1
|
Airola C, Varca S, Del Gaudio A, Pizzolante F. The Covert Side of Ascites in Cirrhosis: Cellular and Molecular Aspects. Biomedicines 2025; 13:680. [PMID: 40149656 PMCID: PMC11940454 DOI: 10.3390/biomedicines13030680] [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: 12/21/2024] [Revised: 02/25/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Ascites, a common complication of portal hypertension in cirrhosis, is characterized by the accumulation of fluid within the peritoneal cavity. While traditional theories focus on hemodynamic alterations and renin-angiotensin-aldosterone system (RAAS) activation, recent research highlights the intricate interplay of molecular and cellular mechanisms. Inflammation, mediated by cytokines (interleukin-1, interleukin-4, interleukin-6, tumor necrosis factor-α), chemokines (chemokine ligand 21, C-X-C motif chemokine ligand 12), and reactive oxygen species (ROS), plays a pivotal role. Besides pro-inflammatory cytokines, hepatic stellate cells (HSCs), sinusoidal endothelial cells (SECs), and smooth muscle cells (SMCs) contribute to the process through their activation and altered functions. Once activated, these cell types can worsen ascites accumulationthrough extracellular matrix (ECM) deposition and paracrine signals. Besides this, macrophages, both resident and infiltrating, through their plasticity, participate in this complex crosstalk by promoting inflammation and dysregulating lymphatic system reabsorption. Indeed, the lymphatic system and lymphangiogenesis, essential for fluid reabsorption, is dysregulated in cirrhosis, exacerbating ascites. The gut microbiota and intestinal barrier alterations which occur in cirrhosis and portal hypertension also play a role by inducing inflammation, creating a vicious circle which worsens portal hypertension and fluid accumulation. This review aims to gather these aspects of ascites pathophysiology which are usually less considered and to date have not been addressed using specific therapy. Nonetheless, it emphasizes the need for further research to understand the complex interactions among these mechanisms, ultimately leading to targeted interventions in specific molecular pathways, aiming towards the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Carlo Airola
- CEMAD Centro Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.V.); (A.D.G.)
- Facoltà di Medicina e Chirurgia, Università Cattolica Sacro Cuore, Largo Agostino Gemelli, 8, 00168 Rome, Italy
| | - Simone Varca
- CEMAD Centro Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.V.); (A.D.G.)
- Facoltà di Medicina e Chirurgia, Università Cattolica Sacro Cuore, Largo Agostino Gemelli, 8, 00168 Rome, Italy
| | - Angelo Del Gaudio
- CEMAD Centro Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.V.); (A.D.G.)
- Facoltà di Medicina e Chirurgia, Università Cattolica Sacro Cuore, Largo Agostino Gemelli, 8, 00168 Rome, Italy
| | - Fabrizio Pizzolante
- CEMAD Centro Malattie dell’Apparato Digerente, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.V.); (A.D.G.)
| |
Collapse
|
2
|
Kwon J, Kawase H, Mattonet K, Guenther S, Hahnefeld L, Shamsara J, Heering J, Kurz M, Kirchhofer S, Krasel C, Ulrich M, Persechino M, Murthy S, Orlandi C, Sadik CD, Geisslinger G, Bünemann M, Kolb P, Offermanns S, Wettschureck N. Orphan G protein-coupled receptor GPRC5B controls macrophage function by facilitating prostaglandin E receptor 2 signaling. Nat Commun 2025; 16:1448. [PMID: 39920161 PMCID: PMC11805951 DOI: 10.1038/s41467-025-56713-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Macrophages express numerous G protein-coupled receptors (GPCRs) that regulate adhesion, migration, and activation, but the function of orphan receptor GPRC5B in macrophages is unknown. Both resident peritoneal and bone marrow-derived macrophages from myeloid-specific GPRC5B-deficient mice show increased migration and phagocytosis, resulting in improved bacterial clearance in a peritonitis model. In other models such as myocardial infarction, increased myeloid cell recruitment has adverse effects. Mechanistically, we found that GPRC5B physically interacts with GPCRs of the prostanoid receptor family, resulting in enhanced signaling through the prostaglandin E receptor 2 (EP2). In GPRC5B-deficient macrophages, EP2-mediated anti-inflammatory effects are diminished, resulting in hyperactivity. Using in silico modelling and docking, we identify residues potentially mediating GPRC5B/EP2 dimerization and show that their mutation results in loss of GPRC5B-mediated facilitation of EP2 signaling. Finally, we demonstrate that decoy peptides mimicking the interacting sequence are able to reduce GPRC5B-mediated facilitation of EP2-induced cAMP signaling in macrophages.
Collapse
Affiliation(s)
- Jeonghyeon Kwon
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Haruya Kawase
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Kenny Mattonet
- Imaging Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Guenther
- Deep sequencing platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Lisa Hahnefeld
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, Institute of Clinical Pharmacology, Frankfurt am Main, Germany
| | - Jamal Shamsara
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Michael Kurz
- Department of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | - Sina Kirchhofer
- Department of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | - Cornelius Krasel
- Department of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | - Michaela Ulrich
- Department of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | | | - Sripriya Murthy
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Christian D Sadik
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany
| | - Gerd Geisslinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, Institute of Clinical Pharmacology, Frankfurt am Main, Germany
| | - Moritz Bünemann
- Department of Pharmacology and Clinical Pharmacy, University of Marburg, Marburg, Germany
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Centre for Molecular Medicine, Medical Faculty, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Centre for Molecular Medicine, Medical Faculty, Goethe-University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
3
|
Li R, Li J, Liu S, Guo X, Lu J, Wang T, Chen J, Zheng Y, Yuan Y, Du J, Zhu B, Wei X, Guo P, Liu L, Xu X, Dai X, Huang R, Liu X, Hu X, Wang S, Ji S. A scATAC-seq atlas of stasis zone in rat skin burn injury wound process. Front Cell Dev Biol 2025; 12:1519926. [PMID: 39845081 PMCID: PMC11752905 DOI: 10.3389/fcell.2024.1519926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
Burn injuries often leave behind a "stasis zone", a region of tissue critically important for determining both the severity of the injury and the potential for recovery. To understand the intricate cellular and epigenetic changes occurring within this critical zone, we utilized single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) to profile over 31,500 cells from both healthy rat skin and the stasis zone at nine different time points after a burn injury. This comprehensive approach revealed 26 distinct cell types and the dynamic shifts in the proportions of these cell types over time. We observed distinct gene activation patterns in different cell types at various stages post-burn, highlighting key players in immune activation, tissue regeneration, and blood vessel repair. Importantly, our analysis uncovered the regulatory networks governing these genes, offering valuable insights into the intricate mechanisms orchestrating burn wound healing. This comprehensive cellular and molecular atlas of the stasis zone provides a powerful resource for developing targeted therapies aimed at improving burn injury recovery and minimizing long-term consequences.
Collapse
Affiliation(s)
- Ruikang Li
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, The College of Life Sciences, Northwest University, Xi’an, China
| | - Jiashan Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shuai Liu
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xinya Guo
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jianyu Lu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Tao Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Hangzhou, China
| | | | - Yue Zheng
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, The College of Life Sciences, Northwest University, Xi’an, China
| | | | - Jiaxin Du
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Hangzhou, China
| | - Bolin Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Hangzhou, China
| | | | | | - Longqi Liu
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- BGI Research, Hangzhou, China
| | - Xun Xu
- BGI Research, Shenzhen, China
| | - Xi Dai
- BGI Research, Hangzhou, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xin Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Research, Shenzhen, China
| | - Xiaoyan Hu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shiwei Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, The College of Life Sciences, Northwest University, Xi’an, China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
4
|
Forni MF, Pizzurro GA, Krause W, Alexander AF, Bridges K, Xu Y, Justynski O, Gabry A, Camara NOS, Miller-Jensen K, Horsley V. Multiomics reveals age-dependent metabolic reprogramming of macrophages by wound bed niche secreted signals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621159. [PMID: 39553941 PMCID: PMC11565841 DOI: 10.1101/2024.10.30.621159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The cellular metabolism of macrophages depends on tissue niches and can control macrophage inflammatory or resolving phenotypes. Yet, the identity of signals within tissue niches that control macrophage metabolism is not well understood. Here, using single-cell RNA sequencing of macrophages in early mouse wounds, we find that, rather than gene expression of canonical inflammatory or resolving polarization markers, metabolic gene expression defines distinct populations of early wound macrophages. Single-cell secretomics and transcriptomics identify inflammatory and resolving cytokines expressed by early wound macrophages, and we show that these signals drive metabolic inputs and mitochondrial metabolism in an age-dependent manner. We show that aging alters the metabolome of early wound macrophages and rewires their metabolism from mitochondria to glycolysis. We further show that macrophage-derived Chi3l3 and IGF-1 can induce metabolic inputs and mitochondrial mass/metabolism in aged and bone marrow-derived macrophages. Together, these findings reveal that macrophage-derived signals drive the mitochondrial metabolism of macrophages within early wounds in an age-dependent manner and have implications for inflammatory diseases, chronic injuries, and age-related inflammatory diseases. In Brief This study reveals that macrophage subsets in early inflammatory stages of skin wound healing are defined by their metabolic profiles rather than polarization phenotype. Using single-cell secretomics, we establish key macrophage cytokines that comprise the in vivo wound niche and drive mitochondrial-based metabolism. Aging significantly alters macrophage heterogeneity and increases glycolytic metabolism, which can be restored to OxPHOS-based metabolism with young niche cytokines. These findings highlight the importance of the tissue niche in driving macrophage phenotypes, with implications for aging-related impairments in wound healing. Highlights Single cell transcriptional analysis reveals that reveals that metabolic gene expression identifies distinct macrophage populations in early skin wounds.Single-cell secretomic data show that young macrophages contribute to the wound bed niche by secreting molecules such as IGF-1 and Chi3l3.Old wound macrophages display altered metabolomics, elevated glycolytic metabolism and glucose uptake, and reduced lipid uptake and mitochondrial mass/metabolism.Chi3l3 but not IGF-1 secretion is altered in macrophages in an age dependent manner.Chi3l3 can restore mitochondrial mass/metabolism in aged macrophages.
Collapse
|
5
|
Lv H, Liu B, Dai Y, Li F, Bellone S, Zhou Y, Mamillapalli R, Zhao D, Venkatachalapathy M, Hu Y, Carmichael GG, Li D, Taylor HS, Huang Y. TET3-overexpressing macrophages promote endometriosis. J Clin Invest 2024; 134:e181839. [PMID: 39141428 PMCID: PMC11527447 DOI: 10.1172/jci181839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024] Open
Abstract
Endometriosis is a debilitating, chronic inflammatory disease affecting approximately 10% of reproductive-age women worldwide with no cure. While macrophages have been intrinsically linked to the pathophysiology of endometriosis, targeting them therapeutically has been extremely challenging due to their high heterogeneity and because these disease-associated macrophages (DAMs) can be either pathogenic or protective. Here, we report identification of pathogenic macrophages characterized by TET3 overexpression in human endometriosis lesions. We show that factors from the disease microenvironment upregulated TET3 expression, transforming macrophages into pathogenic DAMs. TET3 overexpression stimulated proinflammatory cytokine production via a feedback mechanism involving inhibition of let-7 miRNA expression. Remarkably, these cells relied on TET3 overexpression for survival and hence were vulnerable to TET3 knockdown. We demonstrated that Bobcat339, a synthetic cytosine derivative, triggered TET3 degradation in both human and mouse macrophages. This degradation was dependent on a von Hippel-Lindau (VHL) E3 ubiquitin ligase whose expression was also upregulated in TET3-overexpressing macrophages. Furthermore, depleting TET3-overexpressing macrophages either through myeloid-specific Tet3 ablation or using Bobcat339 strongly inhibited endometriosis progression in mice. Our results defined TET3-overexpressing macrophages as key pathogenic contributors to and attractive therapeutic targets for endometriosis. Our findings may also be applicable to other chronic inflammatory diseases where DAMs have important roles.
Collapse
Affiliation(s)
- Haining Lv
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Beibei Liu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
- Center of Reproductive Medicine, National Health Commission Key Laboratory of Advanced Reproductive Medicine and Fertility, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yangyang Dai
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Li
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Anesthesiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Stefania Bellone
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yuping Zhou
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dejian Zhao
- Yale Center for Genome Analysis, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Yali Hu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Gordon G. Carmichael
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Da Li
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
- Center of Reproductive Medicine, National Health Commission Key Laboratory of Advanced Reproductive Medicine and Fertility, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hugh S. Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yingqun Huang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
6
|
Schultze-Rhonhof L, Marzi J, Carvajal Berrio DA, Holl M, Braun T, Schäfer-Ruoff F, Andress J, Bachmann C, Templin M, Brucker SY, Schenke-Layland K, Weiss M. Human tissue-resident peritoneal macrophages reveal resistance towards oxidative cell stress induced by non-invasive physical plasma. Front Immunol 2024; 15:1357340. [PMID: 38504975 PMCID: PMC10949891 DOI: 10.3389/fimmu.2024.1357340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
In the context of multimodal treatments for abdominal cancer, including procedures such as cytoreductive surgery and intraperitoneal chemotherapy, recurrence rates remain high, and long-term survival benefits are uncertain due to post-operative complications. Notably, treatment-limiting side effects often arise from an uncontrolled activation of the immune system, particularly peritoneally localized macrophages, leading to massive cytokine secretion and phenotype changes. Exploring alternatives, an increasing number of studies investigated the potential of plasma-activated liquids (PAL) for adjuvant peritoneal cancer treatment, aiming to mitigate side effects, preserve healthy tissue, and reduce cytotoxicity towards non-cancer cells. To assess the non-toxicity of PAL, we isolated primary human macrophages from the peritoneum and subjected them to PAL exposure. Employing an extensive methodological spectrum, including flow cytometry, Raman microspectroscopy, and DigiWest protein analysis, we observed a pronounced resistance of macrophages towards PAL. This resistance was characterized by an upregulation of proliferation and anti-oxidative pathways, countering PAL-derived oxidative stress-induced cell death. The observed cellular effects of PAL treatment on human tissue-resident peritoneal macrophages unveil a potential avenue for PAL-derived immunomodulatory effects within the human peritoneal cavity. Our findings contribute to understanding the intricate interplay between PAL and macrophages, shedding light on the promising prospects for PAL in the adjuvant treatment of peritoneal cancer.
Collapse
Affiliation(s)
| | - Julia Marzi
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, University of Tübingen, Tübingen, Germany
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - Daniel Alejandro Carvajal Berrio
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, University of Tübingen, Tübingen, Germany
| | - Myriam Holl
- Department of Women’s Health Tübingen, University of Tübingen, Tübingen, Germany
| | - Theresa Braun
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
- University Development, Research and Transfer, University of Konstanz, Konstanz, Germany
| | - Felix Schäfer-Ruoff
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - Jürgen Andress
- Department of Women’s Health Tübingen, University of Tübingen, Tübingen, Germany
| | - Cornelia Bachmann
- Department of Women’s Health Tübingen, University of Tübingen, Tübingen, Germany
| | - Markus Templin
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - Sara Y. Brucker
- Department of Women’s Health Tübingen, University of Tübingen, Tübingen, Germany
| | - Katja Schenke-Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, University of Tübingen, Tübingen, Germany
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - Martin Weiss
- Department of Women’s Health Tübingen, University of Tübingen, Tübingen, Germany
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| |
Collapse
|
7
|
Cyndari KI, Scorza BM, Zacharias ZR, Strand L, Mahachi K, Oviedo JM, Gibbs L, Pessoa-Pereira D, Ausdal G, Hendricks D, Yahashiri R, Elkins JM, Gulbrandsen T, Peterson AR, Willey MC, Fairfax KC, Petersen CA. Resident Synovial Macrophages in Synovial Fluid: Implications for Immunoregulation in Infectious and Inflammatory Arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.29.560183. [PMID: 37873090 PMCID: PMC10592878 DOI: 10.1101/2023.09.29.560183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Objectives Resident synovial macrophages (RSM) provide immune sequestration of the joint space and are likely involved in initiation and perpetuation of the joint-specific immune response. We sought to identify RSM in synovial fluid (SF) and demonstrate migratory ability, in additional to functional changes that may perpetuate a chronic inflammatory response within joint spaces. Methods We recruited human patients presenting with undifferentiated arthritis in multiple clinical settings. We used flow cytometry to identify mononuclear cells in peripheral blood and SF. We used a novel transwell migration assay with human ex-vivo synovium obtained intra-operatively to validate flow cytometry findings. We used single cell RNA-sequencing (scRNA-seq) to further identify macrophage/monocyte subsets. ELISA was used to evaluate the bone-resorption potential of SF. Results We were able to identify a rare population of CD14dim, OPG+, ZO-1+ cells consistent with RSM in SF via flow cytometry. These cells were relatively enriched in the SF during infectious processes, but absolutely decreased compared to healthy controls. Similar putative RSM were identified using ex vivo migration assays when MCP-1 and LPS were used as migratory stimulus. scRNA-seq revealed a population consistent with RSM transcriptionally related to CD56+ cytotoxic dendritic cells and IDO+ M2 macrophages. Conclusion We identified a rare cell population consistent with RSM, indicating these cells are likely migratory and able to initiate or coordinate both acute (septic) or chronic (autoimmune or inflammatory) arthritis. RSM analysis via scRNA-seq indicated these cells are M2 skewed, capable of antigen presentation, and have consistent functions in both septic and inflammatory arthritis.
Collapse
Affiliation(s)
- Karen I Cyndari
- Department of Emergency Medicine, University of Iowa, Iowa City, IA
- Center for Emerging Infectious Diseases
| | - Breanna M Scorza
- Center for Emerging Infectious Diseases
- Department of Epidemiology, University of Iowa, Iowa City, IA
| | - Zeb R Zacharias
- Human Immunology Core, University of Iowa, Iowa City, IA
- Holden Comprehensive Cancer Center, Iowa City, IA
| | | | - Kurayi Mahachi
- Research and Analytics, Enterprise Analytics, Sentara Health
| | | | - Lisa Gibbs
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Danielle Pessoa-Pereira
- Center for Emerging Infectious Diseases
- Department of Epidemiology, University of Iowa, Iowa City, IA
| | - Graham Ausdal
- Center for Emerging Infectious Diseases
- Department of Epidemiology, University of Iowa, Iowa City, IA
| | - Dylan Hendricks
- Center for Emerging Infectious Diseases
- Department of Epidemiology, University of Iowa, Iowa City, IA
| | | | - Jacob M Elkins
- Department of Orthopedics, University of Iowa, Iowa City, IA
| | | | | | | | - Keke C Fairfax
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Christine A Petersen
- Center for Emerging Infectious Diseases
- Department of Epidemiology, University of Iowa, Iowa City, IA
| |
Collapse
|
8
|
Li A, Gu L, He C, Li Y, Peng M, Liao J, Xiao R, Xu L, Guo S. GATA6 promotes fibrotic repair of tracheal injury through NLRP3 inflammasome-mediated epithelial pyroptosis. Int Immunopharmacol 2023; 123:110657. [PMID: 37531826 DOI: 10.1016/j.intimp.2023.110657] [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: 06/06/2023] [Revised: 07/08/2023] [Accepted: 07/13/2023] [Indexed: 08/04/2023]
Abstract
Tracheal injury is a challenging emergency condition that is characterized by the abnormal repair of the trachea. GATA6, a well-established transcription factor, plays a crucial role in tissue injury and epithelial regenerative repair. This study aims to evaluate the role of GATA6 in NF-κB-mediated NLRP3 inflammasome activation and pyroptosis after tracheal injury. Tracheal tissues and serum samples were collected from clinical patients and a rat model of tracheal injury. Upon GATA6 knockdown or overexpression, BEAS-2B and rat tracheal epithelial (RTE) cells were treated with lipopolysaccharides and nigericin before being co-cultured with primary tracheal fibroblasts. The changes of NLRP3 inflammasome activation and pyroptosis and their underlying mechanisms were detected. Additionally, the role of GATA6 downregulation in tracheal injury was verified in rats. GATA6 expression and NLRP3 inflammasome activation were upregulated following tracheal injury in the epithelium of granulation tissues. GATA6 silencing inhibited NLRP3 priming, NLRP3 inflammasome activation, and pyroptosis in BEAS-2B and RTE cells. Mechanistically, GATA6 was determined to have bound to the promoter region of NLRP3 and synergistically upregulated NLRP3 promoter activity with NF-κB. Furthermore, GATA6 overexpression promoted epithelial-mesenchymal transition via modulating the NF-κB/NLRP3 pathway. Epithelial NLRP3 inflammasome activation triggered ECM production in fibroblasts, which was suppressed by GATA6 knockdown and induced by GATA6 overexpression. Finally, the downregulation of GATA6 alleviated NLRP3 inflammasome-mediated pyroptosis induced by tracheal injury in rats, thereby reducing tracheal stenosis, inflammation, and fibrosis. GATA6 promotes fibrotic repair in tracheal injury through NLRP3 inflammasome-mediated epithelial pyroptosis, making it a potential biological therapeutic target for tracheal injury.
Collapse
Affiliation(s)
- Anmao Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Gu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunyan He
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yishi Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingyu Peng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiaxin Liao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Xiao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuliang Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
9
|
Hassan GS, Flores Molina M, Shoukry NH. The multifaceted role of macrophages during acute liver injury. Front Immunol 2023; 14:1237042. [PMID: 37736102 PMCID: PMC10510203 DOI: 10.3389/fimmu.2023.1237042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/15/2023] [Indexed: 09/23/2023] Open
Abstract
The liver is situated at the interface of the gut and circulation where it acts as a filter for blood-borne and gut-derived microbes and biological molecules, promoting tolerance of non-invasive antigens while driving immune responses against pathogenic ones. Liver resident immune cells such as Kupffer cells (KCs), a subset of macrophages, maintain homeostasis under physiological conditions. However, upon liver injury, these cells and others recruited from circulation participate in the response to injury and the repair of tissue damage. Such response is thus spatially and temporally regulated and implicates interconnected cells of immune and non-immune nature. This review will describe the hepatic immune environment during acute liver injury and the subsequent wound healing process. In its early stages, the wound healing immune response involves a necroinflammatory process characterized by partial depletion of resident KCs and lymphocytes and a significant infiltration of myeloid cells including monocyte-derived macrophages (MoMFs) complemented by a wave of pro-inflammatory mediators. The subsequent repair stage includes restoring KCs, initiating angiogenesis, renewing extracellular matrix and enhancing proliferation/activation of resident parenchymal and mesenchymal cells. This review will focus on the multifaceted role of hepatic macrophages, including KCs and MoMFs, and their spatial distribution and roles during acute liver injury.
Collapse
Affiliation(s)
- Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Manuel Flores Molina
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
10
|
Chia DKA, Demuytere J, Ernst S, Salavati H, Ceelen W. Effects of Hyperthermia and Hyperthermic Intraperitoneal Chemoperfusion on the Peritoneal and Tumor Immune Contexture. Cancers (Basel) 2023; 15:4314. [PMID: 37686590 PMCID: PMC10486595 DOI: 10.3390/cancers15174314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/12/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Hyperthermia combined with intraperitoneal (IP) drug delivery is increasingly used in the treatment of peritoneal metastases (PM). Hyperthermia enhances tumor perfusion and increases drug penetration after IP delivery. The peritoneum is increasingly recognized as an immune-privileged organ with its own distinct immune microenvironment. Here, we review the immune landscape of the healthy peritoneal cavity and immune contexture of peritoneal metastases. Next, we review the potential benefits and unwanted tumor-promoting effects of hyperthermia and the associated heat shock response on the tumor immune microenvironment. We highlight the potential modulating effect of hyperthermia on the biomechanical properties of tumor tissue and the consequences for immune cell infiltration. Data from translational and clinical studies are reviewed. We conclude that (mild) hyperthermia and HIPEC have the potential to enhance antitumor immunity, but detailed further studies are required to distinguish beneficial from tumor-promoting effects.
Collapse
Affiliation(s)
- Daryl K. A. Chia
- Department of Surgery, National University Hospital, National University Health System, Singapore 119074, Singapore
| | - Jesse Demuytere
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Sam Ernst
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Hooman Salavati
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Wim Ceelen
- Department of Human Structure and Repair, Experimental Surgery Lab, Ghent University, 9052 Ghent, Belgium; (J.D.); (S.E.); (H.S.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Department of GI Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| |
Collapse
|
11
|
Li A, Gu L, Mu J, Li Y, Wang X, Jiang J, Bai Y, Yang M, He C, Xiao R, Liao J, Jin X, Xiao M, Xiao Y, Zhang X, Tan T, Peng M, Xu L, Guo S. GATA6 triggers fibroblast activation and tracheal fibrosis through the Wnt/β-catenin pathway. Cell Signal 2023; 105:110593. [PMID: 36682592 DOI: 10.1016/j.cellsig.2023.110593] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/21/2022] [Accepted: 01/10/2023] [Indexed: 01/21/2023]
Abstract
Tracheal fibrosis is a key abnormal repair process leading to fatal stenosis, characterized by excessive fibroblast activation and extracellular matrix (ECM) deposition. GATA6, a zinc finger-containing transcription factor, is involved in fibroblast activation, while its role in tracheal fibrosis remains obscure. The present study investigated the potential role of GATA6 as a novel regulator of tracheal fibrosis. It was found that GATA6 and α-smooth muscle actin (α-SMA) were obviously increased in tracheal fibrotic granulations and in TGFβ1-treated primary tracheal fibroblasts. GATA6 silencing inhibited TGFβ1-stimulated fibroblast proliferation and ECM synthesis, promoted cell apoptosis, and inactivated Wnt/β-catenin pathway, whereas GATA6 overexpression showed the reverse effects. SKL2001, an agonist of Wnt/β-catenin signaling, restored collagen1a1 and α-SMA expression which was suppressed by GATA6 silencing. Furthermore, in vivo, knockdown of GATA6 ameliorated tracheal fibrosis, as manifested by reduced tracheal stenosis and ECM deposition. GATA6 inhibition in rat tracheas also impaired granulation proliferation, increased apoptosis, and inactivated Wnt/β-catenin pathway. In conclusion, our findings indicate that GATA6 triggers fibroblast activation, cell proliferation, and apoptosis resistance in tracheal fibrosis via the Wnt/β-catenin signaling pathway. Targeting GATA6 may represent a promising therapeutic approach for tracheal fibrosis.
Collapse
Affiliation(s)
- Anmao Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Gu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junhao Mu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yishi Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaohui Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinyue Jiang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Bai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingjin Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunyan He
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Xiao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiaxin Liao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xingxing Jin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Meiling Xiao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Xiao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xia Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tairong Tan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingyu Peng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Shuliang Guo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
12
|
Yoshihara T, Okabe Y. Aldh1a2 + fibroblastic reticular cells regulate lymphocyte recruitment in omental milky spots. J Exp Med 2023; 220:213908. [PMID: 36880532 PMCID: PMC9997506 DOI: 10.1084/jem.20221813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/29/2022] [Accepted: 02/03/2023] [Indexed: 03/06/2023] Open
Abstract
Lymphoid clusters in visceral adipose tissue omentum, known as milky spots, play a central role in the immunological defense in the abdomen. Milky spots exhibit hybrid nature between secondary lymph organs and ectopic lymphoid tissues, yet their development and maturation mechanisms are poorly understood. Here, we identified a subset of fibroblastic reticular cells (FRCs) that are uniquely present in omental milky spots. These FRCs were characterized by the expression of retinoic acid-converting enzyme, Aldh1a2, and endothelial cell marker, Tie2, in addition to canonical FRC-associated genes. Diphtheria toxin-mediated ablation of Aldh1a2+ FRCs resulted in the alteration in milky spot structure with a significant reduction in size and cellularity. Mechanistically, Aldh1a2+ FRCs regulated the display of chemokine CXCL12 on high endothelial venules (HEVs), which recruit blood-borne lymphocytes from circulation. We further found that Aldh1a2+ FRCs are required for the maintenance of peritoneal lymphocyte composition. These results illustrate the homeostatic roles of FRCs in the formation of non-classical lymphoid tissues.
Collapse
Affiliation(s)
- Tomomi Yoshihara
- Laboratory of Immune Homeostasis, WPI Immunology Frontier Research Center, Osaka University , Osaka, Japan
| | - Yasutaka Okabe
- Laboratory of Immune Homeostasis, WPI Immunology Frontier Research Center, Osaka University , Osaka, Japan.,Center for Infectious Disease Education and Research, Osaka University , Osaka, Japan.,Japan Science and Technology Agency , PRESTO, Kawaguchi, Japan
| |
Collapse
|
13
|
Miyamoto T, Murphy B, Zhang N. Intraperitoneal metastasis of ovarian cancer: new insights on resident macrophages in the peritoneal cavity. Front Immunol 2023; 14:1104694. [PMID: 37180125 PMCID: PMC10167029 DOI: 10.3389/fimmu.2023.1104694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
Ovarian cancer metastasis occurs primarily in the peritoneal cavity. Orchestration of cancer cells with various cell types, particularly macrophages, in the peritoneal cavity creates a metastasis-favorable environment. In the past decade, macrophage heterogeneities in different organs as well as their diverse roles in tumor settings have been an emerging field. This review highlights the unique microenvironment of the peritoneal cavity, consisting of the peritoneal fluid, peritoneum, and omentum, as well as their own resident macrophage populations. Contributions of resident macrophages in ovarian cancer metastasis are summarized; potential therapeutic strategies by targeting such cells are discussed. A better understanding of the immunological microenvironment in the peritoneal cavity will provide a stepping-stone to new strategies for developing macrophage-based therapies and is a key step toward the unattainable eradication of intraperitoneal metastasis of ovarian cancer.
Collapse
Affiliation(s)
- Taito Miyamoto
- Immunology, Metastasis & Microenvironment Program, Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA, United States
| | | | - Nan Zhang
- Immunology, Metastasis & Microenvironment Program, Ellen and Ronald Caplan Cancer Center, The Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
14
|
Jiang T, Zhang H, Li Y, Jayakumar P, Liao H, Huang H, Billiar TR, Deng M. Intraperitoneal injection of class A TLR9 agonist enhances anti-PD-1 immunotherapy in colorectal peritoneal metastases. JCI Insight 2022; 7:e160063. [PMID: 36278484 PMCID: PMC9714777 DOI: 10.1172/jci.insight.160063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/31/2022] [Indexed: 07/02/2024] Open
Abstract
Peritoneal metastases are associated with a low response rate to immune checkpoint blockade (ICB) therapy. The numbers of peritoneal resident macrophages (PRMs) are reversely correlated with the response rate to ICB therapy. We have previously shown that TLR9 in fibroblastic reticular cells (FRCs) plays a critical role in regulating peritoneal immune cell recruitment. However, the role of TLR9 in FRCs in regulating PRMs is unclear. Here, we demonstrated that the class A TLR9 agonist, ODN1585, markedly enhanced the efficacy of anti-PD-1 therapy in mouse models of colorectal peritoneal metastases. ODN1585 injected i.p. reduced the numbers of Tim4+ PRMs and enhanced CD8+ T cell antitumor immunity. Mechanistically, treatment of ODN1585 suppressed the expression of genes required for retinoid metabolism in FRCs, and this was associated with reduced expression of the PRM lineage-defining transcription factor GATA6. Selective deletion of TLR9 in FRCs diminished the benefit of ODN1585 in anti-PD-1 therapy in reducing peritoneal metastases. The crosstalk between PRMs and FRCs may be utilized to develop new strategies to improve the efficacy of ICB therapy for peritoneal metastases.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Tsinghua University School of Medicine, Beijing, China
| | - Hongji Zhang
- Department of Surgery, The Ohio State University, Columbus, Ohio, USA
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Yiming Li
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Preethi Jayakumar
- Department of Surgery, The Ohio State University, Columbus, Ohio, USA
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Hong Liao
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hai Huang
- Department of Surgery, The Ohio State University, Columbus, Ohio, USA
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Meihong Deng
- Department of Surgery, The Ohio State University, Columbus, Ohio, USA
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|