IL-1R signaling in driving enhanced NLRP3 inflammasome activity in wound Mφs. These data also highlight the importance of cell cross-talk in wound tissues and identify JMJD3 and the IL-1R signaling cascade as important upstream therapeutic targets for Mφ NLRP3 inflammasome hyperactivity in nonhealing diabetic wounds. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Sonya J. Wolf
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Christopher O. Audu
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Jadie Y. Moon
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Amrita D. Joshi
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - William J. Melvin
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Emily C. Barrett
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Kevin Mangum
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | | | - Sabrina Rocco
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Sam Buckley
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Zara Ahmed
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | | | - J. Michelle Kahlenberg
- Department of Dermatology, University of Michigan, Ann Arbor, MI
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI
| | | | - Katherine A. Gallagher
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
6
|
Wang J, Feng J, Ni Y, Wang Y, Zhang T, Cao Y, Zhou M, Zhao C. Histone modifications and their roles in macrophage-mediated inflammation: a new target for diabetic wound healing. Front Immunol 2024; 15:1450440. [PMID: 39229271 PMCID: PMC11368794 DOI: 10.3389/fimmu.2024.1450440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024] Open
Abstract
Impaired wound healing is one of the main clinical complications of type 2 diabetes (T2D) and a major cause of lower limb amputation. Diabetic wounds exhibit a sustained inflammatory state, and reducing inflammation is crucial to diabetic wounds management. Macrophages are key regulators in wound healing, and their dysfunction would cause exacerbated inflammation and poor healing in diabetic wounds. Gene regulation caused by histone modifications can affect macrophage phenotype and function during diabetic wound healing. Recent studies have revealed that targeting histone-modifying enzymes in a local, macrophage-specific manner can reduce inflammatory responses and improve diabetic wound healing. This article will review the significance of macrophage phenotype and function in wound healing, as well as illustrate how histone modifications affect macrophage polarization in diabetic wounds. Targeting macrophage phenotype with histone-modifying enzymes may provide novel therapeutic strategies for the treatment of diabetic wound healing.
Collapse
Affiliation(s)
- Jing Wang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiawei Feng
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Ni
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing Wang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ting Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yemin Cao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingmei Zhou
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Zhao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
7
|
Apaza CJ, Cerezo JF, García-Tejedor A, Giménez-Bastida JA, Laparra-Llopis JM. Revisiting the Immunometabolic Basis for the Metabolic Syndrome from an Immunonutritional View. Biomedicines 2024; 12:1825. [PMID: 39200288 PMCID: PMC11352112 DOI: 10.3390/biomedicines12081825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/11/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Metabolic syndrome (MetS) implies different conditions where insulin resistance constitutes a major hallmark of the disease. The disease incurs a high risk for the development of cardiovascular complications, and takes its toll in regard to the gut-liver axis (pancreas, primary liver and colorectal)-associated immunity. The modulation of immunometabolic responses by immunonutritional factors (IFs) has emerged as a key determinant of the gut-liver axis' metabolic and immune health. IFs from plant seeds have shown in vitro and pre-clinical effectiveness primarily in dealing with various immunometabolic and inflammatory diseases. Only recently have immunonutritional studies established the engagement of innate intestinal immunity to effectively control immune alterations in inflamed livers preceding the major features of the MetS. However, integrative analyses and the demonstration of causality between IFs and specific gut-liver axis-associated immunometabolic imbalances for the MetS remain ill-defined in the field. Herein, a better understanding of the IFs with a significant role in the MetS, as well as within the dynamic interplay in the functional differentiation of innate immune key effectors (i.e., monocytes/macrophages), worsening or improving the disease, could be of crucial relevance. The development of an adequate intermediary phenotype of these cells can significantly contribute to maintaining the function of Tregs and innate lymphoid cells for the prevention and treatment of MetS and associated comorbidities.
Collapse
Affiliation(s)
- César Jeri Apaza
- Madrid Institute for Advanced Studies in Food (IMDEA Food), Carretera Cantoblanco 8, 28049 Madrid, Spain
- Bioactivity and Nutritional Immunology Group (BIOINUT), Valencian International University (VIU), Pintor Sorolla 21, 46002 Valencia, Spain
| | - Juan Francisco Cerezo
- Madrid Institute for Advanced Studies in Food (IMDEA Food), Carretera Cantoblanco 8, 28049 Madrid, Spain
| | - Aurora García-Tejedor
- Bioactivity and Nutritional Immunology Group (BIOINUT), Valencian International University (VIU), Pintor Sorolla 21, 46002 Valencia, Spain
| | - Juan Antonio Giménez-Bastida
- Research Group on Quality, Safety and Bioactivity of Plant Foods, Campus de Espinardo, CEBAS-CSIC, P.O. Box 164, 30100 Murcia, Spain;
| | - José Moisés Laparra-Llopis
- Madrid Institute for Advanced Studies in Food (IMDEA Food), Carretera Cantoblanco 8, 28049 Madrid, Spain
- Bioactivity and Nutritional Immunology Group (BIOINUT), Valencian International University (VIU), Pintor Sorolla 21, 46002 Valencia, Spain
| |
Collapse
|
8
|
Ju CC, Liu XX, Liu LH, Guo N, Guan LW, Wu JX, Liu DW. Epigenetic modification: A novel insight into diabetic wound healing. Heliyon 2024; 10:e28086. [PMID: 38533007 PMCID: PMC10963386 DOI: 10.1016/j.heliyon.2024.e28086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 03/04/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Wound healing is an intricate and fine regulatory process. In diabetic patients, advanced glycation end products (AGEs), excessive reactive oxygen species (ROS), biofilm formation, persistent inflammation, and angiogenesis regression contribute to delayed wound healing. Epigenetics, the fast-moving science in the 21st century, has been up to date and associated with diabetic wound repair. In this review, we go over the functions of epigenetics in diabetic wound repair in retrospect, covering transcriptional and posttranscriptional regulation. Among these, we found that histone modification is widely involved in inflammation and angiogenesis by affecting macrophages and endothelial cells. DNA methylation is involved in factors regulation in wound repair but also affects the differentiation phenotype of cells in hyperglycemia. In addition, noncodingRNA regulation and RNA modification in diabetic wound repair were also generalized. The future prospects for epigenetic applications are discussed in the end. In conclusion, the study suggests that epigenetics is an integral regulatory mechanism in diabetic wound healing.
Collapse
Affiliation(s)
- Cong-Cong Ju
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, PR China
- Huankui Academy, Nanchang University, Nanchang, Jiangxi, PR China
| | - Xiao-Xiao Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Li-hua Liu
- Huankui Academy, Nanchang University, Nanchang, Jiangxi, PR China
| | - Nan Guo
- Nanchang University, Nanchang, Jiangxi, PR China
| | - Le-wei Guan
- Huankui Academy, Nanchang University, Nanchang, Jiangxi, PR China
| | - Jun-xian Wu
- Nanchang University, Nanchang, Jiangxi, PR China
| | - De-Wu Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, PR China
| |
Collapse
|
9
|
Zhao Q, Lai K. Role of immune inflammation regulated by macrophage in the pathogenesis of age-related macular degeneration. Exp Eye Res 2024; 239:109770. [PMID: 38145794 DOI: 10.1016/j.exer.2023.109770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
Age-related macular degeneration (AMD) can lead to irreversible impairment of visual function, and the number of patients with AMD has been increasing globally. The immunoinflammatory theory is an important pathogenic mechanism of AMD, with macrophages serving as the primary inflammatory infiltrating cells in AMD lesions. Its powerful immunoinflammatory regulatory function has attracted considerable attention. Herein, we provide an overview of the involvement of macrophage-regulated immunoinflammation in different stages of AMD. Additionally, we summarize novel therapeutic approaches for AMD, focusing on targeting macrophages, such as macrophage/microglia modulators, reduction of macrophage aggregation in the subretinal space, modulation of macrophage effector function, macrophage phenotypic alterations, and novel biomimetic nanocomposites development based on macrophage-associated functional properties. We aimed to provide a basis and reference for the further exploration of AMD pathogenesis, developmental influences, and new therapeutic approaches.
Collapse
Affiliation(s)
- Qin Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, No.7 Jinsui Road, Guangzhou, 510060, China
| | - Kunbei Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, No.7 Jinsui Road, Guangzhou, 510060, China.
| |
Collapse
|
10
|
Lian X, Li Y, Wang W, Zuo J, Yu T, Wang L, Song L. The Modification of H3K4me3 Enhanced the Expression of CgTLR3 in Hemocytes to Increase CgIL17-1 Production in the Immune Priming of Crassostrea gigas. Int J Mol Sci 2024; 25:1036. [PMID: 38256110 PMCID: PMC10816183 DOI: 10.3390/ijms25021036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Increasing evidence confirms that histone modification plays a critical role in preserving long-term immunological memory. Immune priming is a novel form of immunological memory recently verified in invertebrates. Toll-like receptor (TLR) signaling and cytokines have been reported to be involved in the immune priming of the Pacific oyster Crassostrea gigas. In the present study, the expression of Toll-like receptor 3 (CgTLR3), myeloid differentiation factor 88-2 (CgMyd88-2) and interleukin 17-1 (CgIL17-1) was found to be elevated in the hemocytes of C. gigas at 6 h after the secondary stimulation with Vibrio splendidus, which was significantly higher than that at 6 h after the primary stimulation (p < 0.05). A significant increase in histone H3 lysine 4 trimethylation (H3K4me3) enrichment was detected in the promoter region of the CgTLR3 gene at 7 d after the primary stimulation with inactivated V. splendidus (p < 0.05). After the treatment with a histone methyltransferase inhibitor (5'-methylthioadenosine, MTA), the level of H3K4me3 at the promoter of the CgTLR3 gene decreased significantly at 7 d after the primary stimulation with inactivated V. splendidus (p < 0.05), and the expression of CgTLR3, CgMyD88-2 and CgIL17-1 was significantly repressed at 6 h after the secondary stimulation with V. splendidus (p < 0.05). Conversely, the treatment with monomethyl fumarate (MEF, an inhibitor of histone demethylases) resulted in a significant increase in H3K4me3 enrichment levels at the CgTLR3 promoter at 7 d after the primary stimulation (p < 0.05), and the expression of CgTLR3, CgMyD88-2 and CgIL17-1 was observed to increase significantly at 6 h after the secondary stimulation (p < 0.05). These results suggested that H3K4me3 regulated MyD88-dependent TLR signaling in the hemocytes of C. gigas, which defined the role of histone modifications in invertebrate immune priming.
Collapse
Affiliation(s)
- Xingye Lian
- School of Life Science, Liaoning Normal University, Dalian 116029, China; (X.L.); (Y.L.)
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Yinan Li
- School of Life Science, Liaoning Normal University, Dalian 116029, China; (X.L.); (Y.L.)
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Jiajun Zuo
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Tianqi Yu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; (W.W.); (J.Z.); (T.Y.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China
- Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
11
|
Awad AM, Elshaer SL, Gangaraju R, Abdelaziz RR, Nader MA. CysLTR1 antagonism by montelukast can ameliorate diabetes-induced aortic and testicular inflammation. Int Immunopharmacol 2023; 125:111127. [PMID: 37907048 DOI: 10.1016/j.intimp.2023.111127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/12/2023] [Accepted: 10/22/2023] [Indexed: 11/02/2023]
Abstract
AIMS Montelukast, a cysteinyl leukotriene receptor (CysLTR)1 antagonist, is emerging as an attractive strategy to curtail diabetic complications; however, its role in aortic and testicular tissues is unknown. This study investigated the effect of CysLTR1 antagonism by montelukast on toll-like receptor (TLR)4 and nuclear factor kappa B (NF-κB) pathways in diabetes-induced aortic and testicular injury. METHODS Adult male Sprague-Dawley rats were made diabetic with Streptozotocin (STZ, 55 mg/kg). Following this, Streptozotocin-induced diabetic (SD) rats were administered montelukast (10 and 20 mg/kg, orally) for 8 weeks. Blood glucose, serum malondialdehyde (MDA), inflammatory markers, and histopathology were evaluated. RESULTS Montelukast showed protection against diabetic complications, as evidenced by the ameliorative effect against STZ-induced alterations in oxidative stress as indicated by serum MDA levels. Moreover, montelukast conferred a significant decrease in the aortic and testicular levels of CysLTR1, TLR4, and NF-κB with a subsequent decrease in the levels of NOD-like receptor family pyrin domain containing (NLRP)3, caspase 1, interleukin (IL)-1β, IL-6, monocyte chemoattractant protein (MCP)-1, and tumor necrosis factor (TNF)-α. Additionally, administration of montelukast resulted in autophagy stimulation, as shown by decreased p62/Sequestosome (SQSTM)1 levels. Finally, montelukast protection resulted in normal thickness of whole aortic wall, regular tunica (t.) intima, mild vacuolation of smooth muscle fibers in aorta, increased size of seminiferous tubules, and increased spermatogenesis in testis as demonstrated by histopathology. CONCLUSIONS The protective effect of montelukast against diabetes-induced aortic and testicular injury is due to its antioxidant, anti-inflammatory, and autophagy stimulation characteristics.
Collapse
Affiliation(s)
- Ahmed M Awad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt; Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sally L Elshaer
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt
| |
Collapse
|
12
|
Geng K, Ma X, Jiang Z, Huang W, Gu J, Wang P, Luo L, Xu Y, Xu Y. High glucose-induced STING activation inhibits diabetic wound healing through promoting M1 polarization of macrophages. Cell Death Discov 2023; 9:136. [PMID: 37100799 PMCID: PMC10133226 DOI: 10.1038/s41420-023-01425-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/22/2023] [Accepted: 04/03/2023] [Indexed: 04/28/2023] Open
Abstract
Diabetic wound (DW) is characterized by elevated pro-inflammatory cytokines and cellular dysfunction consistent with elevated reactive oxygen species (ROS) levels. Recent advances in immunology have dissected molecular pathways involved in the innate immune system where cytoplasmic DNA can trigger STING-dependent inflammatory responses and play an important role in metabolic-related diseases. We investigated whether STING regulates inflammation and cellular dysfunction in DW healing. We found that STING and M1 macrophages were increased in wound tissues from DW in patients and mice and delayed the wound closure. We also noticed that the massively released ROS in the High glucose (HG) environment activated STING signaling by inducing the escape of mtDNA to the cytoplasm, inducing macrophage polarization into a pro-inflammatory phenotype, releasing pro-inflammatory cytokines, and exacerbating endothelial cell dysfunction. In Conclusion, mtDNA-cGAS-STING pathway activation under diabetic metabolic stress is an important mechanism of DW refractory healing. While using STING gene-edited macrophages for wound treatment by cell therapy can induce the polarization of wound macrophages from pro-inflammatory M1 to anti-inflammatory M2, promote angiogenesis, and collagen deposition to accelerate DW healing. STING may be a promising therapeutic target for DW.
Collapse
Affiliation(s)
- Kang Geng
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao
- Department of Endocrinology and Metabolism, Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Nephropathy, Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, PR China
- Department of plastic and burns surgery, National Key Clinical Construction Specialty, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, PR China
| | - Xiumei Ma
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao
- Department of Endocrinology and Metabolism, Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Nephropathy, Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, PR China
| | - Zongzhe Jiang
- Department of Endocrinology and Metabolism, Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Nephropathy, Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, PR China
| | - Wei Huang
- Department of Endocrinology and Metabolism, Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Nephropathy, Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, PR China
| | - Junling Gu
- Endocrinology Department, The Second People's Hospital of Yibin‧West China Yibin Hospital, Sichuan University, Yibin, Sichuan, PR China
| | - Peng Wang
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao
- Department of Endocrinology and Metabolism, Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Nephropathy, Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, PR China
| | - Lifang Luo
- Department of Endocrinology and Metabolism, Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Nephropathy, Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, PR China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao.
| | - Yong Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao.
- Department of Endocrinology and Metabolism, Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Nephropathy, Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, Sichuan, PR China.
| |
Collapse
|
13
|
Obi AT, Sharma SB, Elfline MA, Luke CE, Dowling AR, Cai Q, Kimball AS, Hollinstat M, Stanger L, Moore BB, Jaffer FA, Henke PK. Experimental venous thrombus resolution is driven by IL-6 mediated monocyte actions. Sci Rep 2023; 13:3253. [PMID: 36828892 PMCID: PMC9951841 DOI: 10.1038/s41598-023-30149-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/16/2023] [Indexed: 02/26/2023] Open
Abstract
Deep venous thrombosis and residual thrombus burden correlates with circulating IL-6 levels in humans. To investigate the cellular source and role of IL-6 in thrombus resolution, Wild type C57BL/6J (WT), and IL-6-/- mice underwent induction of VT via inferior vena cava (IVC) stenosis or stasis. Vein wall (VW) and thrombus were analyzed by western blot, immunohistochemistry, and flow cytometry. Adoptive transfer of WT bone marrow derived monocytes was performed into IL6-/- mice to assess for rescue. Cultured BMDMs from WT and IL-6-/- mice underwent quantitative real time PCR and immunoblotting for fibrinolytic factors and matrix metalloproteinase activity. No differences in baseline coagulation function or platelet function were found between WT and IL-6-/- mice. VW and thrombus IL-6 and IL-6 leukocyte-specific receptor CD126 were elevated in a time-dependent fashion in both VT models. Ly6Clo Mo/MØ were the predominant leukocyte source of IL-6. IL-6-/- mice demonstrated larger, non-resolving stasis thrombi with less neovascularization, despite a similar number of monocytes/macrophages (Mo/MØ). Adoptive transfer of WT BMDM into IL-6-/- mice undergoing stasis VT resulted in phenotype rescue. Human specimens of endophlebectomized tissue showed co-staining of Monocyte and IL-6 receptor. Thrombosis matrix analysis revealed significantly increased thrombus fibronectin and collagen in IL-6-/- mice. MMP9 activity in vitro depended on endogenous IL-6 expression in Mo/MØ, and IL-6-/- mice exhibited stunted matrix metalloproteinase activity. Lack of IL-6 signaling impairs thrombus resolution potentially via dysregulation of MMP-9 leading to impaired thrombus recanalization and resolution. Restoring or augmenting monocyte-mediated IL-6 signaling in IL-6 deficient or normal subjects, respectively, may represent a non-anticoagulant target to improve thrombus resolution.
Collapse
Affiliation(s)
- Andrea T Obi
- Conrad Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, USA.
- University of Michigan Health System, 1500 E. Medical Center Drive, Cardiovascular Center - 5463, Ann Arbor, MI, 48109-5867, USA.
| | - Sriganesh B Sharma
- Conrad Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, USA
| | - Megan A Elfline
- Conrad Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, USA
| | - Catherine E Luke
- Conrad Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, USA
| | - Abigail R Dowling
- Conrad Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, USA
| | - Qing Cai
- Conrad Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, USA
| | - Andrew S Kimball
- Section of Vascular Surgery, University of Alabama Division of Vascular Surgery, University of Michigan Medical School, Ann Arbor, USA
| | - Mike Hollinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, USA
| | - Livia Stanger
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, USA
| | - Bethany B Moore
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, USA
- Cardiovascular Research Center, Cardiology Division, Department of Medicine, University of Michigan Medical School, Ann Arbor, USA
| | - Farouc A Jaffer
- Section of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Peter K Henke
- Conrad Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, USA
| |
Collapse
|
14
|
Sharma SB, Melvin WJ, Audu CO, Bame M, Rhoads N, Wu W, Kanthi Y, Knight JS, Adili R, Holinstat MA, Wakefield TW, Henke PK, Moore BB, Gallagher KA, Obi AT. The histone methyltransferase MLL1/KMT2A in monocytes drives coronavirus-associated coagulopathy and inflammation. Blood 2023; 141:725-742. [PMID: 36493338 PMCID: PMC9743412 DOI: 10.1182/blood.2022015917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
Coronavirus-associated coagulopathy (CAC) is a morbid and lethal sequela of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. CAC results from a perturbed balance between coagulation and fibrinolysis and occurs in conjunction with exaggerated activation of monocytes/macrophages (MO/Mφs), and the mechanisms that collectively govern this phenotype seen in CAC remain unclear. Here, using experimental models that use the murine betacoronavirus MHVA59, a well-established model of SARS-CoV-2 infection, we identify that the histone methyltransferase mixed lineage leukemia 1 (MLL1/KMT2A) is an important regulator of MO/Mφ expression of procoagulant and profibrinolytic factors such as tissue factor (F3; TF), urokinase (PLAU), and urokinase receptor (PLAUR) (herein, "coagulopathy-related factors") in noninfected and infected cells. We show that MLL1 concurrently promotes the expression of the proinflammatory cytokines while suppressing the expression of interferon alfa (IFN-α), a well-known inducer of TF and PLAUR. Using in vitro models, we identify MLL1-dependent NF-κB/RelA-mediated transcription of these coagulation-related factors and identify a context-dependent, MLL1-independent role for RelA in the expression of these factors in vivo. As functional correlates for these findings, we demonstrate that the inflammatory, procoagulant, and profibrinolytic phenotypes seen in vivo after coronavirus infection were MLL1-dependent despite blunted Ifna induction in MO/Mφs. Finally, in an analysis of SARS-CoV-2 positive human samples, we identify differential upregulation of MLL1 and coagulopathy-related factor expression and activity in CD14+ MO/Mφs relative to noninfected and healthy controls. We also observed elevated plasma PLAU and TF activity in COVID-positive samples. Collectively, these findings highlight an important role for MO/Mφ MLL1 in promoting CAC and inflammation.
Collapse
Affiliation(s)
- Sriganesh B. Sharma
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - William J. Melvin
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Christopher O. Audu
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Monica Bame
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI
| | - Nicole Rhoads
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Weisheng Wu
- Bioinformatics Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, MI
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis & Inflammation, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Michael A. Holinstat
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Thomas W. Wakefield
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Peter K. Henke
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI
| | - Katherine A. Gallagher
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI
| | - Andrea T. Obi
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| |
Collapse
|
15
|
Zeng F, Zheng J, Shen L, Herrera-Balandrano DD, Huang W, Sui Z. Physiological mechanisms of TLR4 in glucolipid metabolism regulation: Potential use in metabolic syndrome prevention. Nutr Metab Cardiovasc Dis 2023; 33:38-46. [PMID: 36428186 DOI: 10.1016/j.numecd.2022.10.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/30/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
Over-nourishment or an unbalanced diet has been linked to an increase in the prevalence of metabolic syndrome. An imbalance in glucolipid metabolism is a major cause of metabolic syndrome, which has consequences for human health. Toll-like receptor 4 (TLR4), a member of the innate immune pattern recognition receptor family, is involved in inflammation-related disorders, autoimmune diseases, and tumors. Recent research has shown that TLR4 plays a key role in glucolipid metabolism, which is linked to insulin resistance, intestinal flora, and the development of chronic inflammation. TLR4 activation regulates glucolipid metabolism and contributes to the dynamic relationship between innate immunity and nutrition-related disorders. Further, TLR4 regulates glucolipid metabolism by controlling glycolysis and pyruvate oxidative decarboxylation, interfering with insulin signaling, regulating adipogenic gene expression levels, influencing preadipocyte differentiation and lipid accumulation, and altering the intestinal microbiota and permeability. TLR4 functions may provide new therapeutic applications for the prevention and treatment of metabolic syndrome. The purpose of this review is to enrich mechanistic research of diabetes, atherosclerosis, and other nutrition-related disorders by summarizing the role of TLR4 in the regulation of glucolipid metabolism as well as its physiological mechanisms.
Collapse
Affiliation(s)
- Feng Zeng
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, PR China; Medical College, Yangzhou University, Yangzhou 225000, PR China; Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Jiawei Zheng
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, PR China; School of Food & Bioengineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Li Shen
- Medical College, Yangzhou University, Yangzhou 225000, PR China
| | | | - Wuyang Huang
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, PR China; School of Food & Bioengineering, Jiangsu University, Zhenjiang 212013, PR China.
| | - Zhongquan Sui
- Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| |
Collapse
|
16
|
Worsley AL, Lui DH, Ntow-Boahene W, Song W, Good L, Tsui J. The importance of inflammation control for the treatment of chronic diabetic wounds. Int Wound J 2022. [PMID: 36564054 DOI: 10.1111/iwj.14048] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/25/2022] Open
Abstract
Diabetic chronic wounds cause massive levels of patient suffering and economic problems worldwide. The state of chronic inflammation arises in response to a complex combination of diabetes mellitus-related pathophysiologies. Advanced treatment options are available; however, many wounds still fail to heal, exacerbating morbidity and mortality. This review describes the chronic inflammation pathophysiologies in diabetic ulcers and treatment options that may help address this dysfunction either directly or indirectly. We suggest that treatments to reduce inflammation within these complex wounds may help trigger healing.
Collapse
Affiliation(s)
- Anna L Worsley
- Royal Veterinary College, Department of Pathobiology and Population Sciences, London, UK.,UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, UCL Division of Surgery and Interventional Science, University College London, London, UK
| | - Dennis H Lui
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, UCL Division of Surgery and Interventional Science, University College London, London, UK
| | - Winnie Ntow-Boahene
- Royal Veterinary College, Department of Pathobiology and Population Sciences, London, UK.,UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, UCL Division of Surgery and Interventional Science, University College London, London, UK
| | - Wenhui Song
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, UCL Division of Surgery and Interventional Science, University College London, London, UK
| | - Liam Good
- Royal Veterinary College, Department of Pathobiology and Population Sciences, London, UK
| | - Janice Tsui
- UCL Centre for Biomaterials in Surgical Reconstruction and Regeneration, Department of Surgical Biotechnology, UCL Division of Surgery and Interventional Science, University College London, London, UK
| |
Collapse
|
17
|
Hassanshahi A, Moradzad M, Ghalamkari S, Fadaei M, Cowin AJ, Hassanshahi M. Macrophage-Mediated Inflammation in Skin Wound Healing. Cells 2022; 11:cells11192953. [PMID: 36230913 PMCID: PMC9564023 DOI: 10.3390/cells11192953] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/11/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages are key immune cells that respond to infections, and modulate pathophysiological conditions such as wound healing. By possessing phagocytic activities and through the secretion of cytokines and growth factors, macrophages are pivotal orchestrators of inflammation, fibrosis, and wound repair. Macrophages orchestrate the process of wound healing through the transitioning from predominantly pro-inflammatory (M1-like phenotypes), which present early post-injury, to anti-inflammatory (M2-like phenotypes), which appear later to modulate skin repair and wound closure. In this review, different cellular and molecular aspects of macrophage-mediated skin wound healing are discussed, alongside important aspects such as macrophage subtypes, metabolism, plasticity, and epigenetics. We also highlight previous studies demonstrating interactions between macrophages and these factors for optimal wound healing. Understanding and harnessing the activity and capability of macrophages may help to advance new approaches for improving healing of the skin.
Collapse
Affiliation(s)
- Alireza Hassanshahi
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, SA 5095, Australia
| | - Mohammad Moradzad
- Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj 66179-13446, Iran
| | - Saman Ghalamkari
- Department of Biology, Islamic Azad University, Arsanjan 61349-37333, Iran
| | - Moosa Fadaei
- Department of Biology, Islamic Azad University, Arsanjan 61349-37333, Iran
| | - Allison J. Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, SA 5095, Australia
- Correspondence: (A.J.C.); (M.H.)
| | - Mohammadhossein Hassanshahi
- Vascular Research Centre, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, SA 5005, Australia
- Correspondence: (A.J.C.); (M.H.)
| |
Collapse
|
18
|
Yu H, Wang Y, Wang D, Yi Y, Liu Z, Wu M, Wu Y, Zhang Q. Landscape of the epigenetic regulation in wound healing. Front Physiol 2022; 13:949498. [PMID: 36035490 PMCID: PMC9403478 DOI: 10.3389/fphys.2022.949498] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 07/19/2022] [Indexed: 12/13/2022] Open
Abstract
Wound healing after skin injury is a dynamic and highly coordinated process involving a well-orchestrated series of phases, including hemostasis, inflammation, proliferation, and tissue remodeling. Epigenetic regulation refers to genome-wide molecular events, including DNA methylation, histone modification, and non-coding RNA regulation, represented by microRNA (miRNA), long noncoding RNA (lncRNA), and circular RNA (circRNA). Epigenetic regulation is pervasively occurred in the genome and emerges as a new role in gene expression at the post-transcriptional level. Currently, it is well-recognized that epigenetic factors are determinants in regulating gene expression patterns, and may provide evolutionary mechanisms that influence the wound microenvironments and the entire healing course. Therefore, this review aims to comprehensively summarize the emerging roles and mechanisms of epigenetic remodeling in wound healing. Moreover, we also pose the challenges and future perspectives related to epigenetic modifications in wound healing, which would bring novel insights to accelerated wound healing.
Collapse
Affiliation(s)
| | | | | | | | | | - Min Wu
- *Correspondence: Min Wu, ; Yiping Wu, ; Qi Zhang,
| | - Yiping Wu
- *Correspondence: Min Wu, ; Yiping Wu, ; Qi Zhang,
| | - Qi Zhang
- *Correspondence: Min Wu, ; Yiping Wu, ; Qi Zhang,
| |
Collapse
|
19
|
Al Sadoun H. Macrophage Phenotypes in Normal and Diabetic Wound Healing and Therapeutic Interventions. Cells 2022; 11:2430. [PMID: 35954275 PMCID: PMC9367932 DOI: 10.3390/cells11152430] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/30/2022] [Accepted: 08/02/2022] [Indexed: 11/29/2022] Open
Abstract
Macrophage differentiation and polarization are essential players in the success of the wound-healing process. Acute simple wounds progress from inflammation to proliferation/regeneration and, finally, to remodeling. In injured skin, macrophages either reside in the epithelium or are recruited from monocytes. Their main role is supported by their plasticity, which allows them to adopt different phenotypic states, such as the M1-inflammatory state, in which they produce TNF and NO, and the M2-reparative state, in which they resolve inflammation and exhibit a reparative function. Reparative macrophages are an essential source of growth factors such as TGF-β and VEGF and are not found in nonhealing wounds. This review discusses the differences between macrophage phenotypes in vitro and in vivo, how macrophages originate, and how they cross-communicate with other cellular components in a wound. This review also highlights the dysregulation of macrophages that occurs in nonhealing versus overhealing wounds and fibrosis. Then, the therapeutic manipulation of macrophages is presented as an attractive strategy for promoting healing through the secretion of growth factors for angiogenesis, keratinocyte migration, and collagen production. Finally, Hoxa3 overexpression is discussed as an example of the therapeutic repolarization of macrophages to the normal maturation state and phenotype with better healing outcomes.
Collapse
Affiliation(s)
- Hadeel Al Sadoun
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; ; Tel.: +966-(12)-6400000 (ext. 24277)
- Stem Cell Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
20
|
Mills SJ, Ahangar P, Thomas HM, Hofma BR, Murray RZ, Cowin AJ. Flightless I Negatively Regulates Macrophage Surface TLR4, Delays Early Inflammation, and Impedes Wound Healing. Cells 2022; 11:cells11142192. [PMID: 35883634 PMCID: PMC9318993 DOI: 10.3390/cells11142192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 01/27/2023] Open
Abstract
TLR4 plays a pivotal role in orchestrating inflammation and tissue repair. Its expression has finally been balanced to initiate the early, robust immune response necessary for efficient repair without excessively amplifying and prolonging inflammation, which impairs healing. Studies show Flightless I (Flii) is an immunomodulator that negatively regulates macrophage TLR4 signalling. Using macrophages from Flii+/−, WT, and FliiTg/Tg mice, we have shown that elevated Flii reduces early TLR4 surface expression, delaying and reducing subsequent TNF secretions. In contrast, reduced Flii increases surface TLR4, leading to an earlier robust TNF peak. In Flii+/− mice, TLR4 levels peak earlier during wound repair, and overall healing is accelerated. Fewer neutrophils, monocytes and macrophages are recruited to Flii+/− wounds, leading to fewer TNF-positive macrophages, alongside an early peak and a robust shift to M2 anti-inflammatory, reparative Ym1+ and IL-10+ macrophages. Importantly, in diabetic mice, high Flii levels are found in plasma and unwounded skin, with further increases observed in their wounds, which have impaired healing. Lowering Flii in diabetic mice results in an earlier shift to M2 macrophages and improved healing. Overall, this suggests Flii regulation of TLR4 reduces early inflammation and decreases the M2 macrophage phenotype, leading to impaired healing.
Collapse
Affiliation(s)
- Stuart J. Mills
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide SA 5095, Australia; (P.A.); (H.M.T.); (B.R.H.)
- Correspondence: (S.J.M.); (A.J.C.); Tel.: +61-8-8302-3896 (S.J.M.)
| | - Parinaz Ahangar
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide SA 5095, Australia; (P.A.); (H.M.T.); (B.R.H.)
| | - Hannah M. Thomas
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide SA 5095, Australia; (P.A.); (H.M.T.); (B.R.H.)
| | - Benjamin R. Hofma
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide SA 5095, Australia; (P.A.); (H.M.T.); (B.R.H.)
| | - Rachael Z. Murray
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane QLD 4059, Australia;
| | - Allison J. Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide SA 5095, Australia; (P.A.); (H.M.T.); (B.R.H.)
- Correspondence: (S.J.M.); (A.J.C.); Tel.: +61-8-8302-3896 (S.J.M.)
| |
Collapse
|
21
|
Wolf SJ, Melvin WJ, Gallagher K. Macrophage-mediated inflammation in diabetic wound repair. Semin Cell Dev Biol 2021; 119:111-118. [PMID: 34183242 PMCID: PMC8985699 DOI: 10.1016/j.semcdb.2021.06.013] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 06/07/2021] [Accepted: 06/18/2021] [Indexed: 02/08/2023]
Abstract
Non-healing wounds in Type 2 Diabetes (T2D) patients represent the most common cause of amputation in the US, with an associated 5-year mortality of nearly 50%. Our lab has examined tissue from both T2D murine models and human wounds in order to explore mechanisms contributing to impaired wound healing. Current published data in the field point to macrophage function serving a pivotal role in orchestrating appropriate wound healing. Wound macrophages in mice and patients with T2D are characterized by a persistent inflammatory state; however, the mechanisms that control this persistent inflammatory state are unknown. Current literature demonstrates that gene regulation through histone modifications, DNA modifications, and microRNA can influence macrophage plasticity during wound healing. Further, accumulating studies reveal the importance of cells such as adipocytes, infiltrating immune cells (PMNs and T cells), and keratinocytes secrete factors that may help drive macrophage polarization. This review will examine the role of macrophages in the wound healing process, along with their function and interactions with other cells, and how it is perturbed in T2D. We also explore epigenetic factors that regulate macrophage polarization in wounds, while highlighting the emerging role of other cell types that may influence macrophage phenotype following tissue injury.
Collapse
Affiliation(s)
- Sonya J. Wolf
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - William J. Melvin
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Katherine Gallagher
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA,Correspondence to: Department of Surgery, University of Michigan, 1500 East Medical Center Drive, SPC 5867, Ann Arbor, MI 48109, USA. (K. Gallagher)
| |
Collapse
|
22
|
Geng K, Ma X, Jiang Z, Huang W, Gao C, Pu Y, Luo L, Xu Y, Xu Y. Innate Immunity in Diabetic Wound Healing: Focus on the Mastermind Hidden in Chronic Inflammatory. Front Pharmacol 2021; 12:653940. [PMID: 33967796 PMCID: PMC8097165 DOI: 10.3389/fphar.2021.653940] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
A growing body of evidence suggests that the interaction between immune and metabolic responses is essential for maintaining tissue and organ homeostasis. These interacting disorders contribute to the development of chronic diseases associated with immune-aging such as diabetes, obesity, atherosclerosis, and nonalcoholic fatty liver disease. In Diabetic wound (DW), innate immune cells respond to the Pathogen-associated molecular patterns (PAMAs) and/or Damage-associated molecular patterns (DAMPs), changes from resting to an active phenotype, and play an important role in the triggering and maintenance of inflammation. Furthermore, the abnormal activation of innate immune pathways secondary to immune-aging also plays a key role in DW healing. Here, we review studies of innate immune cellular molecular events that identify metabolic disorders in the local microenvironment of DW and provide a historical perspective. At the same time, we describe some of the recent progress, such as TLR receptor-mediated intracellular signaling pathways that lead to the activation of NF-κB and the production of various pro-inflammatory mediators, NLRP3 inflammatory via pyroptosis, induction of IL-1β and IL-18, cGAS-STING responds to mitochondrial injury and endoplasmic reticulum stress, links sensing of metabolic stress to activation of pro-inflammatory cascades. Besides, JAK-STAT is also involved in DW healing by mediating the action of various innate immune effectors. Finally, we discuss the great potential of targeting these innate immune pathways and reprogramming innate immune cell phenotypes in DW therapy.
Collapse
Affiliation(s)
- Kang Geng
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China.,Department of Plastic and Burn Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,National Key Clinical Construction Specialty, Luzhou, China
| | - Xiumei Ma
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Zongzhe Jiang
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Wei Huang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Chenlin Gao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Yueli Pu
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Lifang Luo
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China
| | - Yong Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, China.,State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Avenida Wai Long, Taipa, China.,Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, China.,Sichuan Clinical Research Center for Nephropathy, Luzhou, China.,Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
23
|
Abstract
The innate immune response is a rapid response to pathogens or danger signals. It is precisely activated not only to efficiently eliminate pathogens but also to avoid excessive inflammation and tissue damage. cis-Regulatory element-associated chromatin architecture shaped by epigenetic factors, which we define as the epiregulome, endows innate immune cells with specialized phenotypes and unique functions by establishing cell-specific gene expression patterns, and it also contributes to resolution of the inflammatory response. In this review, we focus on two aspects: (a) how niche signals during lineage commitment or following infection and pathogenic stress program epiregulomes by regulating gene expression levels, enzymatic activities, or gene-specific targeting of chromatin modifiers and (b) how the programed epiregulomes in turn mediate regulation of gene-specific expression, which contributes to controlling the development of innate cells, or the response to infection and inflammation, in a timely manner. We also discuss the effects of innate immunometabolic rewiring on epiregulomes and speculate on several future challenges to be encountered during the exploration of the master regulators of epiregulomes in innate immunity and inflammation.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China; , .,National Key Laboratory of Medical Immunology, Institute of Immunology, Navy Military Medical University, Shanghai 200433, China
| | - Xuetao Cao
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China; , .,National Key Laboratory of Medical Immunology, Institute of Immunology, Navy Military Medical University, Shanghai 200433, China.,Laboratory of Immunity and Inflammation, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
24
|
Huizinga GP, Singer BH, Singer K. The Collision of Meta-Inflammation and SARS-CoV-2 Pandemic Infection. Endocrinology 2020; 161:bqaa154. [PMID: 32880654 PMCID: PMC7499583 DOI: 10.1210/endocr/bqaa154] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has forced us to consider the physiologic role of obesity in the response to infectious disease. There are significant disparities in morbidity and mortality by sex, weight, and diabetes status. Numerous endocrine changes might drive these varied responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, including hormone and immune mediators, hyperglycemia, leukocyte responses, cytokine secretion, and tissue dysfunction. Studies of patients with severe COVID-19 disease have revealed the importance of innate immune responses in driving immunopathology and tissue injury. In this review we will describe the impact of the metabolically induced inflammation (meta-inflammation) that characterizes obesity on innate immunity. We consider that obesity-driven dysregulation of innate immune responses may drive organ injury in the development of severe COVID-19 and impair viral clearance.
Collapse
Affiliation(s)
- Gabrielle P Huizinga
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Benjamin H Singer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
- Michigan Center for Integrative Research in Critical Care, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kanakadurga Singer
- Department of Pediatrics and Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
25
|
Davis FM, denDekker A, Joshi AD, Wolf SJ, Audu C, Melvin WJ, Mangum K, Riordan MO, Kunkel SL, Gallagher KA. Palmitate-TLR4 signaling regulates the histone demethylase, JMJD3, in macrophages and impairs diabetic wound healing. Eur J Immunol 2020; 50:1929-1940. [PMID: 32662520 DOI: 10.1002/eji.202048651] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/30/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022]
Abstract
Chronic macrophage inflammation is a hallmark of type 2 diabetes (T2D) and linked to the development of secondary diabetic complications. T2D is characterized by excess concentrations of saturated fatty acids (SFA) that activate innate immune inflammatory responses, however, mechanism(s) by which SFAs control inflammation is unknown. Using monocyte-macrophages isolated from human blood and murine models, we demonstrate that palmitate (C16:0), the most abundant circulating SFA in T2D, increases expression of the histone demethylase, Jmjd3. Upregulation of Jmjd3 results in removal of the repressive histone methylation (H3K27me3) mark on NFκB-mediated inflammatory gene promoters driving macrophage-mediated inflammation. We identify that the effects of palmitate are fatty acid specific, as laurate (C12:0) does not regulate Jmjd3 and the associated inflammatory profile. Further, palmitate-induced Jmjd3 expression is controlled via TLR4/MyD88-dependent signaling mechanism, where genetic depletion of TLR4 (Tlr4-/- ) or MyD88 (MyD88-/- ) negated the palmitate-induced changes in Jmjd3 and downstream NFκB-induced inflammation. Pharmacological inhibition of Jmjd3 using a small molecule inhibitor (GSK-J4) reduced macrophage inflammation and improved diabetic wound healing. Together, we conclude that palmitate contributes to the chronic Jmjd3-mediated activation of macrophages in diabetic peripheral tissue and a histone demethylase inhibitor-based therapy may represent a novel treatment for nonhealing diabetic wounds.
Collapse
Affiliation(s)
- Frank M Davis
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Aaron denDekker
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Amrita D Joshi
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Sonya J Wolf
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Christopher Audu
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - William J Melvin
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kevin Mangum
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Mary O Riordan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Steven L Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Katherine A Gallagher
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA.,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|