1
|
Zheng Z, Yang T, Cao H, Yu J, Fang X, He X, Zou L, Tang D, Lu Z, Liu J, Yu L. Liang-Ge-San drives macrophages toward M2 polarization for alleviating lipopolysaccharide-induced acute lung injury via activating the miR-21/PTEN axis. Fitoterapia 2025; 184:106572. [PMID: 40318703 DOI: 10.1016/j.fitote.2025.106572] [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: 01/16/2025] [Revised: 04/17/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
Acute lung injury (ALI) has high clinical mortality currently and no specific drugs available for its treatment. Although Liang-Ge-San (LGS), a traditional Chinese medicine formula, is known to promote inflammation resolution and shorten hospitalization duration of ALI, the mechanism is still unclear. Our results demonstrated that LGS regulated the dynamic balance of macrophage polarization as reflected by up-regulating the expression of anti-inflammatory factors (CD206, Arg-1 and IL-10) in advance to counteract the high expression of pro-inflammatory factors (CD86, iNOS, IL-6 and TNF-α) in vitro. MiR-21 concentration was elevated in LPS-challenged RAW264.7 cells and ALI mice. Moreover, the overexpression of miR-21 mimicked the anti-inflammatory effects of LGS, whereas a miR-21 inhibitor abolished the protective effects of LGS in vitro. Most importantly, LGS protected ALI mice from LPS which could be counteracted by the treatment of miR-21 antagomir. Furthermore, LGS could inhibit the transcriptional activity and protein expression of PTEN by up-regulating miR-21. In summary, LGS functions by regulating the miR-21/PTEN axis to induce a shift in macrophages from a pro-inflammatory phenotype to an anti-inflammatory phenotype, thereby alleviating LPS-induced ALI. This study supports the clinical evidence of LGS in the treatment of ALI.
Collapse
Affiliation(s)
- Zhuping Zheng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China; Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China
| | - Tangjia Yang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China
| | - Huihui Cao
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China
| | - Xiaochuan Fang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China
| | - Xuemei He
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China
| | - Lifang Zou
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China
| | - Dongkai Tang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China
| | - Zibin Lu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China.
| | - Junshan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China.
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, International Joint Labaratory of Zebrafish Models of Human Diseases and Drug Discovery, Guangzhou 510515, PR China; Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
2
|
Gallerand A, Han J, Mintz RL, Chen J, Lee DD, Chan MM, Harmon TT, Lin X, Huckstep CG, Du S, Liu T, Kipnis J, Lavine KJ, Schilling JD, Morley SC, Zinselmeyer BH, Murphy KM, Randolph GJ. Tracing LYVE1 + peritoneal fluid macrophages unveils two paths to resident macrophage repopulation with differing reliance on monocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644175. [PMID: 40166277 PMCID: PMC11957119 DOI: 10.1101/2025.03.19.644175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Mouse resident peritoneal macrophages, called large cavity macrophages (LCM), arise from embryonic progenitors that proliferate as mature, CD73+Gata6+ tissue-specialized macrophages. After injury from irradiation or inflammation, monocytes are thought to replenish CD73+Gata6+ LCMs through a CD73-LYVE1+ LCM intermediate. Here, we show that CD73-LYVE1+ LCMs indeed yield Gata6+CD73+ LCMs through integrin-mediated interactions with mesothelial surfaces. CD73-LYVE1+ LCM repopulation of the peritoneum was reliant upon and quantitatively proportional to recruited monocytes. Unexpectedly, fate mapping indicated that only ~10% of Gata6-dependent LCMs that repopulated the peritoneum after injury depended on the LYVE1+ LCM stage. Further supporting nonoverlapping lifecycles of CD73-LYVE1+ and CD73+Gata6+ LCMs, in mice bearing a paucity of monocytes, Gata6+CD73+ LCMs rebounded after ablative irradiation substantially more efficiently than their presumed LYVE1+ or CD73- LCM upstream precursors. Thus, after inflammatory insult, two temporally parallel pathways, each generating distinct differentiation intermediates with varying dependencies on monocytes, contribute to the replenish hment of Gata6+ resident peritoneal macrophages.
Collapse
Affiliation(s)
- Alexandre Gallerand
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jichang Han
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel L. Mintz
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Biomedical Engineering Graduate Program, Washington University School of Medicine, St. Louis, MO, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Jing Chen
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences Graduate Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel D. Lee
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mandy M. Chan
- Division of Biology and Biomedical Sciences Graduate Program, Washington University School of Medicine, St. Louis, MO, USA
- Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Tyler T. Harmon
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences Graduate Program, Washington University School of Medicine, St. Louis, MO, USA
- Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xue Lin
- Division of Infectious Disease, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher G. Huckstep
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Siling Du
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences Graduate Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Tiantian Liu
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jonathan Kipnis
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kory J. Lavine
- Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Joel D. Schilling
- Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - S. Celeste Morley
- Division of Infectious Disease, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Bernd H. Zinselmeyer
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kenneth M. Murphy
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gwendalyn J. Randolph
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
3
|
Morabbi A, Karimian M. Therapeutic potential of exosomal lncRNAs derived from stem cells in wound healing: focusing on mesenchymal stem cells. Stem Cell Res Ther 2025; 16:62. [PMID: 39934913 PMCID: PMC11816792 DOI: 10.1186/s13287-025-04200-0] [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: 07/12/2024] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
The self-renewal ability and multipotency of stem cells give them great potential for use in wound healing. Stem cell-derived exosomes, owing to their close biological resemblance to their parent cells, offer a more efficient, safer, and economical approach for facilitating cellular communication and interactions within different environments. This potential makes them particularly valuable in the treatment of both acute and chronic wounds, such as lacerations, burns, and diabetic ulcers. Long non-coding RNAs (lncRNAs) enclosed in exosomes, as one of the leading actors of these extracellular microvesicles, through interaction with miRNAs and regulation of various signaling pathways involved in inflammation, angiogenesis, cell proliferation, and migration, could heal the wounds. Exosome-derived lncRNAs from stem cells facilitate extracellular matrix remodeling through interaction between macrophages and fibroblasts. Moreover, alongside regulating the expression of inflammatory cytokines, controlling reactive oxygen species levels, and enhancing autophagic activity, they also modulate immune responses to support wound healing. Regulating the expression of genes and signaling pathways related to angiogenesis, by increasing blood supply and accelerating the delivery of essential substances to the wound environment, is another effect exosomal lncRNAs derived from stem cells for wound healing. These lncRNAs can also enhance skin wound healing by regulating homeostasis, increasing the proliferation and differentiation of cells involved in the wound-healing process, and enhancing fibroblast viability and migration to the injury site. Ultimately, exosome-derived lncRNAs from stem cells offer valuable and novel insights into the molecular mechanisms underlying improved wound healing. They can pave the way for potential therapeutic strategies, fostering further research for a better future. Meanwhile, exosomes derived from mesenchymal stem cells, due to their exceptional regenerative properties, as well as the lncRNAs derived from these exosomes, have emerged as one of the innovative tools in wound healing. This review article aims to narrate the cellular and molecular roles of exosome-derived lncRNAs from stem cells in enhancing wound healing with a focus on mesenchymal stem cells.
Collapse
Affiliation(s)
- Ali Morabbi
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, 47416-95447, Iran
| | - Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, 47416-95447, Iran.
| |
Collapse
|
4
|
De AK, Ponraj P, Bhattacharya D. Depicting "arms race" of Rhipicephalus microplus and its host on a single frame platform. Parasitol Res 2025; 124:18. [PMID: 39903310 PMCID: PMC11794405 DOI: 10.1007/s00436-025-08459-3] [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: 08/02/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
To improve our knowledge on host-parasite interaction, the study was undertaken on Rhipicephalus microplus infestation in cross breed cattle. This tick, being a voracious blood sucker, remains attached to the skin for prolonged period and inoculates saliva during blood feeding. Biomolecules present in the saliva have detrimental effects on host system. The present study deals with the effects of R. microplus in artificially infected nine months old cattle for a period of 21 days. There was physiological alteration during tick infestation in terms of body temperature, pulse, and respiration rate. There was drop in haemoglobin gram percentage, platelet count, total red and white blood cell count. Thrombocytopenia in infested animals was suggestive of iron deficient anaemia after artificial infestation. R. microplus infestation was found to induce stress in experimental animals. Our study on peripheral blood gene expression was suggestive of Th2 polarization since there was increased IL-4 response. Increased IL-6 response indicated skin damage due to R. microplus infestation and we further correlate eosinophilia with up-regulation of IL-6 and IL-8 responses. Increased IL-10 response and decreased IFN-γ response were suggestive of immunosuppressive and anti-inflammatory properties of tick saliva.
Collapse
Affiliation(s)
- Arun Kumar De
- Animal Science Division, ICAR-Central Island Agricultural Research Institute, South Andaman, Andaman and Nicobar Islands-744101, Port Blair, India
| | - Perumal Ponraj
- Animal Science Division, ICAR-Central Island Agricultural Research Institute, South Andaman, Andaman and Nicobar Islands-744101, Port Blair, India
| | - Debasis Bhattacharya
- Animal Science Division, ICAR-Central Island Agricultural Research Institute, South Andaman, Andaman and Nicobar Islands-744101, Port Blair, India.
| |
Collapse
|
5
|
Su J, Tu Y, Hu X, Wang Y, Chen M, Cao X, Shao M, Zhang F, Ding W. Ambient PM 2.5 orchestrates M1 polarization of alveolar macrophages via activating glutaminase 1-mediated glutaminolysis in acute lung injury. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 366:125467. [PMID: 39653263 DOI: 10.1016/j.envpol.2024.125467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
The temporary explosive growth events of atmospheric fine particulate matter (PM2.5) pollution during late autumn and winter seasons still frequently occur in China. High-concentration exposure to PM2.5 aggravates lung inflammation, leading to acute lung injury (ALI). Alveolar macrophages (AMs) participate in PM2.5-induced pulmonary inflammation and injury. The polarization of AMs is dependent on metabolic reprogramming. However, the mechanism underlying the PM2.5-induced glutaminase-mediated glutaminolysis in AM polarization is still largely obscure. In this study, we found that PM2.5-treated mice exhibited pulmonary dysfunction and inflammation. The concentrations of glutamate and succinate were increased in PM2.5-treated lungs and AMs compared with the controls, whereas glutamine and α-ketoglutarate (α-KG) levels were decreased, indicating that glutaminolysis in AMs was aberrantly activated as evidenced by increased mRNA and protein levels of GLS1 after PM2.5 exposure. Moreover, we determined that the GLS1/nuclear factor kappa-B (NF-κB)/hypoxia-inducible factor-1α (HIF-1α) pathway regulated M1 polarization of AMs upon PM2.5 exposure. Inhibition of glutaminolysis by GLS1 specific inhibitor CB-839 and GLS1 siRNA significantly decreased PM2.5-induced M1 macrophage polarization and attenuated pulmonary damage. Taken together, our findings reveal a novel mechanism by which a metabolic program regulates M1 polarization of AMs and suggest that GLS1-mediated glutaminolysis is a potential therapeutic target for treating PM2.5-induced ALI.
Collapse
Affiliation(s)
- Jingran Su
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yikun Tu
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China; Sino-Danish Center for Education and Research, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoqi Hu
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanli Wang
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mo Chen
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China; Sino-Danish Center for Education and Research, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xue Cao
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengyao Shao
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fang Zhang
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenjun Ding
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
6
|
Zhao M, Jankovic D, Link VM, Souza COS, Hornick KM, Oyesola O, Belkaid Y, Lack J, Loke P. Genetic variation in IL-4 activated tissue resident macrophages determines strain-specific synergistic responses to LPS epigenetically. Nat Commun 2025; 16:1030. [PMID: 39863579 PMCID: PMC11762786 DOI: 10.1038/s41467-025-56379-8] [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: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
How macrophages in the tissue environment integrate multiple stimuli depends on the genetic background of the host, but this is still poorly understood. We investigate IL-4 activation of male C57BL/6 and BALB/c strain specific in vivo tissue-resident macrophages (TRMs) from the peritoneal cavity. C57BL/6 TRMs are more transcriptionally responsive to IL-4 stimulation, with induced genes associated with more super enhancers, induced enhancers, and topologically associating domains (TAD) boundaries. IL-4-directed epigenomic remodeling reveals C57BL/6 specific enrichment of NF-κB, IRF, and STAT motifs. Additionally, IL-4-activated C57BL/6 TRMs demonstrate an augmented synergistic response upon in vitro lipopolysaccharide (LPS) exposure, despite naïve BALB/c TRMs displaying a more robust transcriptional response to LPS. Single-cell RNA sequencing (scRNA-seq) analysis of mixed bone marrow chimeras indicates that transcriptional differences and synergy are cell intrinsic within the same tissue environment. Hence, genetic variation alters IL-4-induced cell intrinsic epigenetic reprogramming resulting in strain specific synergistic responses to LPS exposure.
Collapse
Affiliation(s)
- Mingming Zhao
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Dragana Jankovic
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Camila Oliveira Silva Souza
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Katherine M Hornick
- NIAID Collaborative Bioinformatics Resource, Integrated Data Sciences Section, Research Technology Branch, Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Oyebola Oyesola
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource, Integrated Data Sciences Section, Research Technology Branch, Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Png Loke
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
7
|
Zheng B, Cui D, Deng B, Long W, Ye G, Zhang S, Zeng J. Form-deprivation myopia promotes sclera M2-type macrophages polarization in mice. Biochem Biophys Res Commun 2024; 737:150490. [PMID: 39146710 DOI: 10.1016/j.bbrc.2024.150490] [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: 05/27/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE To explore the phenotype of sclera macrophages in form-deprivation (FD) myopia mice and the effects of M2 macrophage in FD myopia development. METHODS C57BL/6 mice were under 2 weeks of unilateral FD treatment. and they were separated into two groups, including an intraperitoneally injected(IP) vehicle group and Panobinostat (LBH589) (10 mg/kg per body weight) treatment group. All biometric parameters were measured before and after treatments, and the type and density of sclera macrophages were identified by immunofluorescence and RT-qPCR. In vitro, we analyzed the M2 macrophage and primary human sclera fibroblast (HSF) co-culture system by using the transcriptome sequencing method. Gene ontology (GO) and KEGG enrichment analyses were used to pinpoint the biological functions and pathways associated with the identified Differentially Expressed Genes (DEGs). The hub genes were investigated using the STRING database and Cytoscape software and were confirmed using RT-qPCR. RESULTS We found that the M2-type sclera macrophage density and expression increased in FD-treated eyes. The results showed that LBH589 inhibited the M2 macrophage polarization, and reduced FDM development. GO and KEGG analyses revealed that the DEGs were predominantly involved in the synthesis and breakdown of the extracellular matrix (ECM), as well as in pathways related to ECM-receptor interaction and the PI3K-Akt signaling pathway. Five hub genes (FN-1, MMP-2, COL1A1, CD44, and IL6) were identified, and RT-qPCR validated the variation in expression levels among these genes. CONCLUSION M2 macrophage polarization occurred in the sclera in FDM mice. Panobinostat-mediated inhibition of M2 macrophage polarization may decrease FDM progression, as M2 macrophages are crucial in controlling ECM remodeling by HSFs.
Collapse
Affiliation(s)
- Bingru Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 Xianlie Road, Guangzhou, 510060, China; Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, 18 Zetian Road, Futian District, Shenzhen, 518040, China
| | - Dongmei Cui
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, 18 Zetian Road, Futian District, Shenzhen, 518040, China
| | - Baodi Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Wen Long
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, 18 Zetian Road, Futian District, Shenzhen, 518040, China
| | - Guitong Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 Xianlie Road, Guangzhou, 510060, China
| | - Shaochong Zhang
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, 18 Zetian Road, Futian District, Shenzhen, 518040, China.
| | - Junwen Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, 54 Xianlie Road, Guangzhou, 510060, China.
| |
Collapse
|
8
|
Li X, Zhang G, Wang M, Lu C, Zhang G, Chen Z, Ji Y. Comparison of stromal vascular fraction cell composition between Coleman fat and extracellular matrix/stromal vascular fraction gel. Adipocyte 2024; 13:2360037. [PMID: 38829527 PMCID: PMC11152101 DOI: 10.1080/21623945.2024.2360037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
As a mechanically condensed product of Coleman fat, extracellular matrix/stromal vascular fraction gel (ECM/SVF-gel) eliminates adipocytes, concentrates SVF cells, and improves fat graft retention. This study aims to compare SVF cell composition between Coleman fat and ECM/SVF-gel. Matched Coleman fat and ECM/SVF-gel of 28 healthy women were subjected to RNA-seq, followed by functional enrichment and cell-type-specific enrichment analyses, and deconvolution of SVF cell subsets, reconstructing SVF cell composition in the transcriptome level. ECM/SVF-gels had 9 upregulated and 73 downregulated differentially expressed genes (DEGs). Downregulated DEGs were mainly associated with inflammatory and immune responses, and enriched in fat macrophages. M2 macrophages, resting CD4+ memory T cells, M1 macrophages, resting mast cells, and M0 macrophages ranked in the top five most prevalent immune cells in the two groups. The proportions of the principal non-immune cells (e.g., adipose-derived stem cells, pericytes, preadipocytes, microvascular endothelial cells) had no statistical differences between the two groups. Our findings reveal ECM/SVF-gels share the same dominant immune cells beneficial to fat graft survival with Coleman fat, but exhibiting obvious losses of immune cells (especially macrophages), while non-immune cells necessary for adipose regeneration might have no significant loss in ECM/SVF-gels and their biological effects could be markedly enhanced by the ECM/SVF-gel's condensed nature.
Collapse
Affiliation(s)
- Xiaoyun Li
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Guohong Zhang
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Mengmeng Wang
- Medical Cosmetic Center, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Changhao Lu
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Guangping Zhang
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Zhehui Chen
- Medical Cosmetic Center, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yingchang Ji
- Medical Cosmetic Center, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
9
|
Rawal S, Randhawa V, Rizvi SHM, Sachan M, Wara AK, Pérez-Cremades D, Weisbrod RM, Hamburg NM, Feinberg MW. miR-369-3p ameliorates diabetes-associated atherosclerosis by regulating macrophage succinate-GPR91 signalling. Cardiovasc Res 2024; 120:1693-1712. [PMID: 38703377 PMCID: PMC11587565 DOI: 10.1093/cvr/cvae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 03/04/2024] [Accepted: 05/02/2024] [Indexed: 05/06/2024] Open
Abstract
AIMS Diabetes leads to dysregulated macrophage immunometabolism, contributing to accelerated atherosclerosis progression. Identifying critical factors to restore metabolic alterations and promote resolution of inflammation remains an unmet goal. MicroRNAs orchestrate multiple signalling events in macrophages, yet their therapeutic potential in diabetes-associated atherosclerosis remains unclear. METHODS AND RESULTS miRNA profiling revealed significantly lower miR-369-3p expression in aortic intimal lesions from Ldlr-/- mice on a high-fat sucrose-containing (HFSC) diet for 12 weeks. miR-369-3p was also reduced in peripheral blood mononuclear cells from diabetic patients with coronary artery disease (CAD). Cell-type expression profiling showed miR-369-3p enrichment in aortic macrophages. In vitro, oxLDL treatment reduced miR-369-3p expression in mouse bone marrow-derived macrophages (BMDMs). Metabolic profiling in BMDMs revealed that miR-369-3p overexpression blocked the oxidized low density lipoprotein (oxLDL)-mediated increase in the cellular metabolite succinate and reduced mitochondrial respiration (OXPHOS) and inflammation [Interleukin (lL)-1β, TNF-α, and IL-6]. Mechanistically, miR-369-3p targeted the succinate receptor (GPR91) and alleviated the oxLDL-induced activation of inflammasome signalling pathways. Therapeutic administration of miR-369-3p mimics in HFSC-fed Ldlr-/- mice reduced GPR91 expression in lesional macrophages and diabetes-accelerated atherosclerosis, evident by a decrease in plaque size and pro-inflammatory Ly6Chi monocytes. RNA-Seq analyses showed more pro-resolving pathways in plaque macrophages from miR-369-3p-treated mice, consistent with an increase in macrophage efferocytosis in lesions. Finally, a GPR91 antagonist attenuated oxLDL-induced inflammation in primary monocytes from human subjects with diabetes. CONCLUSION These findings establish a therapeutic role for miR-369-3p in halting diabetes-associated atherosclerosis by regulating GPR91 and macrophage succinate metabolism.
Collapse
MESH Headings
- Animals
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Macrophages/metabolism
- Macrophages/pathology
- Signal Transduction
- Humans
- Mice, Knockout
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Male
- Mice, Inbred C57BL
- Disease Models, Animal
- Lipoproteins, LDL/metabolism
- Succinic Acid/metabolism
- Plaque, Atherosclerotic
- Mice
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Receptors, LDL/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/metabolism
- Aortic Diseases/genetics
- Aortic Diseases/prevention & control
- Aortic Diseases/immunology
- Cells, Cultured
- Gene Expression Regulation
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/prevention & control
- Female
- Middle Aged
Collapse
Affiliation(s)
- Shruti Rawal
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Vinay Randhawa
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Syed Husain Mustafa Rizvi
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA
- Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Madhur Sachan
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Akm Khyrul Wara
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Daniel Pérez-Cremades
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Department of Physiology, University of Valencia, INCLIVA Biomedical Research Institute, Valencia 46010, Spain
| | - Robert M Weisbrod
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA
- Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Naomi M Hamburg
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA
- Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Mark W Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
10
|
Flores-Sotelo I, Juárez N, González MI, Chávez A, Vannan DT, Eksteen B, Terrazas LI, Reyes JL. Endogenous innate sensor NLRP3 is a key component in peritoneal macrophage dynamics required for cestode establishment. Immunol Res 2024; 72:948-963. [PMID: 38842647 PMCID: PMC11564225 DOI: 10.1007/s12026-024-09496-3] [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: 11/23/2023] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
The NLRP3 receptor can assemble inflammasome platforms to trigger inflammatory responses; however, accumulating evidence suggests that it can also display anti-inflammatory properties. Here, we explored the role of nucleotide-binding oligomerization domain pyrin-containing protein 3 (NLRP3) in Taenia crassiceps experimental infection, which requires immune polarization into a Th2-type profile and peritoneal influx of suppressive macrophages for successful colonization. NLRP3 deficient mice (NLRP3-/-) were highly resistant against T. crassiceps, relative to wild-type (WT) mice. Resistance in NLRP3-/- mice was associated with a diminished IL-4 output, high levels of IL-15, growth factor for both innate and adaptive lymphocytes, and a dramatic decrease in peritoneum-infiltrating suppressive macrophages. Also, a transcriptional analysis on bone marrow-derived macrophages exposed to Taenia-secreted antigens and IL-4 revealed that NLRP3-/- macrophages express reduced transcripts of relm-α and PD-1 ligands, markers of alternative activation and suppressive ability, respectively. Finally, we found that the resistance displayed by NLRP3-/- mice is transferred through intestinal microbiota exchange, since WT mice co-housed with NLRP3-/- mice were significantly more resistant than WT animals preserving their native microbiota. Altogether, these data demonstrate that NLRP3 is a component of innate immunity required for T. crassiceps to establish, most likely contributing to macrophage recruitment, and controlling lymphocyte-stimulating cytokines such as IL-15.
Collapse
Affiliation(s)
- Irán Flores-Sotelo
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Natalia Juárez
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Marisol I González
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Auraamellaly Chávez
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - Danielle T Vannan
- Boston Scientific Corporation, Urology Division, 200 Boston Scientific Way, Marlborough, MA, USA
| | | | - Luis I Terrazas
- Laboratorio de Inmunoparasitología, UBIMED, FES Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico
| | - José L Reyes
- Laboratorio de Inmunología Experimental y Regulación de la Inflamación Hepato-Intestinal, UBIMED, FES, Iztacala, UNAM, Tlalnepantla de Baz, Estado de México, Mexico.
| |
Collapse
|
11
|
Silver SV, Tucker KJ, Vickman RE, Lanman NA, Semmes OJ, Alvarez NS, Popovics P. Characterization of prostate macrophage heterogeneity, foam cell markers, and CXCL17 upregulation in a mouse model of steroid hormone imbalance. Sci Rep 2024; 14:21029. [PMID: 39251671 PMCID: PMC11383972 DOI: 10.1038/s41598-024-71137-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/26/2024] [Indexed: 09/11/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) is a prevalent age-related condition often characterized by debilitating urinary symptoms. Its etiology is believed to stem from hormonal imbalance, particularly an elevated estradiol-to-testosterone ratio and chronic inflammation. Our previous studies using a mouse steroid hormone imbalance model identified a specific increase in macrophages that migrated and accumulated in the prostate lumen where they differentiated into lipid-laden foam cells in mice implanted with testosterone and estradiol pellets, but not in sham animals. The current study focused on further characterizing the cellular heterogeneity of the prostate in this model as well as identifying the specific transcriptomic signature of the recruited foam cells. Moreover, we aimed to identify epithelia-derived signals that drive macrophage infiltration and luminal translocation. Male C57BL/6J mice were implanted with slow-release testosterone and estradiol pellets (T + E2) or sham surgery was performed and the ventral prostates were harvested two weeks later for scRNA-seq analysis. We identified Ear2 + and Cd72 + macrophages that were elevated in response to steroid hormone imbalance, whereas a Mrc1 + resident macrophage population did not change. In addition, an Spp1 + foam cell cluster was almost exclusively found in T + E2 mice. Further markers of foam cells were also identified, including Gpnmb and Trem2, and GPNMB was confirmed as a novel histological marker with immunohistochemistry. Foam cells were also shown to express known pathological factors Vegf, Tgfb1, Ccl6, Cxcl16 and Mmp12. Intriguingly, a screen for chemokines identified the upregulation of epithelia-derived Cxcl17, a known monocyte attractant, in T + E2 prostates suggesting that it might be responsible for the elevated macrophage number as well as their translocation to the lumen. Our study identified macrophage subsets that responded to steroid hormone imbalance as well as further confirmed a potential pathological role of luminal foam cells in the prostate. These results underscore a potential pathological role of the identified prostate foam cells and suggests CXCL17-mediated macrophage migration as a critical initiating event.
Collapse
Affiliation(s)
- Samara V Silver
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Kayah J Tucker
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Renee E Vickman
- Department of Surgery, Endeavor Health, An Academic Affiliate of the University of Chicago Pritzker School of Medicine, Evanston, IL, USA
| | - Nadia A Lanman
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - O John Semmes
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Nehemiah S Alvarez
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Petra Popovics
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA.
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
12
|
Esposito M, Amory JK, Kang Y. The pathogenic role of retinoid nuclear receptor signaling in cancer and metabolic syndromes. J Exp Med 2024; 221:e20240519. [PMID: 39133222 PMCID: PMC11318670 DOI: 10.1084/jem.20240519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/13/2024] [Accepted: 07/26/2024] [Indexed: 08/13/2024] Open
Abstract
The retinoid nuclear receptor pathway, activated by the vitamin A metabolite retinoic acid, has been extensively investigated for over a century. This study has resulted in conflicting hypotheses about how the pathway regulates health and how it should be pharmaceutically manipulated. These disagreements arise from a fundamental contradiction: retinoid agonists offer clear benefits to select patients with rare bone growth disorders, acute promyelocytic leukemia, and some dermatologic diseases, yet therapeutic retinoid pathway activation frequently causes more harm than good, both through acute metabolic dysregulation and a delayed cancer-promoting effect. In this review, we discuss controlled clinical, mechanistic, and genetic data to suggest several disease settings where inhibition of the retinoid pathway may be a compelling therapeutic strategy, such as solid cancers or metabolic syndromes, and also caution against continued testing of retinoid agonists in cancer patients. Considerable evidence suggests a central role for retinoid regulation of immunity and metabolism, with therapeutic opportunities to antagonize retinoid signaling proposed in cancer, diabetes, and obesity.
Collapse
Affiliation(s)
- Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Kayothera, Inc , Seattle, WA, USA
| | | | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Ludwig Institute for Cancer Research Princeton Branch , Princeton, NJ, USA
| |
Collapse
|
13
|
Lu H, Suo Z, Lin J, Cong Y, Liu Z. Monocyte-macrophages modulate intestinal homeostasis in inflammatory bowel disease. Biomark Res 2024; 12:76. [PMID: 39095853 PMCID: PMC11295551 DOI: 10.1186/s40364-024-00612-x] [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: 05/21/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Monocytes and macrophages play an indispensable role in maintaining intestinal homeostasis and modulating mucosal immune responses in inflammatory bowel disease (IBD). Although numerous studies have described macrophage properties in IBD, the underlying mechanisms whereby the monocyte-macrophage lineage modulates intestinal homeostasis during gut inflammation remain elusive. MAIN BODY In this review, we decipher the cellular and molecular mechanisms governing the generation of intestinal mucosal macrophages and fill the knowledge gap in understanding the origin, maturation, classification, and functions of mucosal macrophages in intestinal niches, particularly the phagocytosis and bactericidal effects involved in the elimination of cell debris and pathogens. We delineate macrophage-mediated immunoregulation in the context of producing pro-inflammatory and anti-inflammatory cytokines, chemokines, toxic mediators, and macrophage extracellular traps (METs), and participating in the modulation of epithelial cell proliferation, angiogenesis, and fibrosis in the intestine and its accessory tissues. Moreover, we emphasize that the maturation of intestinal macrophages is arrested at immature stage during IBD, and the deficiency of MCPIP1 involves in the process via ATF3-AP1S2 signature. In addition, we confirmed the origin potential of IL-1B+ macrophages and defined C1QB+ macrophages as mature macrophages. The interaction crosstalk between the intestine and the mesentery has been described in this review, and the expression of mesentery-derived SAA2 is upregulated during IBD, which contributes to immunoregulation of macrophage. Moreover, we also highlight IBD-related susceptibility genes (e.g., RUNX3, IL21R, GTF2I, and LILRB3) associated with the maturation and functions of macrophage, which provide promising therapeutic opportunities for treating human IBD. CONCLUSION In summary, this review provides a comprehensive, comprehensive, in-depth and novel description of the characteristics and functions of macrophages in IBD, and highlights the important role of macrophages in the molecular and cellular process during IBD.
Collapse
Affiliation(s)
- Huiying Lu
- Department of Gastroenterology, Huaihe Hospital of Henan University, Henan Province, Kaifeng, 475000, China
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Shanghai Tenth People's Hospital of Tongji University, No. 301 Yanchang Road, Shanghai, 200072, China
| | - Zhimin Suo
- Department of Gastroenterology, Huaihe Hospital of Henan University, Henan Province, Kaifeng, 475000, China
| | - Jian Lin
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Shanghai Tenth People's Hospital of Tongji University, No. 301 Yanchang Road, Shanghai, 200072, China
| | - Yingzi Cong
- Division of Gastroenterology and Hepatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Center for Human Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Zhanju Liu
- Center for Inflammatory Bowel Disease Research and Department of Gastroenterology, Shanghai Tenth People's Hospital of Tongji University, No. 301 Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
14
|
Shafiei-Jahani P, Yan S, Kazemi MH, Li X, Akbari A, Sakano K, Sakano Y, Hurrell BP, Akbari O. CB2 stimulation of adipose resident ILC2s orchestrates immune balance and ameliorates type 2 diabetes mellitus. Cell Rep 2024; 43:114434. [PMID: 38963763 PMCID: PMC11317174 DOI: 10.1016/j.celrep.2024.114434] [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: 01/30/2024] [Revised: 05/22/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
Development of type 2 diabetes mellitus (T2DM) is associated with low-grade chronic type 2 inflammation and disturbance of glucose homeostasis. Group 2 innate lymphoid cells (ILC2s) play a critical role in maintaining adipose homeostasis via the production of type 2 cytokines. Here, we demonstrate that CB2, a G-protein-coupled receptor (GPCR) and member of the endocannabinoid system, is expressed on both visceral adipose tissue (VAT)-derived murine and human ILC2s. Moreover, we utilize a combination of ex vivo and in vivo approaches to explore the functional and therapeutic impacts of CB2 engagement on VAT ILC2s in a T2DM model. Our results show that CB2 stimulation of ILC2s protects against insulin-resistance onset, ameliorates glucose tolerance, and reverses established insulin resistance. Our mechanistic studies reveal that the therapeutic effects of CB2 are mediated through activation of the AKT, ERK1/2, and CREB pathways on ILC2s. The results reveal that the CB2 agonist can serve as a candidate for the prevention and treatment of T2DM.
Collapse
Affiliation(s)
- Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shi Yan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Mohammad H Kazemi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Xin Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amitis Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kei Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yoshihiro Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
15
|
Chen Y, Wang J, An C, Bao S, Zhang C. The role and research progress of macrophages after heart transplantation. Heliyon 2024; 10:e33844. [PMID: 39027574 PMCID: PMC11255595 DOI: 10.1016/j.heliyon.2024.e33844] [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: 02/25/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
Since the 60s of the 20th century, heart transplantation has been the best treatment for patients with end-stage heart failure. Due to the increasing number of patients, how to expand the number of donor organs and enhance immune compatibility has become an urgent problem to be solved at this stage. Although current immunosuppression is effective, its side effects are also quite obvious, such as opportunistic infections and malignant tumors. In this review, we focus on the important role in macrophages after heart transplantation and their potential targets for achieving allogeneic graft tolerance, in order to improve effective graft survival and reduce infection and the occurrence of malignant tumors.
Collapse
Affiliation(s)
- Yao Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - JianPeng Wang
- School of First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Cheng An
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - ShanQing Bao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - ChengXin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| |
Collapse
|
16
|
Wen J, Li Z, Lv Y, Ding S, Zhu Y, Yang J, Tang J, Zhu M, Zhao Y, Zhao W. A subunit vaccine based on Brucella rBP26 induces Th1 immune responses and M1 macrophage activation. Acta Biochim Biophys Sin (Shanghai) 2024; 56:879-891. [PMID: 38419498 PMCID: PMC11292127 DOI: 10.3724/abbs.2024023] [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: 12/02/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Brucellosis is a global zoonotic infection caused by Brucella bacteria, which poses a significant burden on society. While transmission prevention is currently the most effective method, the absence of a licenced vaccine for humans necessitates the urgent development of a safe and effective vaccine. Recombinant protein-based subunit vaccines are considered promising options, and in this study, the Brucella BP26 protein is expressed using prokaryotic expression systems. The immune responses are evaluated using the well-established adjuvant CpG-ODN. The results demonstrate that rBP26 supplemented with a CpG adjuvant induces M1 macrophage polarization and stimulates cellular immune responses mediated by Th1 cells and CD8 + T cells. Additionally, it generates high levels of rBP26-specific antibodies in immunized mice. Furthermore, rBP26 immunization activates, proliferates, and produces cytokines in T lymphocytes while also maintaining immune memory for an extended period of time. These findings shed light on the potential biological function of rBP26, which is crucial for understanding brucellosis pathogenesis. Moreover, rBP26 holds promise as an effective subunit vaccine candidate for use in endemic areas.
Collapse
Affiliation(s)
- Jia Wen
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- General Medicine DepartmentGeneral Hospital of Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Zihua Li
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Yongxue Lv
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Shuqin Ding
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Yazhou Zhu
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Jihui Yang
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Jing Tang
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Mingxing Zhu
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Yinqi Zhao
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| | - Wei Zhao
- School of Basic MedicineNingxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious DiseasesNingxia Hui Autonomous RegionYinchuan750004China
| |
Collapse
|
17
|
Huang M, Hu J, Chen Y, Xun Y, Zhang X, Cao Y. Mesencephalic astrocyte-derived neurotrophic factor inhibits cervical cancer progression via regulating macrophage phenotype. Mol Biol Rep 2024; 51:654. [PMID: 38735002 DOI: 10.1007/s11033-024-09602-6] [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: 02/27/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Cervical cancer is a common gynecologic malignant tumor, but the critical factors affecting cervical cancer progression are still not well demonstrated. Mesencephalic astrocyte-derived neurotrophic factor (MANF) has been widely recognized as an anti-inflammatory factor to regulate macrophage polarization. In this study, the effect and mechanism of MANF on cervical cancer were preliminarily explored. METHODS AND RESULTS Kaplan-Meier curve was used to show the overall survival time of the involved cervical cancer patients with high and low MANF expression in cervical cancer tissues. MANF was highly expressed in peritumoral tissues of cervical carcinoma by using immunohistochemistry and western blot. MANF mRNA level was detected by using qRT-PCR. Dual-labeled immunofluorescence showed MANF was mainly expressed in macrophages of cervical peritumoral tissues. Moreover, MANF-silenced macrophages promoted HeLa and SiHa cells survival, migration, invasion and EMT via NF-κB signaling activation. The results of tumor formation in nude mice indicated MANF-silenced macrophages promoted cervical tumor formation in vivo. CONCLUSION Our study reveals an inhibitory role of MANF in cervical cancer progression, indicating MANF as a new and valuable therapeutic target for cervical cancer treatment.
Collapse
Affiliation(s)
- Miaomiao Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jingjing Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No. 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Institute of Translational Medicine, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yueran Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Yingying Xun
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| | - Xinru Zhang
- School of Basic Medical Sciences, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Institute of Translational Medicine, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
18
|
Sargsian S, Mondragón-Palomino O, Lejeune A, Ercelen D, Jin WB, Varghese A, Lim YAL, Guo CJ, Loke P, Cadwell K. Functional characterization of helminth-associated Clostridiales reveals covariates of Treg differentiation. MICROBIOME 2024; 12:86. [PMID: 38730492 PMCID: PMC11084060 DOI: 10.1186/s40168-024-01793-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 03/10/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Parasitic helminths influence the composition of the gut microbiome. However, the microbiomes of individuals living in helminth-endemic regions are understudied. The Orang Asli, an indigenous population in Malaysia with high burdens of the helminth Trichuris trichiura, display microbiotas enriched in Clostridiales, an order of spore-forming obligate anaerobes with immunogenic properties. We previously isolated novel Clostridiales that were enriched in these individuals and found that a subset promoted the Trichuris life cycle. In this study, we aimed to further characterize the functional properties of these bacteria. RESULTS Clostridiales isolates were profiled for their ability to perform 57 enzymatic reactions and produce short-chain fatty acids (SCFAs) and hydrogen sulfide, revealing that these bacteria were capable of a range of activities associated with metabolism and host response. Consistent with this finding, monocolonization of mice with individual isolates identified bacteria that were potent inducers of regulatory T-cell (Treg) differentiation in the colon. Comparisons between variables revealed by these studies identified enzymatic properties correlated with Treg induction and Trichuris egg hatching. CONCLUSION We identified Clostridiales species that are sufficient to induce high levels of Tregs. We also identified a set of metabolic activities linked with Treg differentiation and Trichuris egg hatching mediated by these newly isolated bacteria. Altogether, this study provides functional insights into the microbiotas of individuals residing in a helminth-endemic region. Video Abstract.
Collapse
Affiliation(s)
- Shushan Sargsian
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Octavio Mondragón-Palomino
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alannah Lejeune
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Defne Ercelen
- Department of Medicine, Division of Gastroenterology and Hepatology, New York University Langone Health, New York, NY, 10016, USA
| | - Wen-Bing Jin
- Weill Cornell Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Cornell University, New York, NY, 10021, USA
| | - Alan Varghese
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Yvonne A L Lim
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Chun-Jun Guo
- Weill Cornell Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Cornell University, New York, NY, 10021, USA
| | - P'ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Ken Cadwell
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
19
|
Silver SV, Tucker KJ, Vickman RE, Lanman NA, Semmes OJ, Alvarez NS, Popovics P. PROSTATE CELL HETEROGENEITY AND CXCL17 UPREGULATION IN MOUSE STEROID HORMONE IMBALANCE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590980. [PMID: 38712029 PMCID: PMC11071464 DOI: 10.1101/2024.04.24.590980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Benign prostatic hyperplasia (BPH) is a prevalent age-related condition often characterized by debilitating urinary symptoms. Its etiology is believed to stem from hormonal imbalance, particularly an elevated estradiol-to-testosterone ratio and chronic inflammation. Our previous studies using a mouse steroid hormone imbalance model identified a specific increase in macrophages that migrate and accumulate in the prostate lumen where they differentiate into lipid-laden foam cells in mice implanted with testosterone and estradiol pellets, but not in sham animals. The current study focused on further characterizing the cellular heterogeneity of the prostate in this model as well as identifying the specific transcriptomic signature of the recruited foam cells. Moreover, we aimed to identify the epithelia-derived signals that drive macrophage infiltration and luminal translocation. Male C57BL/6J mice were implanted with slow-release testosterone and estradiol pellets (T+E2) and harvested the ventral prostates two weeks later for scRNA-seq analysis, or performed sham surgery. We identified Ear2+ and Cd72+ macrophages that were elevated in response to steroid hormone imbalance, whereas a Mrc1+ resident macrophage population did not change. In addition, an Spp1+ foam cell cluster was almost exclusively found in T+E2 mice. Further markers of foam cells were also identified, including Gpnmb and Trem2, and GPNMB was confirmed as a novel histological marker with immunohistochemistry. Foam cells were also shown to express known pathological factors Vegf, Tgfb1, Ccl6, Cxcl16 and Mmp12. Intriguingly, a screen for chemokines identified the upregulation of epithelial-derived Cxcl17, a known monocyte attractant, in T+E2 prostates suggesting that it might be responsible for the elevated macrophage number as well as their translocation to the lumen. Our study identified macrophage subsets that respond to steroid hormone imbalance as well as further confirmed a potential pathological role of luminal foam cells in the prostate. These results underscore a pathological role of the identified prostate foam cells and suggests CXCL17-mediated macrophage migration as a critical initiating event.
Collapse
Affiliation(s)
- Samara V. Silver
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA
| | - Kayah J. Tucker
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA
| | - Renee E Vickman
- Department of Surgery, Endeavor Health, an Academic Affiliate of the University of Chicago Pritzker School of Medicine, Evanston, IL, USA
| | - Nadia A. Lanman
- Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - O John Semmes
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA
| | - Nehemiah S. Alvarez
- Department of Surgery, Endeavor Health, an Academic Affiliate of the University of Chicago Pritzker School of Medicine, Evanston, IL, USA
| | - Petra Popovics
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA
| |
Collapse
|
20
|
Selvakumar B, Sekar P, Samsudin AR. Intestinal macrophages in pathogenesis and treatment of gut leakage: current strategies and future perspectives. J Leukoc Biol 2024; 115:607-619. [PMID: 38198217 DOI: 10.1093/jleuko/qiad165] [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: 07/02/2023] [Revised: 11/13/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Macrophages play key roles in tissue homeostasis, defense, disease, and repair. Macrophages are highly plastic and exhibit distinct functional phenotypes based on micro-environmental stimuli. In spite of several advancements in understanding macrophage biology and their different functional phenotypes in various physiological and pathological conditions, currently available treatment strategies targeting macrophages are limited. Macrophages' high plasticity and diverse functional roles-including tissue injury and wound healing mechanisms-mark them as potential targets to mine for efficient therapeutics to treat diseases. Despite mounting evidence on association of gut leakage with several extraintestinal diseases, there is no targeted standard therapy to treat gut leakage. Therefore, there is an urgent need to develop therapeutic strategies to treat this condition. Macrophages are the cells that play the largest role in interacting with the gut microbiota in the intestinal compartment and exert their intended functions in injury and repair mechanisms. In this review, we have summarized the current knowledge on the origins and phenotypes of macrophages. The specific role of macrophages in intestinal barrier function, their role in tissue repair mechanisms, and their association with gut microbiota are discussed. In addition, currently available therapies and the putative tissue repair mediators of macrophages for treating microbiota dysbiosis induced gut leakage are also discussed. The overall aim of this review is to convey the intense need to screen for microbiota induced macrophage-released prorepair mediators, which could lead to the identification of potential candidates that could be developed for treating the leaky gut and associated diseases.
Collapse
Affiliation(s)
- Balachandar Selvakumar
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Priyadharshini Sekar
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - A Rani Samsudin
- Department of Microbiota, Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
21
|
Balakrishnan R, Subbarayan R, Shrestha R, Chauhan A, Krishnamoorthy L. Exploring platelet-derived microvesicles in vascular regeneration: unraveling the intricate mechanisms and molecular mediators. Mol Biol Rep 2024; 51:393. [PMID: 38446325 DOI: 10.1007/s11033-024-09302-1] [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: 12/22/2023] [Accepted: 01/30/2024] [Indexed: 03/07/2024]
Abstract
Microvesicles (MVs) serve as biomarkers and transmitters for cell communication and also act as essential contributors to diseases. Platelets release microvesicles when activated voluntarily, making them a significant source. Platelet-derived microvesicles possess a range of characteristics similar to their parent cells and were shown to exert regulatory impacts on vascular and immunological cells. MVs can alter the activity of recipient cells by transferring their internal components. Furthermore, it has been identified that microvesicles derived from platelets possess the ability to exert immunomodulatory effects on different kinds of cells. Recent research has shown that microvesicles have a bidirectional influence of harming and preventing the receptor cells. Nevertheless, the specific characteristics of the active molecules responsible for this phenomenon are still unknown. The primary focus of this review was to explore the mechanism of vascular tissue regeneration and the specific molecules that play a role in mediating various biological effects throughout this process. These molecules exert their effects by influencing autophagy, apoptosis, and inflammatory pathways.
Collapse
Affiliation(s)
- Ranjith Balakrishnan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Rajasekaran Subbarayan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India.
| | | | - Ankush Chauhan
- Faculty of Allied Health Sciences, Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Loganathan Krishnamoorthy
- FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| |
Collapse
|
22
|
Pinos I, Coronel J, Albakri A, Blanco A, McQueen P, Molina D, Sim J, Fisher EA, Amengual J. β-Carotene accelerates the resolution of atherosclerosis in mice. eLife 2024; 12:RP87430. [PMID: 38319073 PMCID: PMC10945528 DOI: 10.7554/elife.87430] [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: 02/07/2024] Open
Abstract
β-Carotene oxygenase 1 (BCO1) catalyzes the cleavage of β-carotene to form vitamin A. Besides its role in vision, vitamin A regulates the expression of genes involved in lipid metabolism and immune cell differentiation. BCO1 activity is associated with the reduction of plasma cholesterol in humans and mice, while dietary β-carotene reduces hepatic lipid secretion and delays atherosclerosis progression in various experimental models. Here we show that β-carotene also accelerates atherosclerosis resolution in two independent murine models, independently of changes in body weight gain or plasma lipid profile. Experiments in Bco1-/- mice implicate vitamin A production in the effects of β-carotene on atherosclerosis resolution. To explore the direct implication of dietary β-carotene on regulatory T cells (Tregs) differentiation, we utilized anti-CD25 monoclonal antibody infusions. Our data show that β-carotene favors Treg expansion in the plaque, and that the partial inhibition of Tregs mitigates the effect of β-carotene on atherosclerosis resolution. Our data highlight the potential of β-carotene and BCO1 activity in the resolution of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Ivan Pinos
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Johana Coronel
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Asma'a Albakri
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Amparo Blanco
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Patrick McQueen
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Donald Molina
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| | - JaeYoung Sim
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| | - Edward A Fisher
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, New York University Grossman School of Medicine, NYU Langone Medical CenterNew YorkUnited States
| | - Jaume Amengual
- Division of Nutritional Sciences, University of Illinois Urbana ChampaignUrbanaUnited States
- Department of Food Science and Human Nutrition, University of Illinois Urbana ChampaignUrbanaUnited States
| |
Collapse
|
23
|
Hamley M, Leyk S, Casar C, Liebold I, Jawazneh AA, Lanzloth C, Böttcher M, Haas H, Richardt U, Rothlin CV, Jacobs T, Huber S, Adlung L, Pelczar P, Henao-Mejia J, Bosurgi L. Nmes1 is a novel regulator of mucosal response influencing intestinal healing potential. Eur J Immunol 2024; 54:e2350434. [PMID: 37971166 DOI: 10.1002/eji.202350434] [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: 02/15/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/19/2023]
Abstract
The initiation of tissue remodeling following damage is a critical step in preventing the development of immune-mediated diseases. Several factors contribute to mucosal healing, leading to innovative therapeutic approaches for managing intestinal disorders. However, uncovering alternative targets and gaining mechanistic insights are imperative to enhance therapy efficacy and broaden its applicability across different intestinal diseases. Here we demonstrate that Nmes1, encoding for Normal Mucosa of Esophagus-Specific gene 1, also known as Aa467197, is a novel regulator of mucosal healing. Nmes1 influences the macrophage response to the tissue remodeling cytokine IL-4 in vitro. In addition, using two murine models of intestinal damage, each characterized by a type 2-dominated environment with contrasting functions, the ablation of Nmes1 results in decreased intestinal regeneration during the recovery phase of colitis, while enhancing parasitic egg clearance and reducing fibrosis during the advanced stages of Schistosoma mansoni infection. These outcomes are associated with alterations in CX3CR1+ macrophages, cells known for their wound-healing potential in the inflamed colon, hence promising candidates for cell therapies. All in all, our data indicate Nmes1 as a novel contributor to mucosal healing, setting the basis for further investigation into its potential as a new target for the treatment of colon-associated inflammation.
Collapse
Affiliation(s)
- Madeleine Hamley
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stephanie Leyk
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Christian Casar
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Bioinformatics Core, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Imke Liebold
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Amirah Al Jawazneh
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Clarissa Lanzloth
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Marius Böttcher
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Ulricke Richardt
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Carla V Rothlin
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Thomas Jacobs
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lorenz Adlung
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Penelope Pelczar
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jorge Henao-Mejia
- The Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lidia Bosurgi
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
24
|
Zhao M, Jankovic D, Hornick KM, Link VM, Souza COS, Belkaid Y, Lack J, Loke P. Genetic variation in IL-4 activated tissue resident macrophages alters the epigenetic state to determine strain specific synergistic responses to LPS. RESEARCH SQUARE 2024:rs.3.rs-3759654. [PMID: 38712032 PMCID: PMC11071541 DOI: 10.21203/rs.3.rs-3759654/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
How macrophages in the tissue environment integrate multiple stimuli will depend on the genetic background of the host, but this is poorly understood. Here, we investigated C57BL/6 and BALB/c strain specific in vivo IL-4 activation of tissue-resident macrophages (TRMs) from the peritoneal cavity. C57BL/6 TRMs are more transcriptionally responsive to IL-4 stimulation, with a greater association of induced genes with super enhancers, induced enhancers, and topologically associating domains (TAD) boundaries. IL-4-directed epigenomic remodeling revealed BL/6 specific enrichment of NF-κB, IRF, and STAT motifs. Additionally, IL-4-activated BL/6 TRMs demonstrated an augmented synergistic response upon in vitro lipopolysaccharide (LPS) exposure compared to BALB/c TRMs, despite naïve BALB/c TRMs displaying a more robust transcriptional response to LPS than naïve BL/6 TRMs. Single-cell RNA sequencing (scRNA-seq) analysis of mixed bone marrow chimeric mice indicated that transcriptional differences between BL/6 and BALB/c TRMs, and synergy between IL-4 and LPS, are cell intrinsic within the same tissue environment. Hence, genetic variation alters IL-4-induced cell intrinsic epigenetic reprogramming resulting in strain specific synergistic responses to LPS exposure.
Collapse
Affiliation(s)
- Mingming Zhao
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Dragana Jankovic
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Katherine M. Hornick
- NIAID Collaborative Bioinformatics Resource, Integrated Data Sciences Section, Research Technology Branch, Division of Intramural Research, NIAID, NIH, Bethesda, MD 20892, USA
| | - Verena M. Link
- Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Camila Oliveira Silva Souza
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Yasmine Belkaid
- Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource, Integrated Data Sciences Section, Research Technology Branch, Division of Intramural Research, NIAID, NIH, Bethesda, MD 20892, USA
| | - P’ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
25
|
Pinos I, Coronel J, Albakri A, Blanco A, McQueen P, Molina D, Sim J, Fisher EA, Amengual J. β-carotene accelerates the resolution of atherosclerosis in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.07.531563. [PMID: 36945561 PMCID: PMC10028884 DOI: 10.1101/2023.03.07.531563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
β-carotene oxygenase 1 (BCO1) catalyzes the cleavage of β-carotene to form vitamin A. Besides its role in vision, vitamin A regulates the expression of genes involved in lipid metabolism and immune cell differentiation. BCO1 activity is associated with the reduction of plasma cholesterol in humans and mice, while dietary β-carotene reduces hepatic lipid secretion and delays atherosclerosis progression in various experimental models. Here we show that β-carotene also accelerates atherosclerosis resolution in two independent murine models, independently of changes in body weight gain or plasma lipid profile. Experiments in Bco1-/- mice implicate vitamin A production in the effects of β-carotene on atherosclerosis resolution. To explore the direct implication of dietary β-carotene on regulatory T cells (Tregs) differentiation, we utilized anti-CD25 monoclonal antibody infusions. Our data show that β-carotene favors Treg expansion in the plaque, and that the partial inhibition of Tregs mitigates the effect of β-carotene on atherosclerosis resolution. Our data highlight the potential of β-carotene and BCO1 activity in the resolution of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Ivan Pinos
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL
| | - Johana Coronel
- Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL
| | - Asma'a Albakri
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL
| | - Amparo Blanco
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL
| | - Patrick McQueen
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL
| | - Donald Molina
- Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL
| | - JaeYoung Sim
- Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL
| | - Edward A Fisher
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, New York University Grossman School of Medicine, NYU Langone Medical Center, NY
| | - Jaume Amengual
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL
- Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL
| |
Collapse
|
26
|
Han J, Gallerand A, Erlich EC, Helmink BA, Mair I, Li X, Eckhouse SR, Dimou FM, Shakhsheer BA, Phelps HM, Chan MM, Mintz RL, Lee DD, Schilling JD, Finlay CM, Allen JE, Jakubzick CV, Else KJ, Onufer EJ, Zhang N, Randolph GJ. Human serous cavity macrophages and dendritic cells possess counterparts in the mouse with a distinct distribution between species. Nat Immunol 2024; 25:155-165. [PMID: 38102487 PMCID: PMC10990619 DOI: 10.1038/s41590-023-01688-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 10/20/2023] [Indexed: 12/17/2023]
Abstract
In mouse peritoneal and other serous cavities, the transcription factor GATA6 drives the identity of the major cavity resident population of macrophages, with a smaller subset of cavity-resident macrophages dependent on the transcription factor IRF4. Here we showed that GATA6+ macrophages in the human peritoneum were rare, regardless of age. Instead, more human peritoneal macrophages aligned with mouse CD206+ LYVE1+ cavity macrophages that represent a differentiation stage just preceding expression of GATA6. A low abundance of CD206+ macrophages was retained in C57BL/6J mice fed a high-fat diet and in wild-captured mice, suggesting that differences between serous cavity-resident macrophages in humans and mice were not environmental. IRF4-dependent mouse serous cavity macrophages aligned closely with human CD1c+CD14+CD64+ peritoneal cells, which, in turn, resembled human peritoneal CD1c+CD14-CD64- cDC2. Thus, major populations of serous cavity-resident mononuclear phagocytes in humans and mice shared common features, but the proportions of different macrophage differentiation stages greatly differ between the two species, and dendritic cell (DC2)-like cells were especially prominent in humans.
Collapse
Affiliation(s)
- Jichang Han
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexandre Gallerand
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Emma C Erlich
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Beth A Helmink
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Iris Mair
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Xin Li
- Departments of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Shaina R Eckhouse
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Francesca M Dimou
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Baddr A Shakhsheer
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah M Phelps
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Mandy M Chan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel L Mintz
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel D Lee
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joel D Schilling
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Conor M Finlay
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Judith E Allen
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Wellcome Trust Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Claudia V Jakubzick
- Departments of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Kathryn J Else
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emily J Onufer
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Nan Zhang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Ellen and Ronald Caplan Cancer Center at the Wistar Institute in Philadelphia, Philadelphia, PA, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
27
|
Ren X, Hou Z, Pang B, Gao C, Tang R. Photosynthetic and Self-Draining Biohybrid Dressing for Accelerating Healing of Diabetic Wound. Adv Healthc Mater 2024; 13:e2302287. [PMID: 37924323 DOI: 10.1002/adhm.202302287] [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: 07/19/2023] [Revised: 10/20/2023] [Indexed: 11/06/2023]
Abstract
Wound healing is a well-orchestrated progress associated with angiogenesis, epithelialization, inflammatory status, and infection control, whereas these processes are seriously disturbed in diabetic wounds. In this study, a biohybrid dressing integrating the inherent ability of Bromeliad leaf (photosynthesis and self-draining) with the therapeutic effect of artificial materials (glucose-degrading and ROS-scavenging) is presented. The dressing consists of double-layered structures as follows: 1) Outer layer, a Bromeliad leaf substrate full of alginate hydrogel-immobilized glucose oxidase (GOx@Alg@Bromeliad substrate, abbreviated as BGA), can generate oxygen to guarantee the GOx-catalyzed glucose oxidation by photosynthesis, reducing local hyperglycemia to stabilize hypoxia inducible factor-1 alpha (HIF-1α) for angiogenesis and producing hydrogen peroxide for killing bacteria on the surface of wound tissue. The sophisticated structure of the leaf drains excessive exudate away via transpiration-mimicking, preventing skin maceration and impeding bacterial growth. 2) Inner layer, microneedles containing catalase (CAT-HA MNs, abbreviated as CHM), reduces excessive oxidative stress in the tissue to promote the proliferation of fibroblasts and inhibits proinflammatory polarization of macrophages, improving re-epithelialization of diabetic wounds. Together, the biohybrid dressing (BGA-CHM, abbreviated as BCHM) can enhance angiogenesis, strengthen re-epithelialization, alleviate chronic inflammation, and suppress bacterial infection, providing a promising strategy for diabetic wound therapy.
Collapse
Affiliation(s)
- Xinyu Ren
- School of Stomatology, Lanzhou University, Lanzhou, 730000, P. R. China
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, Lanzhou, 730000, P. R. China
| | - Zhiming Hou
- School of Stomatology, Lanzhou University, Lanzhou, 730000, P. R. China
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, Lanzhou, 730000, P. R. China
| | - Bo Pang
- School of Stomatology, Lanzhou University, Lanzhou, 730000, P. R. China
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, Lanzhou, 730000, P. R. China
| | - Cen Gao
- School of Stomatology, Lanzhou University, Lanzhou, 730000, P. R. China
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, Lanzhou, 730000, P. R. China
| | - Rongbing Tang
- School of Stomatology, Lanzhou University, Lanzhou, 730000, P. R. China
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, Gansu Province, Lanzhou, 730000, P. R. China
| |
Collapse
|
28
|
Oates TCL, Moura PL, Cross S, Roberts K, Baum HE, Haydn‐Smith KL, Wilson MC, Heesom KJ, Severn CE, Toye AM. Defining the proteomic landscape of cultured macrophages and their polarization continuum. Immunol Cell Biol 2023; 101:947-963. [PMID: 37694300 PMCID: PMC10953363 DOI: 10.1111/imcb.12687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/28/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023]
Abstract
Macrophages have previously been characterized based on phenotypical and functional differences into suggested simplified subtypes of MØ, M1, M2a and M2c. These macrophage subtypes can be generated in a well-established primary monocyte culture model that produces cells expressing accepted subtype surface markers. To determine how these subtypes retain functional similarities and better understand their formation, we generated all four subtypes from the same donors. Comparative whole-cell proteomics confirmed that four distinct macrophage subtypes could be induced from the same donor material, with > 50% of 5435 identified proteins being significantly altered in abundance between subtypes. Functional assessment highlighted that these distinct protein expression profiles are primed to enable specific cell functions, indicating that this shifting proteome is predictive of meaningful changes in cell characteristics. Importantly, the 2552 proteins remained consistent in abundance across all macrophage subtypes examined, demonstrating maintenance of a stable core proteome that likely enables swift polarity changes. We next explored the cross-polarization capabilities of preactivated M1 macrophages treated with dexamethasone. Importantly, these treated cells undergo a partial repolarization toward the M2c surface markers but still retain the M1 functional phenotype. Our investigation of polarized macrophage subtypes therefore provides evidence of a sliding scale of macrophage functionality, with these data sets providing a valuable benchmark resource for further studies of macrophage polarity, with relevance for cell therapy development and drug discovery.
Collapse
Affiliation(s)
- Tiah CL Oates
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| | - Pedro L Moura
- Center for Haematology and Regenerative Medicine, Department of Medicine (MedH)Karolinska InstitutetHuddingeSweden
| | | | - Kiren Roberts
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | - Holly E Baum
- Max Planck Bristol Centre for Minimal Biology, School of ChemistryUniversity of BristolBristolUK
| | - Katy L Haydn‐Smith
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | | | - Kate J Heesom
- Proteomics Facility, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | - Charlotte E Severn
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| |
Collapse
|
29
|
Dietschmann A, Ruhl A, Murray PJ, Günther C, Becker C, Fallon P, Voehringer D. Th2-dependent disappearance and phenotypic conversion of mouse alveolar macrophages. Eur J Immunol 2023; 53:e2350475. [PMID: 37452620 DOI: 10.1002/eji.202350475] [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: 03/08/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Alveolar macrophages (alvMs) play an important role for maintenance of lung function by constant removal of cellular debris in the alveolar space. They further contribute to defense against microbial or viral infections and limit tissue damage during acute lung injury. alvMs arise from embryonic progenitor cells, seed the alveoli before birth, and have life-long self-renewing capacity. However, recruited monocytes may also help to restore the alvM population after depletion caused by toxins or influenza virus infection. At present, the population dynamics and cellular plasticity of alvMs during allergic lung inflammation is poorly defined. To address this point, we used a mouse model of Aspergillus fumigatus-induced allergic lung inflammation and observed that Th2-derived IL-4 and IL-13 caused almost complete disappearance of alvMs. This effect required STAT6 expression in alvMs and also occurred in various other settings of type 2 immunity-mediated lung inflammation or administration of IL-4 complexes to the lung. In addition, Th2 cells promoted conversion of alvMs to alternatively activated macrophages and multinucleated giant cells. Given the well-established role of alvMs for maintenance of lung function, this process may have implications for resolution of inflammation and tissue homeostasis in allergic asthma.
Collapse
Affiliation(s)
- Axel Dietschmann
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Andreas Ruhl
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Peter J Murray
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Claudia Günther
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Padraic Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
30
|
Zarek CM, Dende C, Coronado J, Pendse M, Dryden P, Hooper LV, Reese TA. Preexisting helminth challenge exacerbates infection and reactivation of gammaherpesvirus in tissue resident macrophages. PLoS Pathog 2023; 19:e1011691. [PMID: 37847677 PMCID: PMC10581490 DOI: 10.1371/journal.ppat.1011691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/17/2023] [Indexed: 10/19/2023] Open
Abstract
Even though gammaherpesvirus and parasitic infections are endemic in parts of the world, there is a lack of understanding about the outcome of coinfection. In humans, coinfections usually occur sequentially, with fluctuating order and timing in different hosts. However, experimental studies in mice generally do not address the variables of order and timing of coinfections. We sought to examine the variable of coinfection order in a system of gammaherpesvirus-helminth coinfection. Our previous work demonstrated that infection with the intestinal parasite, Heligmosomoides polygyrus, induced transient reactivation from latency of murine gammaherpesvirus-68 (MHV68). In this report, we reverse the order of coinfection, infecting with H. polygyrus first, followed by MHV68, and examined the effects of preexisting parasite infection on MHV68 acute and latent infection. We found that preexisting parasite infection increased the propensity of MHV68 to reactivate from latency. However, when we examined the mechanism for reactivation, we found that preexisting parasite infection increased the ability of MHV68 to reactivate in a vitamin A dependent manner, a distinct mechanism to what we found previously with parasite-induced reactivation after latency establishment. We determined that H. polygyrus infection increased both acute and latent MHV68 infection in a population of tissue resident macrophages, called large peritoneal macrophages. We demonstrate that this population of macrophages and vitamin A are required for increased acute and latent infection during parasite coinfection.
Collapse
Affiliation(s)
- Christina M. Zarek
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Chaitanya Dende
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jaime Coronado
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Mihir Pendse
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Phillip Dryden
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Lora V. Hooper
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- The Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Tiffany A. Reese
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
31
|
Aronova A, Tosato F, Naser N, Asare Y. Innate Immune Pathways in Atherosclerosis-From Signaling to Long-Term Epigenetic Reprogramming. Cells 2023; 12:2359. [PMID: 37830572 PMCID: PMC10571887 DOI: 10.3390/cells12192359] [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: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Innate immune pathways play a crucial role in the development of atherosclerosis, from sensing initial danger signals to the long-term reprogramming of immune cells. Despite the success of lipid-lowering therapy, anti-hypertensive medications, and other measures in reducing complications associated with atherosclerosis, cardiovascular disease (CVD) remains the leading cause of death worldwide. Consequently, there is an urgent need to devise novel preventive and therapeutic strategies to alleviate the global burden of CVD. Extensive experimental research and epidemiological studies have demonstrated the dominant role of innate immune mechanisms in the progression of atherosclerosis. Recently, landmark trials including CANTOS, COLCOT, and LoDoCo2 have provided solid evidence demonstrating that targeting innate immune pathways can effectively reduce the risk of CVD. These groundbreaking trials mark a significant paradigm shift in the field and open new avenues for atheroprotective treatments. It is therefore crucial to comprehend the intricate interplay between innate immune pathways and atherosclerosis for the development of targeted therapeutic interventions. Additionally, unraveling the mechanisms underlying long-term reprogramming may offer novel strategies to reverse the pro-inflammatory phenotype of immune cells and restore immune homeostasis in atherosclerosis. In this review, we present an overview of the innate immune pathways implicated in atherosclerosis, with a specific focus on the signaling pathways driving chronic inflammation in atherosclerosis and the long-term reprogramming of immune cells within atherosclerotic plaque. Elucidating the molecular mechanisms governing these processes presents exciting opportunities for the development of a new class of immunotherapeutic approaches aimed at reducing inflammation and promoting plaque stability. By addressing these aspects, we can potentially revolutionize the management of atherosclerosis and its associated cardiovascular complications.
Collapse
Affiliation(s)
| | | | | | - Yaw Asare
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), 80539 Munich, Germany
| |
Collapse
|
32
|
Attiogbe E, Larochelle S, Chaib Y, Mainzer C, Mauroux A, Bordes S, Closs B, Gilbert C, Moulin VJ. An in vitro autologous, vascularized, and immunocompetent Tissue Engineered Skin model obtained by the self-assembled approach. Acta Biomater 2023; 168:361-371. [PMID: 37419164 DOI: 10.1016/j.actbio.2023.06.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023]
Abstract
A complete in vitro skin model, containing resident cell types is needed to understand physiology and to consider the role of immune and endothelial cells in dermal drug testing. In this study, a cell extraction technique was developed to isolate resident skin cells from the same human donor while preserving the immune and endothelial cells. Then those cells were used to reconstruct an autologous, vascularized, and immunocompetent Tissue-Engineered Skin model, aviTES. Phenotypic characterization of the viable cells was performed on freshly isolated cells and after thawing through flow cytometry. Dermal cell extracts were characterized as fibroblasts, endothelial and immune cells, and the average amount of each cell type represents 4, 0.5, and 1 million viable cells per g of the dermis, respectively. The 3D models, TES and aviTES, were characterized by a fully differentiated epidermis that showed an increase in the presence of Ki67+ cells in the basolateral layer of the aviTES model. Capillary-like network formation, through the self-assembly of endothelial cells, and the presence of functional immune cells were identified through immunofluorescence staining in aviTES. In addition, the aviTES model was immunocompetent, as evidenced by its capacity to increase the production of pro-inflammatory cytokines TNF-α, MIP-1α, and GM-CSF following LPS stimulation. This study describes an autologous skin model containing a functional resident skin immune system and a capillary network. It provides a relevant tool to study the contribution of the immune system to skin diseases and inflammatory responses and to investigate resident skin cell interactions and drug development. STATEMENT OF SIGNIFICANCE: There is an urgent need for a complete in vitro skin model containing the resident cell types to better understand the role of immune and endothelial cells in skin and to be able to use it for drug testing. Actual 3D models of human skin most often contain only fibroblasts and keratinocytes with a limited number of models containing endothelial cells or a limited variety of immune cells. This study describes an autologous skin model containing a functional resident skin immune system and a capillary network. It provides a relevant tool to study the contribution of the immune system to skin diseases and inflammatory responses and to investigate interactions between resident skin cell, improving our capacity to develop new drugs.
Collapse
Affiliation(s)
- Emilie Attiogbe
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Sébastien Larochelle
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Yanis Chaib
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | | | | | | | | | - Caroline Gilbert
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Véronique J Moulin
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
33
|
Zheng H, Cheng X, Jin L, Shan S, Yang J, Zhou J. Recent advances in strategies to target the behavior of macrophages in wound healing. Biomed Pharmacother 2023; 165:115199. [PMID: 37517288 DOI: 10.1016/j.biopha.2023.115199] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 08/01/2023] Open
Abstract
Chronic wounds and scar formation are widespread due to limited suitable remedies. The macrophage is a crucial regulator in wound healing, controlling the onset and termination of inflammation and regulating other processes related to wound healing. The current breakthroughs in developing new medications and drug delivery methods have enabled the accurate targeting of macrophages in oncology and rheumatic disease therapies through clinical trials. These successes have cleared the way to utilize drugs targeting macrophages in various disorders. This review thus summarizes macrophage involvement in normal and pathologic wound healing. It further details the targets available for macrophage intervention and therapeutic strategies for targeting the behavior of macrophages in tissue repair and regeneration.
Collapse
Affiliation(s)
- Hongkun Zheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xinwei Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lu Jin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Jia Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
34
|
Huang T, Zhang Q, Yi J, Wang R, Zhang Z, Luo P, Zeng R, Wang Y, Tu M. PEG-Sheddable Nanodrug Remodels Tumor Microenvironment to Promote Effector T Cell Infiltration and Revise Their Exhaustion for Breast Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301749. [PMID: 37211704 DOI: 10.1002/smll.202301749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/30/2023] [Indexed: 05/23/2023]
Abstract
Low infiltration of cytotoxic T lymphocytes and their exhaustion manifest the two concurrent main hurdles for achieving effective tumor immunotherapy of triple-negative breast cancer. It is found that Galectin-9 blockage can revise the exhaustion of effector T cells, meanwhile the repolarization of protumoral M2 tumor-associated macrophages (TAMs) into tumoricidal M1-like ones can recruit effector T cells infiltrating into tumor to boost immune responses. Herein, a sheddable PEG-decorated and M2-TAMs targeted nanodrug incorporating Signal Transducer and Activator of Transcription 6 inhibitor (AS) and anti-Galectin-9 antibody (aG-9) is prepared. The nanodrug responds to acidic tumor microenvironment (TME) with the shedding of PEG corona and the release of aG-9, exerting local blockade of PD-1/Galectin-9/TIM-3 interaction to augment effector T cells via exhaustion reversing. Synchronously, targeted repolarization of M2-TAMs into M1 phenotype by AS-loaded nanodrug is achieved, which promotes tumor infiltration of effector T cells and thus synergizes with aG-9 blockade to boost the therapeutic efficacy. Besides, the PEG-sheddable approach endows nanodrug with stealth ability to reduce immune-related adverse effects caused by AS and aG-9. This PEG sheddable nanodrug holds the potential to reverse the immunosuppressive TME and increase effector T cell infiltration, which dramatically enhances immunotherapy in highly malignant breast cancer.
Collapse
Affiliation(s)
- Tao Huang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Qiaoyun Zhang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Jing Yi
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Rongze Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Zekun Zhang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Pin Luo
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Rong Zeng
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Yong Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Mei Tu
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
35
|
Prat M, Coulson K, Blot C, Jacquemin G, Romano M, Renoud ML, AlaEddine M, Le Naour A, Authier H, Rahabi MC, Benmoussa K, Salon M, Parny M, Delord JP, Ferron G, Lefèvre L, Couderc B, Coste A. PPARγ activation modulates the balance of peritoneal macrophage populations to suppress ovarian tumor growth and tumor-induced immunosuppression. J Immunother Cancer 2023; 11:e007031. [PMID: 37586764 PMCID: PMC10432661 DOI: 10.1136/jitc-2023-007031] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Ovarian adenocarcinoma (OVAD) frequently metastasizes to the peritoneal cavity and manifests by the formation of ascites, which constitutes a tumor-promoting microenvironment. In the peritoneal cavity, two developmentally, phenotypically and functionally distinct macrophage subsets, immunocompetent large peritoneal macrophages (LPM) and immunosuppressive small peritoneal macrophages (SPM), coexist. Because peroxisome proliferator-activated receptor γ (PPARγ) is a critical factor participating in macrophage differentiation and cooperates with CCAAT/enhancer binding protein β (C/EBPβ), a transcription factor essential for SPM-to-LPM differentiation, PPARγ could be also involved in the regulation of SPM/LPM balance and could be a promising therapeutic target. METHODS To evaluate the 15(S)-hydroxyeicosatetraenoic acid (HETE), a PPARγ endogenous ligand, impact on ovarian tumor growth, we intraperitoneally injected 15(S)-HETE into a murine ovarian cancer model. This experimental model consists in the intraperitoneally injection of ID8 cells expressing luciferase into syngeneic C57BL/6 female mice. This ID8 orthotopic mouse model is a well-established experimental model of end-stage epithelial OVAD. Tumor progression was monitored using an in vivo imaging system. Peritoneal immune cells in ascites were analyzed by flow cytometry and cell sorting. To determine whether the impact of 15(S)-HETE in tumor development is mediated through the macrophages, these cells were depleted by injection of liposomal clodronate. To further dissect how 15(S)-HETE mediated its antitumor effect, we assessed the tumor burden in tumor-bearing mice in which the PPARγ gene was selectively disrupted in myeloid-derived cells and in mice deficient of the recombination-activating gene Rag2. Finally, to validate our data in humans, we isolated and treated macrophages from ascites of individuals with OVAD. RESULTS Here we show, in the murine experimental model of OVAD, that 15(S)-HETE treatment significantly suppresses the tumor growth, which is associated with the differentiation of SPM into LPM and the LPM residency in the peritoneal cavity. We demonstrate that C/EBPβ and GATA6 play a central role in SPM-to-LPM differentiation and in LPM peritoneal residence through PPARγ activation during OVAD. Moreover, this SPM-to-LPM switch is associated with the increase of the effector/regulatory T-cell ratio. Finally, we report that 15(S)-HETE attenuates immunosuppressive properties of human ovarian tumor-associated macrophages from ascites. CONCLUSION Altogether, these results promote PPARγ as a potential therapeutic target to restrain OVAD development and strengthen the use of PPARγ agonists in anticancer therapy.
Collapse
Affiliation(s)
- Mélissa Prat
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Kimberley Coulson
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Clément Blot
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Godefroy Jacquemin
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Mathilde Romano
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Marie-Laure Renoud
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Mohamad AlaEddine
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Augustin Le Naour
- UMR1037 Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse, INSERM, Toulouse, France
| | - Hélène Authier
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Mouna Chirine Rahabi
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Khaddouj Benmoussa
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Marie Salon
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Mélissa Parny
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | | | - Gwenaël Ferron
- Institut Claudius Regaud, IUCT Oncopole, Toulouse, France
| | - Lise Lefèvre
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| | - Bettina Couderc
- UMR1037 Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse, INSERM, Toulouse, France
- Institut Claudius Regaud, IUCT Oncopole, Toulouse, France
| | - Agnès Coste
- RESTORE Research Center, Université de Toulouse, INSERM-1301, CNRS-5070, EFS, ENVT, Toulouse, France
| |
Collapse
|
36
|
Korotkaja K, Jansons J, Spunde K, Rudevica Z, Zajakina A. Establishment and Characterization of Free-Floating 3D Macrophage Programming Model in the Presence of Cancer Cell Spheroids. Int J Mol Sci 2023; 24:10763. [PMID: 37445941 DOI: 10.3390/ijms241310763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Reprogramming of tumor-associated macrophages (TAMs) is a promising strategy for cancer immunotherapy. Several studies have shown that cancer cells induce/support the formation of immunosuppressive TAMs phenotypes. However, the specific factors that orchestrate this immunosuppressive process are unknown or poorly studied. In vivo studies are expensive, complex, and ethically constrained. Therefore, 3D cell interaction models could become a unique framework for the identification of important TAMs programming factors. In this study, we have established and characterized a new in vitro 3D model for macrophage programming in the presence of cancer cell spheroids. First, it was demonstrated that the profile of cytokines, chemokines, and surface markers of 3D-cultured macrophages did not differ conceptually from monolayer-cultured M1 and M2-programmed macrophages. Second, the possibility of reprogramming macrophages in 3D conditions was investigated. In total, the dynamic changes in 6 surface markers, 11 cytokines, and 22 chemokines were analyzed upon macrophage programming (M1 and M2) and reprogramming (M1→M2 and M2→M1). According to the findings, the reprogramming resulted in a mixed macrophage phenotype that expressed both immunosuppressive and anti-cancer immunostimulatory features. Third, cancer cell spheroids were shown to stimulate the production of immunosuppressive M2 markers as well as pro-tumor cytokines and chemokines. In summary, the newly developed 3D model of cancer cell spheroid/macrophage co-culture under free-floating conditions can be used for studies on macrophage plasticity and for the development of targeted cancer immunotherapy.
Collapse
Affiliation(s)
- Ksenija Korotkaja
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, k.1, LV-1067 Riga, Latvia
| | - Juris Jansons
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, k.1, LV-1067 Riga, Latvia
| | - Karina Spunde
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, k.1, LV-1067 Riga, Latvia
| | - Zhanna Rudevica
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, k.1, LV-1067 Riga, Latvia
| | - Anna Zajakina
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, k.1, LV-1067 Riga, Latvia
| |
Collapse
|
37
|
Zhao T, Zhang Z, Li Y, Sun Z, Liu L, Deng X, Guo J, Zhu D, Cao S, Chai Y, Nikolaevna UV, Maratbek S, Wang Z, Zhang H. Brucella abortus modulates macrophage polarization and inflammatory response by targeting glutaminases through the NF-κB signaling pathway. Front Immunol 2023; 14:1180837. [PMID: 37325614 PMCID: PMC10266586 DOI: 10.3389/fimmu.2023.1180837] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/05/2023] [Indexed: 06/17/2023] Open
Abstract
Objectives The mechanism of Brucella infection regulating macrophage phenotype has not been completely elucidated until now. This study aimed to determine the mechanism of Brucella abortus in the modulation of macrophage phenotype using RAW264.7 cells as a model. Materials and methods RT-qPCR, ELISA and flow cytometry were used to detect the inflammatory factor production and phenotype conversion associated with M1/M2 polarization of macrophages by Brucella abortus infection. Western blot and immunofluorescence were used to analyze the role of nuclear factor kappa B (NF-κB) signaling pathway in regulation of Brucella abortus-induced macrophage polarization. Chromatin immunoprecipitation sequencing (Chip-seq), bioinformatics analysis and luciferase reporter assay were used to screen and validate NF-κB target genes associated with macrophage polarization and further verify its function. Results The results demonstrate that B. abortus induces a macrophage phenotypic switch and inflammatory response in a time-dependent manner. With the increase of infection time, B. abortus infection-induced M1-type increased first, peaked at 12 h, and then decreased, whereas the M2-type decreased first, trough at 12 h, and then increased. The trend of intracellular survival of B. abortus was consistent with that of M2 type. When NF-κB was inhibited, M1-type polarization was inhibited and M2-type was promoted, and the intracellular survival of B. abortus increased significantly. Chip-seq and luciferase reporter assay results showed that NF-κB binds to the glutaminase gene (Gls). Gls expression was down-regulated when NF-κB was inhibited. Furthermore, when Gls was inhibited, M1-type polarization was inhibited and M2-type was promoted, the intracellular survival of B. abortus increased significantly. Our data further suggest that NF-κB and its key target gene Gls play an important role in controlling macrophage phenotypic transformation. Conclusions Taken together, our study demonstrates that B. abortus infection can induce dynamic transformation of M1/M2 phenotype in macrophages. Highlighting NF-κB as a central pathway that regulates M1/M2 phenotypic transition. This is the first to elucidate the molecular mechanism of B. abortus regulation of macrophage phenotype switch and inflammatory response by regulating the key gene Gls, which is regulated by the transcription factor NF-κB.
Collapse
Affiliation(s)
- Tianyi Zhao
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Zedan Zhang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Yitao Li
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Zhihua Sun
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Liangbo Liu
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Xingmei Deng
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Jia Guo
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Dexin Zhu
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Shuzhu Cao
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Yingjin Chai
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Usevich Vera Nikolaevna
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- College of Veterinary, Ural State Agricultural University, Yekaterinburg, Russia
| | - Suleimenov Maratbek
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- College of Veterinary, National Agricultural University of Kazakhstan, Nur Sultan, Kazakhstan
| | - Zhen Wang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Hui Zhang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| |
Collapse
|
38
|
Finlay CM, Parkinson JE, Zhang L, Chan BHK, Ajendra J, Chenery A, Morrison A, Kaymak I, Houlder EL, Murtuza Baker S, Dickie BR, Boon L, Konkel JE, Hepworth MR, MacDonald AS, Randolph GJ, Rückerl D, Allen JE. T helper 2 cells control monocyte to tissue-resident macrophage differentiation during nematode infection of the pleural cavity. Immunity 2023; 56:1064-1081.e10. [PMID: 36948193 PMCID: PMC7616141 DOI: 10.1016/j.immuni.2023.02.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 10/07/2022] [Accepted: 02/21/2023] [Indexed: 03/24/2023]
Abstract
The recent revolution in tissue-resident macrophage biology has resulted largely from murine studies performed in C57BL/6 mice. Here, using both C57BL/6 and BALB/c mice, we analyze immune cells in the pleural cavity. Unlike C57BL/6 mice, naive tissue-resident large-cavity macrophages (LCMs) of BALB/c mice failed to fully implement the tissue-residency program. Following infection with a pleural-dwelling nematode, these pre-existing differences were accentuated with LCM expansion occurring in C57BL/6, but not in BALB/c mice. While infection drove monocyte recruitment in both strains, only in C57BL/6 mice were monocytes able to efficiently integrate into the resident pool. Monocyte-to-macrophage conversion required both T cells and interleukin-4 receptor alpha (IL-4Rα) signaling. The transition to tissue residency altered macrophage function, and GATA6+ tissue-resident macrophages were required for host resistance to nematode infection. Therefore, during tissue nematode infection, T helper 2 (Th2) cells control the differentiation pathway of resident macrophages, which determines infection outcome.
Collapse
Affiliation(s)
- Conor M Finlay
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK; Trinity Health Kidney Centre, Trinity Translational Medicine Institute, Trinity College, Dublin D08 W9RT, Ireland.
| | - James E Parkinson
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Lili Zhang
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Brian H K Chan
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Jesuthas Ajendra
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Alistair Chenery
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Anya Morrison
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Irem Kaymak
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Emma L Houlder
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Syed Murtuza Baker
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK; Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Ben R Dickie
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Salford M6 8HD, UK
| | | | - Joanne E Konkel
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Matthew R Hepworth
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Gwendalyn J Randolph
- Department of Pathology & Immunology, Washington University, St. Louis, MO 63110, USA
| | - Dominik Rückerl
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Judith E Allen
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
39
|
Lin J, Chen P, Tan Z, Sun Y, Tam WK, Ao D, Shen W, Leung VYL, Cheung KMC, To MKT. Application of silver nanoparticles for improving motor recovery after spinal cord injury via reduction of pro-inflammatory M1 macrophages. Heliyon 2023; 9:e15689. [PMID: 37234658 PMCID: PMC10205515 DOI: 10.1016/j.heliyon.2023.e15689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Silver nanoparticles (AgNPs) possess anti-inflammatory activities and have been widely deployed for promoting tissue repair. Here we explored the efficacy of AgNPs on functional recovery after spinal cord injury (SCI). Our data indicated that, in a SCI rat model, local AgNPs delivery could significantly recover locomotor function and exert neuroprotection through reducing of pro-inflammatory M1 survival. Furthermore, in comparison with Raw 264.7-derived M0 and M2, a higher level of AgNPs uptake and more pronounced cytotoxicity were detected in M1. RNA-seq analysis revealed the apoptotic genes in M1 were upregulated by AgNPs, whereas in M0 and M2, pro-apoptotic genes were downregulated and PI3k-Akt pathway signaling pathway was upregulated. Moreover, AgNPs treatment preferentially reduced cell viability of human monocyte-derived M1 comparing to M2, supporting its effect on M1 in human. Overall, our findings reveal AgNPs could suppress M1 activity and imply its therapeutic potential in promoting post-SCI motor recovery.
Collapse
Affiliation(s)
- Jie Lin
- Department of Orthopaedics & Traumatology, The University of Hong Kong Shenzhen Hospital, School of Clinical Medicine, The University of Hong Kong, Shenzhen, Guangdong, 518053, China
- Department of Orthopaedics & Traumatology, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Peikai Chen
- Department of Orthopaedics & Traumatology, The University of Hong Kong Shenzhen Hospital, School of Clinical Medicine, The University of Hong Kong, Shenzhen, Guangdong, 518053, China
| | - Zhijia Tan
- Department of Orthopaedics & Traumatology, The University of Hong Kong Shenzhen Hospital, School of Clinical Medicine, The University of Hong Kong, Shenzhen, Guangdong, 518053, China
| | - Yi Sun
- Department of Sports Medicine, Peking University-Shenzhen Hospital, Shenzhen, Guangdong, 518034, China
| | - Wai Kit Tam
- Department of Orthopaedics & Traumatology, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Di Ao
- Department of Orthopaedics & Traumatology, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Wei Shen
- Department of Orthopaedics & Traumatology, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Victor Yu-Leong Leung
- Department of Orthopaedics & Traumatology, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Kenneth Man Chee Cheung
- Department of Orthopaedics & Traumatology, The University of Hong Kong Shenzhen Hospital, School of Clinical Medicine, The University of Hong Kong, Shenzhen, Guangdong, 518053, China
- Department of Orthopaedics & Traumatology, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Michael Kai Tsun To
- Department of Orthopaedics & Traumatology, The University of Hong Kong Shenzhen Hospital, School of Clinical Medicine, The University of Hong Kong, Shenzhen, Guangdong, 518053, China
- Department of Orthopaedics & Traumatology, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
40
|
Yang Q, Dai H, Wang B, Xu J, Zhang Y, Chen Y, Ma Q, Xu F, Cheng H, Sun D, Wang C. Nanoplastics Shape Adaptive Anticancer Immunity in the Colon in Mice. NANO LETTERS 2023; 23:3516-3523. [PMID: 37043775 DOI: 10.1021/acs.nanolett.3c00644] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The impact of nanoplastics (NPs) on human health is still not well understood, and more research is needed to better understand the risks associated with these particles. In this study, we found that oral administration of polyethylene (PE) NPs in a mice model significantly disrupted the intestinal microenvironment, which shapes adaptive immune response and favors the established in situ colorectal tumor growth. Using single-cell RNA sequencing technology, we show that NPs triggered colon IL-1β-producing macrophages by inducing lysosome damage, leading to colonic Treg and Th17 differentiation associated with T cell exhaustion, which creates a colon environment that favors the tumor initiation and progress. A similar effect is also observed in polystyrene NPs. Our result provides insight into the potential link between NPs ingestion and colon tumorigenesis, and the urgency of addressing plastic pollution worldwide.
Collapse
Affiliation(s)
- Qianyu Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Huaxing Dai
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Beilei Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yue Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yitong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Qingle Ma
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Fang Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Dongdong Sun
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
41
|
Pinos I, Yu J, Pilli N, Kane MA, Amengual J. Functional characterization of interleukin 4 and retinoic acid signaling crosstalk during alternative macrophage activation. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159291. [PMID: 36754230 PMCID: PMC9974901 DOI: 10.1016/j.bbalip.2023.159291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 02/09/2023]
Abstract
Retinoic acid possesses potent immunomodulatory properties in various cell types, including macrophages. In this study, we first investigated the effects at the transcriptional and functional levels of exogenous retinoic acid in murine bone marrow-derived macrophages (BMDMs) in the presence or absence of interleukin 4 (IL4), a cytokine with potent anti-inflammatory properties. We examined the effect of IL4 on vitamin A homeostasis in macrophages by quantifying retinoid synthesis and secretion. Our RNAseq data show that exogenous retinoic acid synergizes with IL4 to regulate anti-inflammatory pathways such as oxidative phosphorylation and phagocytosis. Efferocytosis and lysosomal degradation assays validated gene expression changes at the functional level. IL4 treatment altered the expression of several genes involved in vitamin A transport and conversion to retinoic acid. Radiolabeling experiments and mass spectrometry assays revealed that IL4 stimulates retinoic acid production and secretion in a signal transducer and activator of transcription 6 (STAT6)-dependent manner. In summary, our studies highlight the key role of exogenous and endogenous retinoic acid in shaping the anti-inflammatory response of macrophages.
Collapse
Affiliation(s)
- Ivan Pinos
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL, United States
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Nageswara Pilli
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Jaume Amengual
- Division of Nutritional Sciences, University of Illinois Urbana Champaign, Urbana, IL, United States; Department of Food Science and Human Nutrition, University of Illinois Urbana Champaign, Urbana, IL, United States.
| |
Collapse
|
42
|
Wen X, Xi K, Tang Y, Bian J, Qin Y, Xiao W, Pan T, Cheng X, Ge Z, Cui W. Immunized Microspheres Engineered Hydrogel Membrane for Reprogramming Macrophage and Mucosal Repair. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207030. [PMID: 36604983 DOI: 10.1002/smll.202207030] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/07/2022] [Indexed: 06/17/2023]
Abstract
The "double-edged sword" effect of macrophages under the influence of different microenvironments determines the outcome and prognosis of tissue injury. Accurate and stable reprogramming macrophages (Mφ) are the key to rapid wound healing. In this study, an immunized microsphere-engineered GelMA hydrogel membrane is constructed for oral mucosa treatment. The nanoporous poly(lactide-co-glycolide) (PLGA) microsphere drug delivery system combined with the photo-cross-linkable hydrogel is used to release the soybean lecithin (SL)and IL-4 complexes (SL/IL-4) sustainedly. In this way, it is realized effective wound fit, improvement of drug encapsulation, and stable triphasic release of interleukin-4 (IL-4). In both in vivo and in vitro experiments, it is demonstrated that the hydrogel membrane can reprogram macrophages in the microenvironment into M2Mφ anti-inflammatory types, thereby inhibiting the local excessive inflammatory response. Meanwhile, high levels of platelet-derived growth factor (PDGF) secreted by M2Mφ macrophages enhanced neovascular maturation by 5.7-fold, which assisted in achieving rapid healing of oral mucosa. These findings suggest that the immuno-engineered hydrogel membrane system can re-modulating the biological effects of Mφ, and potentiating the maturation of neovascularization, ultimately achieving the rapid repair of mucosal tissue. This new strategy is expected to be a safe and promising immunomodulatory biomimetic material for clinical translation.
Collapse
Affiliation(s)
- Xiao Wen
- Department of Stomatology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Kun Xi
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Yu Tang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jie Bian
- Department of Stomatology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Yu Qin
- Department of Stomatology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wanshu Xiao
- Department of Stomatology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Tingzheng Pan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Xiaoming Cheng
- Department of Stomatology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Zili Ge
- Department of Stomatology, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
43
|
Li J, Zhao C, Xu Y, Song L, Chen Y, Xu Y, Ma Y, Wang S, Xu A, He F. Remodeling of the osteoimmune microenvironment after biomaterials implantation in murine tibia: Single-cell transcriptome analysis. Bioact Mater 2023; 22:404-422. [PMID: 36311047 PMCID: PMC9588995 DOI: 10.1016/j.bioactmat.2022.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/28/2022] [Accepted: 10/05/2022] [Indexed: 11/12/2022] Open
Abstract
Osseointegration seems to be a foreign body reaction equilibrium due to the complicated interactions between the immune and skeletal systems. The heterogeneity of the osteoimmune microenvironment in the osseointegration of implant materials remains elusive. Here, a single-cell study involving 40043 cells is conducted, and a total of 10 distinct cell clusters are identified from five different groups. A preliminary description of the osteoimmune microenvironment revealed the diverse cellular heterogeneity and dynamic changes modulated by implant properties. The increased immature neutrophils, Ly6C + CCR2hi monocytes, and S100a8hi macrophages induce an aggressive inflammatory response and eventually lead to the formation of fibrous capsule around the stainless steel implant. The enrichment of mature neutrophils, FcgR1hi and differentiated immunomodulatory macrophages around the titanium implant indicates favorable osseointegration under moderate immune response. Neutrophil-depletion mice are conducted to explore the role of neutrophils in osseointegration. Neutrophils may improve bone formation by enhancing the recruitment of BMSCs via the CXCL12/CXCR3 signal axis. These findings contribute to a better knowledge of osteoimmunology and are valuable for the design and modification of 'osteoimmune-smart' biomaterials in the bone regeneration field.
Collapse
Key Words
- BMP2, Bone Morphogenetic Proteins 2
- CXCL12, Chemokine (C-X-C mode) Ligand 12
- CXCR, CXC Chemokine Receptor
- FcgR, Fc Gamma Receptor
- IFN-γ, Interferon-gamma
- IL-1β, Interleukin-1 beta
- Implant
- MHC, Major Histocompatibility Complex
- MIP, Macrophage inflammatory cytokines
- MPO, Myeloperoxidase
- NE, Neutrophil Elastase
- NF-κB, Nuclear Factor Kappa-light-chain-enhancer of Activated B cells
- NOD, Nucleotide Binding Oligomerization Domain
- Neutrophil
- OPG, Osteoprotegerin
- Osseointegration
- Osteoimmunology
- RANKL, Nuclear Factor B receptor Activator Ligand
- RUNX2, Runt-related Transcription Factor 2
- S100a8, S100 Calcium Binding Protein A8
- SDF-1α, Stromal Cell-derived Factor-1 alpha
- STAT, Signal Transduction and Transcription Activator
- Single-cell transcriptomics
- TLR, Toll Like Receptor
- TNFα, Tumor Necrosis Factor-alpha
- TRAP, Tartrate Resistant Acid Phosphatase
Collapse
Affiliation(s)
- Jia Li
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Congrui Zhao
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yangbo Xu
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Lu Song
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yanqi Chen
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yuzi Xu
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yang Ma
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Siyuan Wang
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Antian Xu
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Fuming He
- Department of Prosthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| |
Collapse
|
44
|
Wang G, Lin Z, Li Y, Chen L, Reddy SK, Hu Z, Garza LA. Colonizing microbiota is associated with clinical outcomes in diabetic wound healing. Adv Drug Deliv Rev 2023; 194:114727. [PMID: 36758858 PMCID: PMC10163681 DOI: 10.1016/j.addr.2023.114727] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
With the development of society and the improvement of life quality, more than 500 million people are affected by diabetes. More than 10 % of people with diabetes will suffer from diabetic wounds, and 80 % of diabetic wounds will reoccur, so the development of new diabetic wound treatments is of great importance. The development of skin microbe research technology has gradually drawn people's attention to the complex relationship between microbes and diabetic wounds. Many studies have shown that skin microbes are associated with the outcome of diabetic wounds and can even be used as one of the indicators of wound prognosis. Skin microbes have also been found to have the potential to treat diabetic wounds. The wound colonization of different bacteria can exert opposing therapeutic effects. It is necessary to fully understand the skin microbes in diabetic wounds, which can provide valuable guidance for clinical diabetic wound treatment.
Collapse
Affiliation(s)
- Gaofeng Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China; Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| | - Zhen Lin
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Yue Li
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Lu Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Sashank K Reddy
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - L A Garza
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| |
Collapse
|
45
|
Papotto PH, Yilmaz B, Pimenta G, Mensurado S, Cunha C, Fiala GJ, Gomes da Costa D, Gonçalves-Sousa N, Chan BHK, Blankenhaus B, Domingues RG, Carvalho T, Hepworth MR, Macpherson AJ, Allen JE, Silva-Santos B. Maternal γδ T cells shape offspring pulmonary type 2 immunity in a microbiota-dependent manner. Cell Rep 2023; 42:112074. [PMID: 36787741 PMCID: PMC7615642 DOI: 10.1016/j.celrep.2023.112074] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/21/2022] [Accepted: 01/23/2023] [Indexed: 02/15/2023] Open
Abstract
Immune development is profoundly influenced by vertically transferred cues. However, little is known about how maternal innate-like lymphocytes regulate offspring immunity. Here, we show that mice born from γδ T cell-deficient (TCRδ-/-) dams display an increase in first-breath-induced inflammation, with a pulmonary milieu selectively enriched in type 2 cytokines and type 2-polarized immune cells, when compared with the progeny of γδ T cell-sufficient dams. Upon helminth infection, mice born from TCRδ-/- dams sustain an increased type 2 inflammatory response. This is independent of the genotype of the pups. Instead, the offspring of TCRδ-/- dams harbors a distinct intestinal microbiota, acquired during birth and fostering, and decreased levels of intestinal short-chain fatty acids (SCFAs), such as pentanoate and hexanoate. Importantly, exogenous SCFA supplementation inhibits type 2 innate lymphoid cell function and suppresses first-breath- and infection-induced inflammation. Taken together, our findings unravel a maternal γδ T cell-microbiota-SCFA axis regulating neonatal lung immunity.
Collapse
Affiliation(s)
- Pedro H Papotto
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
| | - Bahtiyar Yilmaz
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland; Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gonçalo Pimenta
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sofia Mensurado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Carolina Cunha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Gina J Fiala
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Daniel Gomes da Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Natacha Gonçalves-Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Brian H K Chan
- Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Birte Blankenhaus
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rita G Domingues
- Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Tânia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Matthew R Hepworth
- Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Andrew J Macpherson
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland; Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Judith E Allen
- Lydia Becker Institute for Immunology & Infection, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK; Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
46
|
Xia S, Vila Ellis L, Winkley K, Menden H, Mabry SM, Venkatraman A, Louiselle D, Gibson M, Grundberg E, Chen J, Sampath V. Neonatal hyperoxia induces activated pulmonary cellular states and sex-dependent transcriptomic changes in a model of experimental bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L123-L140. [PMID: 36537711 PMCID: PMC9902224 DOI: 10.1152/ajplung.00252.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
Hyperoxia disrupts lung development in mice and causes bronchopulmonary dysplasia (BPD) in neonates. To investigate sex-dependent molecular and cellular programming involved in hyperoxia, we surveyed the mouse lung using single cell RNA sequencing (scRNA-seq), and validated our findings in human neonatal lung cells in vitro. Hyperoxia-induced inflammation in alveolar type (AT) 2 cells gave rise to damage-associated transient progenitors (DATPs). It also induced a new subpopulation of AT1 cells with reduced expression of growth factors normally secreted by AT1 cells, but increased mitochondrial gene expression. Female alveolar epithelial cells had less EMT and pulmonary fibrosis signaling in hyperoxia. In the endothelium, expansion of Car4+ EC (Cap2) was seen in hyperoxia along with an emergent subpopulation of Cap2 with repressed VEGF signaling. This regenerative response was increased in females exposed to hyperoxia. Mesenchymal cells had inflammatory signatures in hyperoxia, with a new distal interstitial fibroblast subcluster characterized by repressed lipid biosynthesis and a transcriptomic signature resembling myofibroblasts. Hyperoxia-induced gene expression signatures in human neonatal fibroblasts and alveolar epithelial cells in vitro resembled mouse scRNA-seq data. These findings suggest that neonatal exposure to hyperoxia programs distinct sex-specific stem cell progenitor and cellular reparative responses that underpin lung remodeling in BPD.
Collapse
Affiliation(s)
- Sheng Xia
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Lisandra Vila Ellis
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Konner Winkley
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Heather Menden
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Sherry M Mabry
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Aparna Venkatraman
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Daniel Louiselle
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Margaret Gibson
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Elin Grundberg
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
- Children's Mercy Research Institute, Kansas City, Missouri
| | - Jichao Chen
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Venkatesh Sampath
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
- Children's Mercy Research Institute, Kansas City, Missouri
| |
Collapse
|
47
|
Yan Y, Li X, Yang Q, Zhang H, Hettinga K, Li H, Chen W. Dietary d-Lactate Intake Facilitates Inflammatory Resolution by Modulating M1 Macrophage Polarization. Mol Nutr Food Res 2022; 66:e2200196. [PMID: 36239154 DOI: 10.1002/mnfr.202200196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/01/2022] [Indexed: 01/18/2023]
Abstract
SCOPE Given the d-lactate dehydrogenase (D-LDH) deficiency, L- but not d-lactate is assumed to be the physiological isomer in mammals. Paradoxically, many fermented foods (e.g., yogurt, sauerkraut, cheeses) often contain substantial amounts of d-lactate. In the present study, dietary d-lactate may be a previously unrecognized nutrient aiding in inflammatory resolution is hypothesized. METHODS AND RESULTS The anti-inflammatory properties of d-lactate are evaluated in experimental colitis and endotoxemia. Oral administration of d-lactate favorably affects acute inflammation in two different mouse models. Analysis of lactate-the lactate receptor (the hydroxycarboxylic acid receptor 1 HCA1, formerly GPR81) signal axis in inflammation is performed in primary peritoneal macrophages and wild-type (WT) or GPR81 knockout (KO) mice. GPR81 KO mice are susceptible to endotoxic shock than WT mice, while d-lactate exerts its anti-inflammatory activities in a GPR81-dependent manner. Mechanistically, the activation of lactate-GPR81 axis may suppress LPS-TLR4 signaling to modulate M1 macrophage polarization. Although D-LDH deficiency in mammals impairs d-lactate clearance, it might prolong its plasma terminal half-life, and thus provide a pharmacokinetic advantage of d-lactate over l-lactate. CONCLUSION This study highlights housekeeping function of the lactate-GPR81 axis in inflammation control, and suggests that dietary intake of d-lactate may underlie Metchnikoff's probiotic yogurt theory of life prolongation.
Collapse
Affiliation(s)
- Yongheng Yan
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Xiu Li
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Qin Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Hao Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Kasper Hettinga
- Dairy Science and Technology, Food Quality and Design, Wageningen University and Research, 6700 AA, Wageningen, P.O. Box 17, The Netherlands
| | - Haitao Li
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Wei Chen
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
48
|
Liu XH, Zhou JT, Yan CX, Cheng C, Fan JN, Xu J, Zheng Q, Bai Q, Li Z, Li S, Li X. Single-cell RNA sequencing reveals a novel inhibitory effect of ApoA4 on NAFL mediated by liver-specific subsets of myeloid cells. Front Immunol 2022; 13:1038401. [PMID: 36426356 PMCID: PMC9678944 DOI: 10.3389/fimmu.2022.1038401] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/20/2022] [Indexed: 10/24/2023] Open
Abstract
The liver immune microenvironment is a key element in the development of hepatic inflammation in NAFLD. ApoA4 deficiency increases the hepatic lipid burden, insulin resistance, and metabolic inflammation. However, the effect of ApoA4 on liver immune cells and the precise immune cell subsets that exacerbate fatty liver remain elusive. The aim of this study was to profile the hepatic immune cells affected by ApoA4 in NAFL. We performed scRNA-seq on liver immune cells from WT and ApoA4-deficient mice administered a high-fat diet. Immunostaining and qRT-PCR analysis were used to validate the results of scRNA-seq. We identified 10 discrete immune cell populations comprising macrophages, DCs, granulocytes, B, T and NK&NKT cells and characterized their subsets, gene expression profiles, and functional modules. ApoA4 deficiency led to significant increases in the abundance of specific subsets, including inflammatory macrophages (2-Mφ-Cxcl9 and 4-Mφ-Cxcl2) and activated granulocytes (0-Gran-Wfdc17). Moreover, ApoA4 deficiency resulted in higher Lgals3, Ctss, Fcgr2b, Spp1, Cxcl2, and Elane levels and lower Nr4a1 levels in hepatic immune cells. These genes were consistent with human NAFLD-associated marker genes linked to disease severity. The expression of NE and IL-1β in granulocytes and macrophages as key ApoA4 targets were validate in the presence or absence of ApoA4 by immunostaining. The scRNA-seq data analyses revealed reprogramming of liver immune cells resulted from ApoA4 deficiency. We uncovered that the emergence of ApoA4-associated immune subsets (namely Cxcl9+ macrophage, Cxcl2+ macrophage and Wfdc17+ granulocyte), pathways, and NAFLD-related marker genes may promote the development of NAFL. These findings may provide novel therapeutic targets for NAFL and the foundations for further studying the effects of ApoA4 on immune cells in various diseases.
Collapse
Affiliation(s)
- Xiao-Huan Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Jin-Ting Zhou
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Chun-xia Yan
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Department of Pathology, Bio-Evidence Sciences Academy, The Western China Science and Technology Innovation Port, Xi’an Jiaotong University, Xi’an, China
| | - Cheng Cheng
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Jing-Na Fan
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Jing Xu
- Division of Endocrinology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qiangsun Zheng
- Division of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qiang Bai
- Laboratory of Immunophysiology, GIGA Institute, Liège University, Liège, Belgium
| | - Zongfang Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Shengbin Li
- Key laboratory of Ministry of Public Health for Forensic Sciences, Western China Science & Technology Innovation Harbour, Xi’an, China
| | - Xiaoming Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
49
|
Dolitzky A, Hazut I, Avlas S, Grisaru-Tal S, Itan M, Zaffran I, Levi-Schaffer F, Gerlic M, Munitz A. Differential regulation of Type 1 and Type 2 mouse eosinophil activation by apoptotic cells. Front Immunol 2022; 13:1041660. [PMID: 36389786 PMCID: PMC9662748 DOI: 10.3389/fimmu.2022.1041660] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/07/2022] [Indexed: 08/18/2023] Open
Abstract
Eosinophils are multifunctional, evolutionary conserved leukocytes that are involved in a plethora of responses ranging from regulation of tissue homeostasis, host defense and cancer. Although eosinophils have been studied mostly in the context of Type 2 inflammatory responses, it is now evident that they participate in Type 1 inflammatory responses and can respond to Type 1 cytokines such as IFN-γ. Notably, both Type 1- and Type 2 inflammatory environments are characterized by tissue damage and cell death. Collectively, this raises the possibility that eosinophils can interact with apoptotic cells, which can alter eosinophil activation in the inflammatory milieu. Herein, we demonstrate that eosinophils can bind and engulf apoptotic cells. We further show that exposure of eosinophils to apoptotic cells induces marked transcriptional changes in eosinophils, which polarize eosinophils towards an anti-inflammatory phenotype that is associated with wound healing and cell migration. Using an unbiased RNA sequencing approach, we demonstrate that apoptotic cells suppress the inflammatory responses of eosinophils that were activated with IFN-γ + E. coli (e.g., Type 1 eosinophils) and augment IL-4-induced eosinophil activation (e.g., Type 2 eosinophils). These data contribute to the growing understanding regarding the heterogeneity of eosinophil activation patterns and highlight apoptotic cells as potential regulators of eosinophil polarization.
Collapse
Affiliation(s)
- Avishay Dolitzky
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inbal Hazut
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shmulik Avlas
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Grisaru-Tal
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Itan
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Zaffran
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Motti Gerlic
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Munitz
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
50
|
Bahr JC, Li XY, Feinberg TY, Jiang L, Weiss SJ. Divergent regulation of basement membrane trafficking by human macrophages and cancer cells. Nat Commun 2022; 13:6409. [PMID: 36302921 PMCID: PMC9613642 DOI: 10.1038/s41467-022-34087-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/13/2022] [Indexed: 12/25/2022] Open
Abstract
Macrophages and cancer cells populations are posited to navigate basement membrane barriers by either mobilizing proteolytic enzymes or deploying mechanical forces. Nevertheless, the relative roles, or identity, of the proteinase -dependent or -independent mechanisms used by macrophages versus cancer cells to transmigrate basement membrane barriers harboring physiologically-relevant covalent crosslinks remains ill-defined. Herein, both macrophages and cancer cells are shown to mobilize membrane-anchored matrix metalloproteinases to proteolytically remodel native basement membranes isolated from murine tissues while infiltrating the underlying interstitial matrix ex vivo. In the absence of proteolytic activity, however, only macrophages deploy actomyosin-generated forces to transmigrate basement membrane pores, thereby providing the cells with proteinase-independent access to the interstitial matrix while simultaneously exerting global effects on the macrophage transcriptome. By contrast, cancer cell invasive activity is reliant on metalloproteinase activity and neither mechanical force nor changes in nuclear rigidity rescue basement membrane transmigration. These studies identify membrane-anchored matrix metalloproteinases as key proteolytic effectors of basement membrane remodeling by macrophages and cancer cells while also defining the divergent invasive strategies used by normal and neoplastic cells to traverse native tissue barriers.
Collapse
Affiliation(s)
- Julian C Bahr
- Cancer Biology Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xiao-Yan Li
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tamar Y Feinberg
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Long Jiang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stephen J Weiss
- Cancer Biology Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA.
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|