1
|
Hao H, Hu J, Kuai Z, Hao F, Jiang W, Ran N, He Y, Zhang Y, Huang Y, Qi Y, Luo Q. Enzyme-mediated multifunctional self-healing lysozyme hydrogel for synergistic treatment of chronic diabetic wounds. Int J Biol Macromol 2024; 282:136719. [PMID: 39437956 DOI: 10.1016/j.ijbiomac.2024.136719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Self-healing hydrogels have attracted significant attention in chronic diabetic wound healing due to their potential to minimize the risk of secondary infections caused by joint movement or dressing rupture. Herein, a multifunctional self-healing hydrogel mediated utilizing an enzyme-triggered cascade reaction based on dynamic imine bonds was designed. The hydrogel employs three enzymes: lysozyme (LYZ), glucose oxidase (GOx), and catalase (CAT), as building blocks. GOx catalyzes the conversion of glucose and 1-thio-β-d-glucose (β-GlcSH) into hydrogen peroxide (H2O2), gluconic acid (GA), and hydrogen sulfide (H2S). Subsequently, CAT eliminates H2O2, protecting the imine bonds from oxidative damage. The acidic environment created by GA decreases the pH and regulates the crosslinking density of imine bonds, enhancing the self-healing capability and porosity of the hydrogel. This feature enables the sustained release of the drug rosuvastatin calcium (RCa) to promote endothelial cell migration and vascular regeneration. Combined with the antioxidative and anti-inflammatory effects of released H2S gas and the antibacterial properties of lysozyme, this hydrogel exhibits promising therapeutic efficacy for the synergistic treatment of chronic diabetic wounds.
Collapse
Affiliation(s)
- Hao Hao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Juntao Hu
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin 130033, China
| | - Ziyu Kuai
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin 130033, China
| | - Fengjie Hao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wantong Jiang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Nana Ran
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yuting He
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yanping Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yibing Huang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yanfei Qi
- Centenary Institute of Cancer Medicine and Cell Biology, The University of Sydney, Camperdown, NSW 2050, Australia.
| | - Quan Luo
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China; Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| |
Collapse
|
2
|
Li Y, Xia Y, Liu X, Wang J, Sun Y, Huang J, Guo Z, Jia S, Chen Y, Wang J, Wang L, Li J, Feng J, Wang L, Li X. Rational design of bioengineered recombinant collagen-like protein enhances GelMA hydrogel for diabetic wound healing. Int J Biol Macromol 2024; 280:136012. [PMID: 39326607 DOI: 10.1016/j.ijbiomac.2024.136012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/08/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Gelatin methacryloyl (GelMA) holds significant potential in tissue engineering; however, its clinical applications are often constrained by its lack of functional groups. To overcome this limitation, recombinant proteins with multiple biofunctional domains present a promising strategy for GelMA functionalization, enhancing its biological properties. In this study, we developed a rationally designed recombinant collagen-like protein (RC) engineered with multiple biofunctional domains, which demonstrated the ability to upregulate collagen 1α (COL-1α) expression in NIH-3 T3 cells. By utilizing EDC/NHS chemistry, the purified RC was conjugated to GelMA, resulting in GelMA-RC hydrogels that significantly improved cell viability and migration compared to unmodified GelMA. Subsequent in vivo studies showed that RC-modified GelMA exhibited superior wound healing efficacy, largely attributed to enhanced expression of cytokeratin-14 (CK-14) and COL-1α. These findings underscore the potential of RC-functionalized GelMA in promoting diabetic wound repair and suggest broader applicability for functionalizing other biomaterials.
Collapse
Affiliation(s)
- Yimiao Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Yan Xia
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Xing Liu
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Jieqi Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Yinan Sun
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Jinxia Huang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Zhao Guo
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Shuang Jia
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Yufang Chen
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Jie Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Liping Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Jiaqi Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Jian Feng
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Liyao Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China
| | - Xinyu Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, People's Republic of China.
| |
Collapse
|
3
|
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
|
4
|
Reynolds MB, Klein B, McFadden MJ, Judge NK, Navarrete HE, Michmerhuizen BC, Awad D, Schultz TL, Harms PW, Zhang L, O'Meara TR, Sexton JZ, Lyssiotis CA, Kahlenberg JM, O'Riordan MX. Type I interferon governs immunometabolic checkpoints that coordinate inflammation during Staphylococcal infection. Cell Rep 2024; 43:114607. [PMID: 39126652 DOI: 10.1016/j.celrep.2024.114607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/09/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Macrophage metabolic plasticity is central to inflammatory programming, yet mechanisms of coordinating metabolic and inflammatory programs during infection are poorly defined. Here, we show that type I interferon (IFN) temporally guides metabolic control of inflammation during methicillin-resistant Staphylococcus aureus (MRSA) infection. We find that staggered Toll-like receptor and type I IFN signaling in macrophages permit a transient energetic state of combined oxidative phosphorylation (OXPHOS) and aerobic glycolysis followed by inducible nitric oxide synthase (iNOS)-mediated OXPHOS disruption. This disruption promotes type I IFN, suppressing other pro-inflammatory cytokines, notably interleukin-1β. Upon infection, iNOS expression peaks at 24 h, followed by lactate-driven Nos2 repression via histone lactylation. Type I IFN pre-conditioning prolongs infection-induced iNOS expression, amplifying type I IFN. Cutaneous MRSA infection in mice constitutively expressing epidermal type I IFN results in elevated iNOS levels, impaired wound healing, vasculopathy, and lung infection. Thus, kinetically regulated type I IFN signaling coordinates immunometabolic checkpoints that control infection-induced inflammation.
Collapse
Affiliation(s)
- Mack B Reynolds
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Benjamin Klein
- Department of Internal Medicine, Division of Rheumatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Michael J McFadden
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Norah K Judge
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hannah E Navarrete
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Britton C Michmerhuizen
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Dominik Awad
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tracey L Schultz
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Paul W Harms
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Li Zhang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Teresa R O'Meara
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jonathan Z Sexton
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - J Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
5
|
Melvin WJ, Bauer TM, Mangum KD, Audu CO, Shadiow J, Barrett EC, Joshi AD, Moon JY, Bogle R, Mazumder P, Wolf SJ, Kunke SL, Gudjonsson JE, Davis FM, Gallagher KA. The histone methyltransferase Mixed-lineage-leukemia-1 drives T cell phenotype via Notch signaling in diabetic tissue repair. JCI Insight 2024; 9:e179012. [PMID: 39250432 PMCID: PMC11463913 DOI: 10.1172/jci.insight.179012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 08/15/2024] [Indexed: 09/11/2024] Open
Abstract
Immune cell-mediated inflammation is important in normal tissue regeneration but can be pathologic in diabetic wounds. Limited literature exists on the role of CD4+ T cells in normal or diabetic wound repair; however, the imbalance of CD4+ Th17/Tregs has been found to promote inflammation in other diabetic tissues. Here, using human tissue and murine transgenic models, we identified that the histone methyltransferase Mixed-lineage-leukemia-1 (MLL1) directly regulates the Th17 transcription factor RORγ via an H3K4me3 mechanism and increases expression of Notch receptors and downstream Notch signaling. Furthermore, we found that Notch receptor signaling regulates CD4+ Th cell differentiation and is critical for normal wound repair, and loss of upstream Notch pathway mediators or receptors in CD4+ T cells resulted in the loss of CD4+ Th cell differentiation in wounds. In diabetes, MLL1 and Notch-receptor signaling was upregulated in wound CD4+ Th cells, driving CD4+ T cells toward the Th17 cell phenotype. Treatment of diabetic wound CD4+ T cells with a small molecule inhibitor of MLL1 (MI-2) yielded a significant reduction in CD4+ Th17 cells and IL-17A. This is the first study to our knowledge to identify the MLL1-mediated mechanisms responsible for regulating the Th17/Treg balance in normal and diabetic wounds and to define the complex role of Notch signaling in CD4+ T cells in wounds, where increased or decreased Notch signaling both result in pathologic wound repair. Therapeutic targeting of MLL1 in diabetic CD4+ Th cells may decrease pathologic inflammation through regulation of CD4+ T cell differentiation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sonya J. Wolf
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Steven L. Kunke
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Katherine A. Gallagher
- Section of Vascular Surgery, Department of Surgery
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
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
|
Audu CO, Wolf SJ, Joshi AD, Moon JY, Melvin WJ, Sharma SB, Davis FM, Obi AT, Wasikowski R, Tsoi LC, Barrett EC, Mangum KD, Bauer TM, Kunkel SL, Moore BB, Gallagher KA. Histone demethylase JARID1C/KDM5C regulates Th17 cells by increasing IL-6 expression in diabetic plasmacytoid dendritic cells. JCI Insight 2024; 9:e172959. [PMID: 38912581 PMCID: PMC11383169 DOI: 10.1172/jci.insight.172959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 05/10/2024] [Indexed: 06/25/2024] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are first responders to tissue injury, where they prime naive T cells. The role of pDCs in physiologic wound repair has been examined, but little is known about pDCs in diabetic wound tissue and their interactions with naive CD4+ T cells. Diabetic wounds are characterized by increased levels of inflammatory IL-17A cytokine, partly due to increased Th17 CD4+ cells. This increased IL-17A cytokine, in excess, impairs tissue repair. Here, using human tissue and murine wound healing models, we found that diabetic wound pDCs produced excess IL-6 and TGF-β and that these cytokines skewed naive CD4+ T cells toward a Th17 inflammatory phenotype following cutaneous injury. Further, we identified that increased IL-6 cytokine production by diabetic wound pDCs is regulated by a histone demethylase, Jumonji AT-rich interactive domain 1C histone demethylase (JARID1C). Decreased JARID1C increased IL-6 transcription in diabetic pDCs, and this process was regulated upstream by an IFN-I/TYK2/JAK1,3 signaling pathway. When inhibited in nondiabetic wound pDCs, JARID1C skewed naive CD4+ T cells toward a Th17 phenotype and increased IL-17A production. Together, this suggests that diabetic wound pDCs are epigenetically altered to increase IL-6 expression that then affects T cell phenotype. These findings identify a therapeutically manipulable pathway in diabetic wounds.
Collapse
Affiliation(s)
- Christopher O Audu
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - Sonya J Wolf
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Amrita D Joshi
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jadie Y Moon
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - William J Melvin
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sriganesh B Sharma
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Frank M Davis
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Andrea T Obi
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Rachel Wasikowski
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lam C Tsoi
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Emily C Barrett
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kevin D Mangum
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Tyler M Bauer
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Steven L Kunkel
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Pathology, School of Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Beth B Moore
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Katherine A Gallagher
- Section of Vascular Surgery, Department of Surgery, and
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
8
|
Li TM, Zyulina V, Seltzer ES, Dacic M, Chinenov Y, Daamen AR, Veiga KR, Schwartz N, Oliver DJ, Cabahug-Zuckerman P, Lora J, Liu Y, Shipman WD, Ambler WG, Taber SF, Onel KB, Zippin JH, Rashighi M, Krueger JG, Anandasabapathy N, Rogatsky I, Jabbari A, Blobel CP, Lipsky PE, Lu TT. The interferon-rich skin environment regulates Langerhans cell ADAM17 to promote photosensitivity in lupus. eLife 2024; 13:e85914. [PMID: 38860651 PMCID: PMC11213570 DOI: 10.7554/elife.85914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 06/10/2024] [Indexed: 06/12/2024] Open
Abstract
The autoimmune disease lupus erythematosus (lupus) is characterized by photosensitivity, where even ambient ultraviolet radiation (UVR) exposure can lead to development of inflammatory skin lesions. We have previously shown that Langerhans cells (LCs) limit keratinocyte apoptosis and photosensitivity via a disintegrin and metalloprotease 17 (ADAM17)-mediated release of epidermal growth factor receptor (EGFR) ligands and that LC ADAM17 sheddase activity is reduced in lupus. Here, we sought to understand how the lupus skin environment contributes to LC ADAM17 dysfunction and, in the process, differentiate between effects on LC ADAM17 sheddase function, LC ADAM17 expression, and LC numbers. We show through transcriptomic analysis a shared IFN-rich environment in non-lesional skin across human lupus and three murine models: MRL/lpr, B6.Sle1yaa, and imiquimod (IMQ) mice. IFN-I inhibits LC ADAM17 sheddase activity in murine and human LCs, and IFNAR blockade in lupus model mice restores LC ADAM17 sheddase activity, all without consistent effects on LC ADAM17 protein expression or LC numbers. Anti-IFNAR-mediated LC ADAM17 sheddase function restoration is associated with reduced photosensitive responses that are dependent on EGFR signaling and LC ADAM17. Reactive oxygen species (ROS) is a known mediator of ADAM17 activity; we show that UVR-induced LC ROS production is reduced in lupus model mice, restored by anti-IFNAR, and is cytoplasmic in origin. Our findings suggest that IFN-I promotes photosensitivity at least in part by inhibiting UVR-induced LC ADAM17 sheddase function and raise the possibility that anifrolumab ameliorates lupus skin disease in part by restoring this function. This work provides insight into IFN-I-mediated disease mechanisms, LC regulation, and a potential mechanism of action for anifrolumab in lupus.
Collapse
Affiliation(s)
- Thomas Morgan Li
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
| | - Victoria Zyulina
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Department of Microbiology and Immunology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Ethan S Seltzer
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
| | - Marija Dacic
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Physiology, Biophysics, and Systems Biology Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Yurii Chinenov
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery Research InstituteNew YorkUnited States
| | - Andrea R Daamen
- Department of Medicine, AMPEL BioSolutionsCharlottesvilleUnited States
| | - Keila R Veiga
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Pediatric Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
- Department of Pediatrics, Weill Cornell Medical CollegeNew YorkUnited States
| | - Noa Schwartz
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
| | - David J Oliver
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery Research InstituteNew YorkUnited States
| | - Pamela Cabahug-Zuckerman
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
| | - Jose Lora
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Physiology, Biophysics, and Systems Biology Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Yong Liu
- Department of Dermatology, Weill Cornell Medical CollegeNew YorkUnited States
| | - William D Shipman
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, Weill Cornell Medical CollegeNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - William G Ambler
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Pediatric Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
- Department of Pediatrics, Weill Cornell Medical CollegeNew YorkUnited States
| | - Sarah F Taber
- Pediatric Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
- Department of Pediatrics, Weill Cornell Medical CollegeNew YorkUnited States
| | - Karen B Onel
- Pediatric Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
- Department of Pediatrics, Weill Cornell Medical CollegeNew YorkUnited States
| | - Jonathan H Zippin
- Department of Dermatology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Mehdi Rashighi
- Department of Dermatology, University of Massachusetts Medical SchoolWorcesterUnited States
| | - James G Krueger
- Laboratory of Investigative Dermatology, Rockefeller UniversityNew YorkUnited States
| | - Niroshana Anandasabapathy
- Department of Dermatology, Weill Cornell Medical CollegeNew YorkUnited States
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, Weill Cornell Medical CollegeNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Inez Rogatsky
- Department of Microbiology and Immunology, Weill Cornell Medical CollegeNew YorkUnited States
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Ali Jabbari
- Laboratory of Investigative Dermatology, Rockefeller UniversityNew YorkUnited States
| | - Carl P Blobel
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Physiology, Biophysics, and Systems Biology Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medical CollegeNew YorkUnited States
| | - Peter E Lipsky
- Department of Medicine, AMPEL BioSolutionsCharlottesvilleUnited States
| | - Theresa T Lu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research InstituteNew YorkUnited States
- Department of Microbiology and Immunology, Weill Cornell Medical CollegeNew YorkUnited States
- Pediatric Rheumatology, Department of Medicine, Hospital for Special SurgeryNew YorkUnited States
- Department of Pediatrics, Weill Cornell Medical CollegeNew YorkUnited States
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| |
Collapse
|
9
|
Loftus SN, Gharaee-Kermani M, Xu B, Moore TM, Hannoudi A, Mallbris MJ, Klein B, Gudjonsson JE, Kahlenberg JM. Interferon alpha promotes caspase-8 dependent ultraviolet light-mediated keratinocyte apoptosis via interferon regulatory factor 1. Front Immunol 2024; 15:1384606. [PMID: 38660315 PMCID: PMC11039837 DOI: 10.3389/fimmu.2024.1384606] [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] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Ultraviolet (UV) light is a known trigger of both cutaneous and systemic disease manifestations in lupus patients. Lupus skin has elevated expression of type I interferons (IFNs) that promote increased keratinocyte (KC) death after UV exposure. The mechanisms by which KC cell death is increased by type I IFNs are unknown. Methods Here, we examine the specific cell death pathways that are activated in KCs by type I IFN priming and UVB exposure using a variety of pharmacological and genetic approaches. Mice that overexpress Ifnk in the epidermis were exposed to UVB light and cell death was measured. RNA-sequencing from IFN-treated KCs was analyzed to identify candidate genes for further analysis that could drive enhanced cell death responses after UVB exposure. Results We identify enhanced activation of caspase-8 dependent apoptosis, but not other cell death pathways, in type I IFN and UVB-exposed KCs. In vivo, overexpression of epidermal Ifnk resulted in increased apoptosis in murine skin after UVB treatment. This increase in KC apoptosis was not dependent on known death ligands but rather dependent on type I IFN-upregulation of interferon regulatory factor 1 (IRF1). Discussion These data suggest that enhanced sensitivity to UV light exhibited by lupus patients results from type I IFN priming of KCs that drives IRF1 expression resulting in caspase-8 activation and increased apoptosis after minimal exposures to UVB.
Collapse
Affiliation(s)
- Shannon N. Loftus
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, United States
| | - Mehrnaz Gharaee-Kermani
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Bin Xu
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Tyson M. Moore
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Andrew Hannoudi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Mischa J. Mallbris
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Benjamin Klein
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | | | - J. Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
10
|
Hu X, Wang L, Deng J, Xu H, Song K, Bu L, Pang B. Dietary nitrate accelerates the healing of infected skin wounds in mice by increasing microvascular density. Biochem Biophys Res Commun 2023; 686:149176. [PMID: 37924670 DOI: 10.1016/j.bbrc.2023.149176] [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: 10/22/2023] [Accepted: 10/27/2023] [Indexed: 11/06/2023]
Abstract
As skin injuries resulting from acute trauma, burns, and chronic diseases present significant challenges to healthcare systems worldwide, the promotion of skin wound healing remains an unmet therapeutic area. Dietary nitrate serves as a crucial pathway for the production of nitric oxide, which plays various physiological roles in the body, including vasodilation, increased blood flow, and antioxidant activity. However, the impact of dietary nitrate on skin wound healing remains uncertain. This study aimed to investigate the role of dietary nitrate in infected skin wound healing using a mouse model. We created a full-thickness wound infection model in mice and examine the effects of dietary nitrate (0.5 mmol/kg/d and 1 mmol/kg/d) on wound healing. The results demonstrated that dietary nitrate significantly increased serum nitrate and nitrite levels, leading to accelerated wound healing by increasing microvascular density, promoting collagen deposition and re-epithelialization. Moreover, nitrate supplementation exhibited a certain degree of reduction in inflammatory factors within the body. Our study also found that 1 mmol/kg/d nitrate has a more effective therapeutic effect and can increase blood perfusion and expedite the formation of new blood vessels, thereby promoting skin wound healing. These results indicate that dietary nitrate presents a novel therapeutic approach for infected skin wound healing.
Collapse
Affiliation(s)
- Xiaodan Hu
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China; School of Stomatology of Qingdao University, Qingdao, 288003, China.
| | - Lin Wang
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China; School of Stomatology of Qingdao University, Qingdao, 288003, China.
| | - Jiangzhi Deng
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China; School of Stomatology of Qingdao University, Qingdao, 288003, China
| | - Haoyue Xu
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China; School of Stomatology of Qingdao University, Qingdao, 288003, China
| | - Kai Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China; School of Stomatology of Qingdao University, Qingdao, 288003, China
| | - Lingxue Bu
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China; School of Stomatology of Qingdao University, Qingdao, 288003, China.
| | - Baoxing Pang
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, China; School of Stomatology of Qingdao University, Qingdao, 288003, China.
| |
Collapse
|
11
|
Hou X, Cui B, Qiu A, Liu N, Zhuang S. Pharmacological inhibition of the mixed lineage leukemia 1-menin interaction aggravates acute kidney injury induced by folic acid and ischemia-reperfusion in mice. Am J Physiol Renal Physiol 2023; 325:F669-F680. [PMID: 37733875 DOI: 10.1152/ajprenal.00287.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 08/16/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
Mixed lineage leukemia 1 (MLL1) is a methyltransferase that induces histone H3 lysine 4 trimethylation (H3K4me3) and partially exerts its untoward functional effects by interacting with multiple subunits including menin and WD repeat-containing protein 5 (WDR5). In this study, we investigated the role and mechanisms of MLL1 in murine models of acute kidney injury induced by folic acid (FA) and ischemia-reperfusion. Injury to the kidney elevated the expression of MLL1, menin, WDR5, and H3K4Me3, which was accompanied by increased serum creatinine and blood urea nitrogen, renal tubular injury, and apoptosis. Pharmacological inhibition of MLL1 activity with MI503 to disrupt the interaction between MLL1 with menin further increased serum creatinine and blood urea nitrogen levels, enhanced expression of neutrophil gelatinase-associated lipocalin and kidney injury molecule-1, and induced more apoptosis in the kidney following FA and ischemia-reperfusion injury. In contrast, MI503 treatment decreased the expression of vimentin and proliferating cell nuclear antigens. Similarly, treatment with MM102 to disrupt the interaction between MLL1 and WDR5 also worsened renal dysfunction, aggravated tubular cell injury, increased apoptosis, and inhibited cellular dedifferentiation and proliferation in mice following FA injection. Moreover, MI503 inhibited FA-induced phosphorylation of epidermal growth factor receptor, signal transducer and activator of transcription 3, and extracellular signal-regulated kinase-1/2 in injured kidneys. Collectively, these data suggest that MLL1 contributes to renal protection and functional recovery and promotes renal regeneration through a mechanism associated with activation of the epidermal growth factor receptor signaling pathway.NEW & NOTEWORTHY Mixed lineage leukemia 1 (MLL1) is a methyltransferase that induces histone H3 lysine 4 trimethylation and exerts its functional roles by interacting with multiple subunits. In this study, we demonstrated that inhibition of MLL1 activity by MI503 or MM102 aggravated renal injury and apoptosis and suppressed renal tubular cell dedifferentiation and proliferation, suggesting that MLL1 activation during acute kidney injury acts as an intrinsic protective mechanism to mediate renal tubular cell survival and regeneration.
Collapse
Affiliation(s)
- Xiying Hou
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Binbin Cui
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Andong Qiu
- School of Life Science and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai, China
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island, United States
| |
Collapse
|
12
|
Schmidt BM, Holmes CM, Najarian K, Gallagher K, Haus JM, Shadiow J, Ye W, Ang L, Burant A, Baker N, Katona A, Martin CL, Pop-Busui R. On diabetic foot ulcer knowledge gaps, innovation, evaluation, prediction markers, and clinical needs. J Diabetes Complications 2022; 36:108317. [PMID: 36215794 PMCID: PMC10087892 DOI: 10.1016/j.jdiacomp.2022.108317] [Citation(s) in RCA: 8] [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: 06/27/2022] [Revised: 08/22/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022]
Abstract
Diabetic foot ulcers (DFUs) remain a very prevalent and challenging complication of diabetes worldwide due to high morbidity, high risks of lower extremity amputation and associated mortality. Despite major advances in diabetes treatment in general, there is a paucity of FDA approved technologies and therapies to promote successful healing. Furthermore, accurate biomarkers to identify patients at risk of non-healing and monitor response-to-therapy are significantly lacking. To date, research has been slowed by a lack of coordinated efforts among basic scientists and clinical researchers and confounded by non-standardized heterogenous collection of biospecimen and patient associated data. Novel technologies, especially those in the single and 'multiomics' arena, are being used to advance the study of diabetic foot ulcers but require pragmatic study design to ensure broad adoption following validation. These high throughput analyses offer promise to investigate potential biomarkers across wound trajectories and may support information on wound healing and pathophysiology not previously well understood. Additionally, these biomarkers may be used at the point-of-care. In combination with national scalable research efforts, which seek to address the limitations and better inform clinical practice, coordinated and integrative insights may lead to improved limb salvage rates.
Collapse
Affiliation(s)
- Brian M Schmidt
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America.
| | - Crystal M Holmes
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Kayvan Najarian
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States of America
| | - Katherine Gallagher
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Abor, MI 48109, United States of America
| | - Jacob M Haus
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States of America
| | - James Shadiow
- School of Kinesiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Wen Ye
- Biostatistics Department, School of Public Health, University of Michigan, Ann Arbor, MI, United States of America
| | - Lynn Ang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Aaron Burant
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Nicole Baker
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Aimee Katona
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Catherine L Martin
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Rodica Pop-Busui
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
| |
Collapse
|
13
|
Audu CO, Melvin WJ, Joshi AD, Wolf SJ, Moon JY, Davis FM, Barrett EC, Mangum KD, Deng H, Xing X, Wasikowski R, Tsoi LC, Sharma SB, Bauer TM, Shadiow J, Corriere MA, Obi AT, Kunkel SL, Levi B, Moore BB, Gudjonsson JE, Smith AM, Gallagher KA. Macrophage-specific inhibition of the histone demethylase JMJD3 decreases STING and pathologic inflammation in diabetic wound repair. Cell Mol Immunol 2022; 19:1251-1262. [PMID: 36127466 PMCID: PMC9622909 DOI: 10.1038/s41423-022-00919-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/09/2022] [Indexed: 02/01/2023] Open
Abstract
Macrophage plasticity is critical for normal tissue repair following injury. In pathologic states such as diabetes, macrophage plasticity is impaired, and macrophages remain in a persistent proinflammatory state; however, the reasons for this are unknown. Here, using single-cell RNA sequencing of human diabetic wounds, we identified increased JMJD3 in diabetic wound macrophages, resulting in increased inflammatory gene expression. Mechanistically, we report that in wound healing, JMJD3 directs early macrophage-mediated inflammation via JAK1,3/STAT3 signaling. However, in the diabetic state, we found that IL-6, a cytokine increased in diabetic wound tissue at later time points post-injury, regulates JMJD3 expression in diabetic wound macrophages via the JAK1,3/STAT3 pathway and that this late increase in JMJD3 induces NFκB-mediated inflammatory gene transcription in wound macrophages via an H3K27me3 mechanism. Interestingly, RNA sequencing of wound macrophages isolated from mice with JMJD3-deficient myeloid cells (Jmjd3f/fLyz2Cre+) identified that the STING gene (Tmem173) is regulated by JMJD3 in wound macrophages. STING limits inflammatory cytokine production by wound macrophages during healing. However, in diabetic mice, its role changes to limit wound repair and enhance inflammation. This finding is important since STING is associated with chronic inflammation, and we found STING to be elevated in human and murine diabetic wound macrophages at late time points. Finally, we demonstrate that macrophage-specific, nanoparticle inhibition of JMJD3 in diabetic wounds significantly improves diabetic wound repair by decreasing inflammatory cytokines and STING. Taken together, this work highlights the central role of JMJD3 in tissue repair and identifies cell-specific targeting as a viable therapeutic strategy for nonhealing diabetic wounds.
Collapse
Affiliation(s)
- Christopher O Audu
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - William J Melvin
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, Section of General Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Amrita D Joshi
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Sonya J Wolf
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Jadie Y Moon
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Frank M Davis
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Emily C Barrett
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, Section of General Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kevin D Mangum
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Hongping Deng
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Champaign, IL, USA
| | - Xianying Xing
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Rachel Wasikowski
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Sriganesh B Sharma
- Department of Surgery, Section of General Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Tyler M Bauer
- Department of Surgery, Section of General Surgery, University of Michigan, Ann Arbor, MI, USA
| | - James Shadiow
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew A Corriere
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Andrea T Obi
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Steven L Kunkel
- Department of Surgery, Section of General Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Benjamin Levi
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bethany B Moore
- Department of Surgery, Section of General Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Andrew M Smith
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Champaign, IL, USA
| | - Katherine A Gallagher
- Department of Surgery, Section of Vascular Surgery, University of Michigan, Ann Arbor, MI, USA.
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|