1
|
Meng C, Lin K, Shi W, Teng H, Wan X, DeBruine A, Wang Y, Liang X, Leo J, Chen F, Gu Q, Zhang J, Van V, Maldonado KL, Gan B, Ma L, Lu Y, Zhao D. Histone methyltransferase ASH1L primes metastases and metabolic reprogramming of macrophages in the bone niche. Nat Commun 2025; 16:4681. [PMID: 40394007 PMCID: PMC12092585 DOI: 10.1038/s41467-025-59381-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 04/22/2025] [Indexed: 05/22/2025] Open
Abstract
Bone metastasis is a major cause of cancer death; however, the epigenetic determinants driving this process remain elusive. Here, we report that histone methyltransferase ASH1L is genetically amplified and is required for bone metastasis in men with prostate cancer. ASH1L rewires histone methylations and cooperates with HIF-1α to induce pro-metastatic transcriptome in invading cancer cells, resulting in monocyte differentiation into lipid-associated macrophage (LA-TAM) and enhancing their pro-tumoral phenotype in the metastatic bone niche. We identified IGF-2 as a direct target of ASH1L/HIF-1α and mediates LA-TAMs' differentiation and phenotypic changes by reprogramming oxidative phosphorylation. Pharmacologic inhibition of the ASH1L-HIF-1α-macrophages axis elicits robust anti-metastasis responses in preclinical models. Our study demonstrates epigenetic alterations in cancer cells reprogram metabolism and features of myeloid components, facilitating metastatic outgrowth. It establishes ASH1L as an epigenetic driver priming metastasis and macrophage plasticity in the bone niche, providing a bona fide therapeutic target in metastatic malignancies.
Collapse
Affiliation(s)
- Chenling Meng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wei Shi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hongqi Teng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xinhai Wan
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Anna DeBruine
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Yin Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xin Liang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Javier Leo
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Feiyu Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Qianlin Gu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vivien Van
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kiersten L Maldonado
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Di Zhao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Castellano M, Blanco V, Li Calzi M, Costa B, Witwer K, Hill M, Cayota A, Segovia M, Tosar JP. Ribonuclease activity undermines immune sensing of naked extracellular RNA. CELL GENOMICS 2025; 5:100874. [PMID: 40334662 DOI: 10.1016/j.xgen.2025.100874] [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: 04/27/2024] [Revised: 02/26/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025]
Abstract
Cell membranes are thought of as barriers to extracellular RNA (exRNA) uptake. While naked exRNAs can be spontaneously internalized by certain cells, functional cytosolic delivery has been rarely observed. Here, we show that extracellular ribonucleases (RNases)-primarily from cell culture supplements-have obscured the study of exRNA functionality. When ribonuclease inhibitor (RI) is added to cell cultures, naked exRNAs can trigger pro-inflammatory responses in dendritic cells and macrophages, largely via endosomal Toll-like receptors (TLRs). Moreover, naked exRNAs can escape endosomes, engaging cytosolic RNA sensors. In addition, naked extracellular mRNAs can be spontaneously internalized and translated by various cell types in an RI-dependent manner. In vivo, RI co-injection amplifies naked-RNA-induced activation of splenic lymphocytes and myeloid leukocytes. Furthermore, naked RNA is inherently pro-inflammatory in RNase-poor compartments like the peritoneal cavity. These findings demonstrate that naked RNA is bioactive without requiring vesicular encapsulation, making a case for nonvesicular-exRNA-mediated intercellular communication.
Collapse
Affiliation(s)
- Mauricio Castellano
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Immunoregulation and Inflammation Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Analytical Biochemistry Unit, School of Science, Universidad de la República, Montevideo 11400, Uruguay
| | - Valentina Blanco
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Marco Li Calzi
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Bruno Costa
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Analytical Biochemistry Unit, School of Science, Universidad de la República, Montevideo 11400, Uruguay
| | - Kenneth Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; EV Core Facility "EXCEL," Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Marcelo Hill
- Immunoregulation and Inflammation Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Academic Unit of Immunobiology, School of Medicine, Universidad de la República, Montevideo 11800, Uruguay
| | - Alfonso Cayota
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Hospital de Clínicas, Universidad de la República, Montevideo 11600, Uruguay
| | - Mercedes Segovia
- Immunoregulation and Inflammation Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Academic Unit of Immunobiology, School of Medicine, Universidad de la República, Montevideo 11800, Uruguay.
| | - Juan Pablo Tosar
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay; Analytical Biochemistry Unit, School of Science, Universidad de la República, Montevideo 11400, Uruguay.
| |
Collapse
|
3
|
Yao W, Liu M, Li Z, Qu L, Sui S, Xiang C, Jiang L, Wang S, Liu G, Chen Y, Yang L. CD38 hi macrophages promote fibrotic transition following acute kidney injury by modulating NAD + metabolism. Mol Ther 2025:S1525-0016(25)00318-1. [PMID: 40349105 DOI: 10.1016/j.ymthe.2025.04.039] [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: 02/24/2025] [Revised: 04/05/2025] [Accepted: 04/25/2025] [Indexed: 05/14/2025] Open
Abstract
Acute kidney injury (AKI) encompasses a spectrum of conditions, varying from mild and self-limiting to severe cases that can lead to chronic kidney disease (CKD). Macrophages are crucial in the progression from AKI to CKD, yet the diversity of macrophage subsets complicates the identification of key functional types. We established a detailed single-cell atlas of mononuclear macrophages from the onset of AKI through its progression to CKD. Our results indicate that a macrophage subset with high CD38 expression is closely linked to renal fibrosis following AKI in both mouse model and AKI patients. These CD38hi macrophages, derived from resident macrophages via Csf1 signaling, secrete the NAD-depleting enzyme CD38, inducing senescence in renal tubular cells and promoting chronic inflammation and renal fibrosis. Knocking out CD38 in macrophages elevated renal NAD levels, reducing senescence and fibrotic responses. Furthermore, we initiated a dosing regimen for a CD38 inhibitor, demonstrating its potential to reduce fibrosis post-AKI, suggesting that targeting CD38hi macrophages-mediated NAD+ metabolism could be a promising therapy to halt AKI to CKD progression.
Collapse
Affiliation(s)
- Weijian Yao
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)-Ministry of Education of China, Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases-Chinese Academy of Medical Sciences, Beijing Key Laboratory of Precision Medicine and New-drug/Equipment Development for Severe Kidney Disease, Peking University First Hospital, Beijing 100034, P.R. China
| | - Menghan Liu
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)-Ministry of Education of China, Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases-Chinese Academy of Medical Sciences, Beijing Key Laboratory of Precision Medicine and New-drug/Equipment Development for Severe Kidney Disease, Peking University First Hospital, Beijing 100034, P.R. China
| | - Zehua Li
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)-Ministry of Education of China, Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases-Chinese Academy of Medical Sciences, Beijing Key Laboratory of Precision Medicine and New-drug/Equipment Development for Severe Kidney Disease, Peking University First Hospital, Beijing 100034, P.R. China
| | - Lei Qu
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)-Ministry of Education of China, Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases-Chinese Academy of Medical Sciences, Beijing Key Laboratory of Precision Medicine and New-drug/Equipment Development for Severe Kidney Disease, Peking University First Hospital, Beijing 100034, P.R. China
| | - Shuang Sui
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)-Ministry of Education of China, Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases-Chinese Academy of Medical Sciences, Beijing Key Laboratory of Precision Medicine and New-drug/Equipment Development for Severe Kidney Disease, Peking University First Hospital, Beijing 100034, P.R. China
| | - Chengang Xiang
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)-Ministry of Education of China, Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases-Chinese Academy of Medical Sciences, Beijing Key Laboratory of Precision Medicine and New-drug/Equipment Development for Severe Kidney Disease, Peking University First Hospital, Beijing 100034, P.R. China
| | - Lei Jiang
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)-Ministry of Education of China, Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases-Chinese Academy of Medical Sciences, Beijing Key Laboratory of Precision Medicine and New-drug/Equipment Development for Severe Kidney Disease, Peking University First Hospital, Beijing 100034, P.R. China
| | - Suxia Wang
- Laboratory of Electron Microscopy Pathological Center, Peking University First Hospital, Beijing 100034, P.R. China
| | - Gang Liu
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)-Ministry of Education of China, Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases-Chinese Academy of Medical Sciences, Beijing Key Laboratory of Precision Medicine and New-drug/Equipment Development for Severe Kidney Disease, Peking University First Hospital, Beijing 100034, P.R. China
| | - Ying Chen
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)-Ministry of Education of China, Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases-Chinese Academy of Medical Sciences, Beijing Key Laboratory of Precision Medicine and New-drug/Equipment Development for Severe Kidney Disease, Peking University First Hospital, Beijing 100034, P.R. China.
| | - Li Yang
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)-Ministry of Education of China, Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases-Chinese Academy of Medical Sciences, Beijing Key Laboratory of Precision Medicine and New-drug/Equipment Development for Severe Kidney Disease, Peking University First Hospital, Beijing 100034, P.R. China.
| |
Collapse
|
4
|
Cheng HY, Chu J, Limjunyawong N, Chen J, Ye Y, Chen KH, Koylass N, Sun S, Dong X, Qiu Z. Phagosome-mediated anti-bacterial immunity is governed by the proton-activated chloride channel in peritoneal macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640612. [PMID: 40060571 PMCID: PMC11888413 DOI: 10.1101/2025.02.27.640612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
The success of phagosome degradation relies on the ability of phagocytes to regulate the maturation of phagosomes. However, its underlying molecular mechanisms remain poorly understood. Here, we identify the proton-activated chloride (PAC) channel as a key negative regulator of phagosome maturation. PAC deletion enhanced phagosomal acidification and protease activities, leading to augmented bacterial killing in large peritoneal macrophages (LPMs) upon Escherichia coli infection in mice. Surprisingly, phagosome degradation also stimulated STING-IRF3-interferon responses and inflammasome activation in LPMs, both of which are enhanced upon PAC deletion. The increased inflammasome activation induced the release of cleaved gasdermin D, which localized to the surface of bacteria in the peritoneum and further contributed to their killing. Finally, enhanced bacterial clearance by PAC-deficient LPMs reduced proinflammatory immune cell infiltration and peritoneal inflammation, resulting in improved survival in mice. Our study thus provides new insights into the molecular mechanism of phagosome maturation and the dynamics of host defense response following phagosome-mediated bacterial degradation in peritoneal macrophages. Summary The PAC channel mediates phagosome maturation during bacterial infection in macrophages. PAC deletion promotes phagosome-mediated STING-interferon signaling and inflammasome-mediated gasdermin D secretion during bacterial infection in peritoneal macrophages.
Collapse
|
5
|
Lebegge E, Kancheva D, Van Craenenbroeck J, Ernst S, Bardet PMR, Caro AA, Kiss M, Jumapili NA, Barthelmess RM, Zivalj M, Assaf N, Ali L, Elkrim Y, Demuytere J, De Jonge J, Raes G, Hadadi E, Devoogdt N, Vincke C, Mohavedi K, Vereecke L, Ceelen W, Stijlemans B, Laoui D, Arnouk SM, Van Ginderachter JA. VSIG4-Expressing Macrophages Contribute to Antiparasitic and Antimetastatic Responses in the Peritoneal Cavity. Eur J Immunol 2025; 55:e202551804. [PMID: 40346775 PMCID: PMC12064866 DOI: 10.1002/eji.202551804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/12/2025]
Abstract
Large peritoneal macrophages (LPMs) play a role as gatekeepers of peritoneal homeostasis by providing a first line of defense against pathogens. A third of the LPMs express the surface receptor VSIG4, but it is unclear whether these cells differ from their VSIG4-negative counterparts and perform dedicated functions. We demonstrate that VSIG4+, but not VSIG4-, LPMs are in the majority derived from embryonal precursors, and their occurrence is largely independent of sex and microbiota but increases with age. Although their transcriptome and surface proteome are indistinguishable from VSIG4- LPMs at steady-state, VSIG4+ LPMs are superior in phagocytosing S. aureus bioparticles and colorectal carcinoma (CRC) cells. Anti-VSIG4 nanobody constructs that are ADCC-enabled allowed a selective elimination of the VSIG4+ LPM subset without affecting overall LPM abundance. This strategy uncovered a role for VSIG4+ LPMs in lowering the first peak of parasitemia in a Trypanosoma brucei brucei infection model and in reducing CRC outgrowth in the peritoneal cavity, a prime metastatic site in CRC patients. Altogether, our data uncover a protective role for VSIG4+ LPMs in infectious and oncological diseases in the peritoneal cavity.
Collapse
|
6
|
Qin Y, Wang X, Zhang X, Nong L, Hou Q, Chen Y, Li Y, Lin W, Mao X, Wu K, Nong W, Wang T, Meng L, Song J. Retinoic acid modulates peritoneal macrophage function and distribution to enhance antibacterial defense during inflammation. Front Nutr 2025; 12:1545720. [PMID: 40370796 PMCID: PMC12075188 DOI: 10.3389/fnut.2025.1545720] [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: 12/15/2024] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Background Peritoneal macrophages, comprising large peritoneal macrophages (LPMs) and small peritoneal macrophages (SPMs), play a vital role in maintaining immune defenses during inflammation. However, the molecular mechanisms governing their responses, particularly the impact of retinoic acid (RA), remain poorly understood. This study aims to elucidate the role of RA in modulating macrophage function, distribution, and immune responses during bacterial infections. Methods A murine model of peritonitis was established using Escherichia coli expressing a tdTomato fluorescence marker. The effects of RA on macrophage phagocytic capacity, population dynamics, and transcriptomic profiles were assessed using immunofluorescence, flow cytometry, RNA sequencing, and quantitative PCR. Additionally, RA-loaded ZIF-8 nanoparticles were employed to investigate the sustained effects of RA delivery. Results RA significantly enhanced macrophage phagocytic activity, delayed functional decline, and promoted the recruitment of SPMs in the peritoneal cavity. Transcriptomic analysis revealed upregulation of leukocyte migration and cell adhesion pathways in RA-treated SPMs. RA treatment also induced distinct gene expression profiles in macrophage subpopulations, reflecting its role in immune modulation. Notably, RA-loaded ZIF-8 nanoparticles prolonged RA retention within macrophages, sustaining its effects. Conclusion RA enhances antibacterial defense by modulating macrophage activity, providing new insights into immune regulation. These findings underscore the therapeutic potential of RA and its nanoparticle formulations in managing bacterial infections and inflammation.
Collapse
Affiliation(s)
- Yujuan Qin
- Graduate School, Youjiang Medical University for Nationalities, Baise, China
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Xi Wang
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
- School of Medicine, Guangxi University, Nanning, China
| | - Xiamin Zhang
- Graduate School, Youjiang Medical University for Nationalities, Baise, China
- Department of Digestive Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Lianting Nong
- Graduate School, Youjiang Medical University for Nationalities, Baise, China
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Qiyan Hou
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Yuhong Chen
- Graduate School, Youjiang Medical University for Nationalities, Baise, China
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Yuting Li
- Graduate School, Youjiang Medical University for Nationalities, Baise, China
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Wenxian Lin
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Xiuli Mao
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Kezhao Wu
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Wenqian Nong
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Tonghua Wang
- Department of Digestive Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Lingzhang Meng
- Graduate School, Youjiang Medical University for Nationalities, Baise, China
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Jian Song
- Graduate School, Youjiang Medical University for Nationalities, Baise, China
- Institute of Cardiovascular Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| |
Collapse
|
7
|
Du X, Huang J, Zhao C, Hu Z, Zhang L, Xu Z, Liu X, Li X, Zhang Z, Guo S, Yin T, Wang G. Retrospective perspectives and future trends in nanomedicine treatment: from single membranes to hybrid membranes. NANOSCALE 2025; 17:9738-9763. [PMID: 40136036 DOI: 10.1039/d4nr04999c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
At present, various diseases seriously threaten human life and health, and the development of nanodrug delivery systems has brought about a turnaround for traditional drug treatments, with nanoparticles being precisely targeted to improve bioavailability. Surface modification of nanoparticles can prolong blood circulation time and enhance targeting ability. The application of cell membrane-coated nanoparticles further improves their biocompatibility and active targeting ability, providing new hope for the treatment of various diseases. Various types of cell membrane biomimetic nanoparticles have gradually attracted increasing attention due to their unique advantages. However, the pathological microenvironment of different diseases is complex and varied, and the single-cell membrane has several limitations because a single functional property cannot fully meet the requirements of disease treatment. Hybrid cell membranes integrate the advantages of multiple biological membranes and have become an emerging research hotspot. This review summarizes the application of cell membrane biomimetic nanoparticles in the treatment of various diseases and discusses the advantages, challenges and future development of biomimetic nanoparticles. We propose that the fusion of multiple membranes may be a reasonable trend in the future to provide some ideas and directions for the treatment of various diseases.
Collapse
Affiliation(s)
- Xinya Du
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Junyang Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Chuanrong Zhao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Ziqiu Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | | | - Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xiaoying Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Xinglei Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Zhengcai Zhang
- Lepu Medical Technology (Beijing) Co., Ltd, Beijing, China
| | - Songtao Guo
- College of Computer Science, Chongqing University, Chongqing, China.
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, National Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
- JinFeng Laboratory, Chongqing, China.
| |
Collapse
|
8
|
Pal R, Thomas CM, Salamat K, Jenkins SJ, Bradford BM, Mabbott NA. Acute LPS exposure enhances susceptibility to peripheral prion infection. Sci Rep 2025; 15:9754. [PMID: 40119036 PMCID: PMC11928655 DOI: 10.1038/s41598-025-94003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
After peripheral infections, the initial accumulation of prions within secondary lymphoid tissues is essential for the transmission of disease to the brain. Macrophages are considered to sequester or destroy prions, but little was known of their impact on disease susceptibility after a peripheral infection. Inflammation in the peritoneal cavity can trigger the macrophage disappearance reaction, whereby the macrophages are temporarily contained within cellular aggregates on the mesothelium. We studied the impact of the bacterial lipopolysaccharide (LPS)-mediated macrophage disappearance reaction on susceptibility to an intraperitoneal prion infection. Intraperitoneal LPS injection significantly enhanced prion disease susceptibility approximately 100X when given 24-3 h before infection. The effects on disease susceptibility coincided with the reduced abundance of macrophages within the peritoneal cavity at the time of infection and the enhanced early accumulation of prions in the spleen. This suggests that the reduced recoverable abundance of macrophages in the peritoneal cavity following acute LPS-treatment, increased disease susceptibility by enhancing the initial propagation of the prions from site of exposure (peritoneal cavity) to the spleen from where they subsequently spread to the brain. Further studies may help identify novel macrophage-targeted treatments that can reduce susceptibility to peripherally acquired prion infections.
Collapse
Affiliation(s)
- Reiss Pal
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Charlotte M Thomas
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Khalid Salamat
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Stephen J Jenkins
- Queens Medical Research Institute, University of Edinburgh Centre for Inflammation Research, Edinburgh, UK
| | - Barry M Bradford
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK.
| | - Neil A Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Entsie P, Amoafo EB, Kang Y, Gustad T, Dorsam GP, Frey MR, Liverani E. Sex-specific activation of platelet purinergic signaling is key in local cytokine release and phagocytosis in the peritoneal cavity in intra-abdominal sepsis. Am J Physiol Cell Physiol 2025; 328:C791-C805. [PMID: 39854048 DOI: 10.1152/ajpcell.00116.2024] [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: 02/20/2024] [Revised: 03/07/2024] [Accepted: 01/17/2025] [Indexed: 01/26/2025]
Abstract
Intra-abdominal sepsis is a life-threatening complex syndrome caused by microbes in the gut microbiota invading the peritoneal cavity. It is one of the major complications of intra-abdominal surgery. To date, only supportive therapies are available. No studies have investigated the progression of intra-abdominal sepsis in the peritoneal cavity. Our group has shown that platelets play an essential role during sepsis, and blocking purinergic signaling in platelets through P2Y1 and P2Y12 antagonism significantly lowered inflammatory levels and improved survival in a murine model of sepsis. Here, we tested whether antagonizing purinergic signaling in platelets in the peritoneal cavity can reduce the local release of cytokines and modulate platelet interaction with the immune system. We used cecal ligation and puncture (CLP) to induce sepsis followed by intraperitoneal administration of MRS2279 (P2Y1 antagonist) or ticagrelor (P2Y12 antagonist) in male and female mice. The peritoneal cavity fluid (PCF) was collected 4 or 24 h post-CLP and analyzed for cell recruitment, platelet markers, cytokines, and platelet immune cell interactions. Platelet markers were increased 24 h after CLP, although the total platelet count in the peritoneal cavity was lower than the blood. Blocking P2Y12 or P2Y1 improved bacterial clearance in the PCF in a sex-dependent manner. The influx of immune cells in the peritoneal cavity was altered by blocking P2Y12 or P2Y1 sex-dependently. Blocking P2Y1 and P2Y12 receptors can enhance the phagocytic activity in the peritoneal cavity in a sex- and time-related manner, and platelets significantly contribute to the development and progression of sepsis in the peritoneal cavity.NEW & NOTEWORTHY Intra-abdominal sepsis is a challenging complication postabdominal surgery caused by perforations of the gastrointestinal tract where microbes invade the peritoneal cavity. This leads to local cytokine release and immune cell dysfunction. Our data identify platelets as key players in mediating inflammation in intra-abdominal sepsis. We have shown that blocking purinergic signaling in the peritoneal cavity reduced cytokine release and cell-cell interactions differently in males and females, hence a sex-specific strategy to improve intra-abdominal sepsis.
Collapse
Affiliation(s)
- Philomena Entsie
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Emmanuel Boadi Amoafo
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Ying Kang
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Thomas Gustad
- Department of Microbiological Sciences, College of Agriculture, Food Systems and Natural Resources, North Dakota State University, Fargo, North Dakota, United States
| | - Glenn P Dorsam
- Department of Microbiological Sciences, College of Agriculture, Food Systems and Natural Resources, North Dakota State University, Fargo, North Dakota, United States
| | - Mark R Frey
- Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Elisabetta Liverani
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| |
Collapse
|
12
|
Yang Y, Hu Q, Shao Q, Peng Y, Yu B, Luo F, Chen J, Xu C, Li Z, Tam M, Ju Z, Zhang R, Xing F, Zha Z, Zhang H. A Baicalin-Based Functional Polymer in Dynamic Reversible Networks Alleviates Osteoarthritis by Cellular Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410951. [PMID: 39840483 PMCID: PMC11904974 DOI: 10.1002/advs.202410951] [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: 09/07/2024] [Revised: 12/07/2024] [Indexed: 01/23/2025]
Abstract
Osteoarthritis (OA) is increasingly recognized as a whole-organ disease predominantly affecting the elderly, characterized by typical alterations in subchondral bone and cartilage, along with recurrent synovial inflammation. Despite the availability of various therapeutics and medications, a complete resolution of OA remains elusive. In this study, novel functional hydrogels are developed by integrating natural bioactive molecules for OA treatment. Specifically, baicalin (Bai) is combined with 2-hydroxyethyl acrylate (HEA) to form a polymerizable monomer (HEA-Bai) through esterification, which is subjected to reversible addition-fragmentation chain transfer (RAFT) polymerization to produce Bai-based polymer (Pm). These macromolecules are incorporated into Schiff-base hydrogels, which demonstrate excellent mechanical properties and self-healing performance. Notably, the Bai-based formulations are taken up by fibroblast-like synoviocytes (FLSs), where they regulate glycolysis. Mechanistically, inhibition of yes-associated protein 1 (YAP1) by the formulations suppressed the FLSs glycolysis and reduced the secretion of inflammatory factors, including interleukin 1β (IL-1β), IL-6, and IL-8. Furthermore, the functional hydrogel (AG-Pm)-OC, severing as a lubricant and nutrient, prolonged joint retention of Bai, thereby reducing cartilage degradation and synovial inflammation. Meanwhile, (AG-Pm)-OC alleviated joint pain by targeting the YAP1 signaling and inhibiting macrophage recruitment and polarization. Taken together, this flavonoid-based injectable hydrogel exhibits enhanced biocompatibility and efficacy against OA.
Collapse
Affiliation(s)
- Yili Yang
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
- Department of Immunobiology, College of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Qinxiao Hu
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Qingfeng Shao
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Yachen Peng
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Bo Yu
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Fangji Luo
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Jiajing Chen
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Chenhao Xu
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Zhenyan Li
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Manseng Tam
- Macau Medical Science and Technology Research AssociationMacao999078China
| | - Zhenyu Ju
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Ronghua Zhang
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine InformatizationCollege of PharmacyJinan UniversityGuangzhouGuangdong510630China
| | - Feiyue Xing
- Department of ImmunobiologyCollege of Life Science and TechnologyJinan UniversityGuangzhouGuangdong510632China
- Department of ImmunobiologyMOE Key Laboratory of Tumor Molecular BiologyJinan UniversityGuangzhou510632China
| | - Zhengang Zha
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| | - Huan‐Tian Zhang
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Jinan UniversityKey Laboratory of Regenerative Medicine of Ministry of EducationJinan UniversityGuangzhouGuangdong510630China
| |
Collapse
|
13
|
Moerings BGJ, Govers C, van Bergenhenegouwen J, Mes JJ, van Dijk M, Witkamp RF, van Norren K, Abbring S. Induction of endotoxin tolerance in murine monocyte and macrophage cell populations - optimal LPS dose and compartment-specific reversal by β-glucan. Food Funct 2025; 16:1576-1587. [PMID: 39917820 PMCID: PMC11803501 DOI: 10.1039/d4fo05223d] [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: 10/25/2024] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Beta-glucans, naturally present in foods like wheat, mushrooms, and yeast, have shown potential in reversing immunosuppression. However, the existing evidence solely relies on ex vivo studies assessing direct effects of β-glucans on macrophages. To investigate whether such effects also occur after their oral administration, this study first systematically examined the immunosuppressive effects of LPS in mice. Subsequently, we assessed the ability of yeast-derived whole β-glucan particles (yWGP), administered through the diet, to counteract LPS-induced immunological tolerance. Immunosuppression following intraperitoneal administration of 20, 200, or 2000 μg kg-1 LPS was demonstrated by reduced TNF-α and IL-6 release upon ex vivo LPS stimulation of immune cells harvested from the peritoneal fluid, spleen, and bone marrow. Immunosuppression in blood was detected only after 200 and 2000 μg kg-1 LPS. LPS tolerance extended to heterologous stimuli (PAM3Cys, heat-killed Pseudomonas aeruginosa), indicating cross-tolerance. Due to animal discomfort at 2000 μg kg-1 LPS, as evidenced by a significantly enhanced clinical severity score, a dose of 200 μg kg-1 LPS was selected for the follow-up trial. In this experiment, mice fed a yWGP-supplemented diet for two weeks prior to LPS administration showed effective reversal of LPS tolerance, reflected by restored TNF-α levels in peritoneal cells but not in other monocyte- and macrophage-containing cell populations. Together, these studies demonstrate that peritoneal administration of 200 μg kg-1 LPS induced ex vivo LPS tolerance in all immunological organs studied, without significantly compromising animal welfare. The selective efficacy of dietary β-glucans to counteract immunosuppression, which is often observed in vulnerable and immunocompromised patient populations, warrants further clinical evaluation.
Collapse
Affiliation(s)
- Bart G J Moerings
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Coen Govers
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Jurriaan J Mes
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Renger F Witkamp
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
| | - Klaske van Norren
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
| | - Suzanne Abbring
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
- Wageningen Food and Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
14
|
Shirazi R, Morrison J. The Emerging Role of Pleural Macrophages in Influenza Defense. DNA Cell Biol 2025; 44:127-131. [PMID: 39868992 DOI: 10.1089/dna.2024.0283] [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] [Indexed: 01/28/2025] Open
Abstract
The pleural cavity is gaining recognition as an important player in lung infections. Our recent research revealed that pleural macrophages (PMs) migrate from the pleural cavity into the lung during influenza virus infection, contributing to improved disease outcomes. This summary highlights key findings on the role of PMs in influencing viral lung infection outcomes and explores the potential directions for advancing this emerging field of study.
Collapse
Affiliation(s)
- Roksana Shirazi
- Department of Microbiology, University of California Riverside, Riverside, California, USA
| | - Juliet Morrison
- Department of Microbiology, University of California Riverside, Riverside, California, USA
| |
Collapse
|
15
|
Pellegrini JM, González-Espinoza G, Shayan RR, Hysenaj L, Rouma T, Arce-Gorvel V, Lelouard H, Popoff D, Zhao Y, Hanniffy S, Castillo-Zeledón A, Loperena-Barber M, Celis-Gutierrez J, Mionnet C, Bosilkovski M, Solera J, Muraille E, Barquero-Calvo E, Moreno E, Conde-Álvarez R, Moriyón I, Gorvel JP, Mémet S. Brucella abortus impairs T lymphocyte responsiveness by mobilizing IL-1RA-secreting omental neutrophils. Nat Commun 2025; 16:862. [PMID: 39833171 PMCID: PMC11747348 DOI: 10.1038/s41467-024-55799-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/30/2024] [Indexed: 01/22/2025] Open
Abstract
Immune evasion strategies of Brucella, the etiologic agent of brucellosis, a global zoonosis, remain partially understood. The omentum, a tertiary lymphoid organ part of visceral adipose tissue, has never been explored as a Brucella reservoir. We report that B. abortus infects and replicates within murine omental macrophages. Throughout the chronic phase of infection, the omentum accumulates macrophages, monocytes and neutrophils. The maintenance of PD-L1+Sca-1+ macrophages, monocytes and neutrophils in the omentum depends on the wadC-encoded determinant of Brucella LPS. We demonstrate that PD-L1+Sca-1+ murine omental neutrophils produce high levels of IL-1RA leading to T cell hyporesponsiveness. These findings corroborate brucellosis patient analysis of whole blood displaying upregulation of PDL1 and Ly6E genes, and of serum exhibiting high levels of IL-1RA. Overall, the omentum, a reservoir for B. abortus, promotes bacterial persistence and causes CD4+ and CD8+ T cell immunosuppression by IL-1RA secreted by PD-L1+Sca-1+ neutrophils.
Collapse
Affiliation(s)
| | | | | | - Lisiena Hysenaj
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Thomas Rouma
- Unité de Recherche en Biologie des Microorganismes (URBM)-Laboratoire d'Immunologie et de Microbiologie, NARILIS, University of Namur, Namur, Belgium
- Laboratoire de de Parasitologie, and ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles, Gosselies, Belgium
| | | | - Hugues Lelouard
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Dimitri Popoff
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Yun Zhao
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Sean Hanniffy
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Amanda Castillo-Zeledón
- Universidad Nacional, Pathology Department, Escuela de Medicina Veterinaria, Heredia, Costa Rica
| | - Maite Loperena-Barber
- Universidad de Navarra, Instituto de Salud Tropical e Departamento de Microbiología y Parasitología, Pamplona, Spain
| | | | - Cyrille Mionnet
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Mile Bosilkovski
- University Clinic for Infectious Diseases and Febrile Conditions, Skopje, Republic of North Macedonia
| | - Javier Solera
- Hospital General Universitario, Facultad de Medicina, Universidad Castilla la Mancha Albacete, Albacete, Spain
| | - Eric Muraille
- Unité de Recherche en Biologie des Microorganismes (URBM)-Laboratoire d'Immunologie et de Microbiologie, NARILIS, University of Namur, Namur, Belgium
- Laboratoire de de Parasitologie, and ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles, Gosselies, Belgium
| | - Elías Barquero-Calvo
- Universidad Nacional, Pathology Department, Escuela de Medicina Veterinaria, Heredia, Costa Rica
| | - Edgardo Moreno
- Universidad Nacional, Pathology Department, Escuela de Medicina Veterinaria, Heredia, Costa Rica
| | - Raquel Conde-Álvarez
- Universidad de Navarra, Instituto de Salud Tropical e Departamento de Microbiología y Parasitología, Pamplona, Spain
| | - Ignacio Moriyón
- Universidad de Navarra, Instituto de Salud Tropical e Departamento de Microbiología y Parasitología, Pamplona, Spain
| | | | - Sylvie Mémet
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France.
| |
Collapse
|
16
|
Freitas IL, Macedo MF, Oliveira L, Oliveira P, do Vale A, dos Santos NM. AIP56, an AB toxin secreted by Photobacterium damselae subsp. piscicida, has tropism for myeloid cells. Front Immunol 2025; 15:1527088. [PMID: 39872526 PMCID: PMC11769971 DOI: 10.3389/fimmu.2024.1527088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction The AB-type toxin AIP56 is a key virulence factor of Photobacterium damselae subsp. piscicida (Phdp), inducing apoptosis in fish immune cells. The discovery of AIP56-like and AIP56-related toxins in diverse organisms, including human-associated Vibrio strains, highlights the evolutionary conservation of this toxin family, suggesting that AIP56 and its homologs may share conserved receptors across species. These toxins have potential for biotechnological applications, such as therapeutic protein delivery and immune modulation. Methods Herein, the cell specificity of AIP56 for immune cells was characterized. The tropism of AIP56 for cells of the sea bass, mouse and human immune system was analyzed by following toxin internalization by flow cytometry and arrival of the toxin in the cytosol by evaluating the cleavage of NF-kB p65 by western blotting. Results Only a small population of sea bass neutrophils internalized AIP56, indicating that most of the neutrophilic destruction during Phdp infection and/or AIP56 intoxication does not result from the direct action of the toxin. Moreover, the cellular tropism of AIP56 for myeloid cells was observed in the three species, including its preference for macrophages. Further, mouse and human M0 and M2-like macrophages internalized more toxin than M1-like macrophages. Despite the limited interaction of lymphoid cells with AIP56, mouse B1-cells were able to internalize the toxin, possibly due to its myeloid features. Conclusion AIP56 has tropism for sea bass, mouse and human myeloid cells, with greater affinity for macrophages. This points to an evolutionary conservation of its receptor(s) and mechanism of action across species, raising the possibility that AIP56-like and -related toxins may also play a role in pathogenesis. These findings are relevant for both pathogenicity and biomedical contexts.
Collapse
Affiliation(s)
- Inês Lua Freitas
- Fish Immunology and Vaccinology Group, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Fish Immunology and Vaccinology Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- McBiology Doctoral Program, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Maria Fátima Macedo
- Cell Activation and Gene Expression, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Cell Activation and Gene Expression, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Liliana Oliveira
- Cell Activation and Gene Expression, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Cell Activation and Gene Expression, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Pedro Oliveira
- EPIUnit, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana do Vale
- Fish Immunology and Vaccinology Group, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Fish Immunology and Vaccinology Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Nuno M.S. dos Santos
- Fish Immunology and Vaccinology Group, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Fish Immunology and Vaccinology Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| |
Collapse
|
17
|
Gallerand A, Han J, Ivanov S, Randolph GJ. Mouse and human macrophages and their roles in cardiovascular health and disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1424-1437. [PMID: 39604762 DOI: 10.1038/s44161-024-00580-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/11/2024] [Indexed: 11/29/2024]
Abstract
The past 15 years have witnessed a leap in understanding the life cycle, gene expression profiles, origins and functions of mouse macrophages in many tissues, including macrophages of the artery wall and heart that have critical roles in cardiovascular health. Here, we review the phenotypical and functional diversity of macrophage populations in multiple organs and discuss the roles that proliferation, survival, and recruitment and replenishment from monocytes have in maintaining macrophages in homeostasis and inflammatory states such as atherosclerosis and myocardial infarction. We also introduce emerging data that better characterize the life cycle and phenotypic profiles of human macrophages. We discuss the similarities and differences between murine and human macrophages, raising the possibility that tissue-resident macrophages in humans may rely more on bone marrow-derived monocytes than in mouse.
Collapse
Affiliation(s)
- Alexandre Gallerand
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jichang Han
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
18
|
Weiss FD, Alvarez Y, Shakeri F, Sahu A, Leka P, Dernst A, Rollheiser J, Vasconcelos M, Geraci A, Duthie F, Stahl R, Lee HE, Gellner AK, Buness A, Latz E, Meissner F. Retention of ES cell-derived 129S genome drives NLRP1 hypersensitivity and transcriptional deregulation in Nlrp3 tm1Flv mice. Cell Death Differ 2024; 31:1717-1729. [PMID: 39289506 PMCID: PMC11618613 DOI: 10.1038/s41418-024-01379-2] [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: 07/23/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024] Open
Abstract
Immune response genes are highly polymorphic in humans and mice, with heterogeneity amongst loci driving strain-specific host defence responses. The inadvertent retention of polymorphic loci can introduce confounding phenotypes, leading to erroneous conclusions, and impeding scientific advancement. In this study, we employ a combination of RNAseq and variant calling analyses to identify a substantial region of 129S genome, including the highly polymorphic Nlrp1 locus, proximal to Nlrp3, in one of the most commonly used mouse models of NLRP3 deficiency (Nlrp3tm1Flv). We show that the presence of the Nlrp1129S locus leads to an increase in NLRP1B protein expression, and a sensitising of Nlrp3tm1Flv macrophages to NLRP1 inflammasome activation, independent of NLRP3 deficiency. Retention of 129S genome further leads to protein sequence differences and altered gene regulation across multiple cell types, including of the key tissue-resident macrophage marker, TIM4. Using alternative models of NLRP3 deficiency, including a previously undescribed conditional Nlrp3 allele enabling precise temporal and cell-type specific control over Nlrp3 deletion, we further show that NLRP3 contributes to Talabostat-driven IL-1β release. Our study also establishes a generic framework to identify functionally relevant SNPs and assess genomic contamination in transgenic mice using RNAseq data. This allows for unambiguous attribution of phenotypes to the target gene and advances the precision and reliability of research in the field of host defence responses.
Collapse
Affiliation(s)
- Felix D Weiss
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany.
| | - Yubell Alvarez
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Farhad Shakeri
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University Hospital Bonn, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Anshupa Sahu
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University Hospital Bonn, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Petro Leka
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
| | - Alesja Dernst
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jessika Rollheiser
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Matilde Vasconcelos
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
| | - Adriana Geraci
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Fraser Duthie
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
| | - Rainer Stahl
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
| | - Hye Eun Lee
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
| | - Anne-Kathrin Gellner
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
- Institute of Physiology II, Medical Faculty, University of Bonn, Bonn, Germany
| | - Andreas Buness
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University Hospital Bonn, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Felix Meissner
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|
19
|
Yuan X, Song X, Zhang X, Hu L, Zhou D, Zhang J, Dai C. Unraveling host-pathogen dynamics in a murine Model of septic peritonitis induced by vancomycin-resistant Enterococcus faecium. Virulence 2024; 15:2367659. [PMID: 38951957 PMCID: PMC11221476 DOI: 10.1080/21505594.2024.2367659] [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: 11/27/2023] [Accepted: 06/09/2024] [Indexed: 07/03/2024] Open
Abstract
Vancomycin-resistant Enterococcus faecium (E. faecium) infection is associated with higher mortality rates. Previous studies have emphasized the importance of innate immune cells and signalling pathways in clearing E. faecium, but a comprehensive analysis of host-pathogen interactions is lacking. Here, we investigated the interplay of host and E. faecium in a murine model of septic peritonitis. Following injection with a sublethal dose, we observed significantly increased murine sepsis score and histological score, decreased weight and bacterial burden, neutrophils and macrophages infiltration, and comprehensive activation of cytokine-mediated signalling pathway. In mice receiving a lethal dose, hypothermia significantly improved survival, reduced bacterial burden, cytokines, and CD86 expression of MHC-II+ recruited macrophages compared to the normothermia group. A mathematical model constructed by observational data from 80 animals, recapitulated the host-pathogen interplay, and further verified the benefits of hypothermia. These findings indicate that E. faecium triggers a severe activation of cytokine-mediated signalling pathway, and hypothermia can improve outcomes by reducing bacterial burden and inflammation.
Collapse
Affiliation(s)
- Xin Yuan
- School of Life Sciences, Ludong University, Yantai, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaolin Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xi Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Lingfei Hu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jianlong Zhang
- School of Life Sciences, Ludong University, Yantai, China
| | - Chenxi Dai
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
20
|
Fattori V, Zaninelli TH, Rasquel-Oliveira FS, Heintz OK, Jain A, Sun L, Seshan ML, Peterse D, Lindholm AE, Anchan RM, Verri WA, Rogers MS. Nociceptor-to-macrophage communication through CGRP/RAMP1 signaling drives endometriosis-associated pain and lesion growth in mice. Sci Transl Med 2024; 16:eadk8230. [PMID: 39504351 DOI: 10.1126/scitranslmed.adk8230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 06/18/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024]
Abstract
Endometriosis is a debilitating and painful gynecological inflammatory disease affecting up to 15% of women and transgender men. Current treatments are ineffective for a substantial proportion of patients, underscoring the need for additional therapies with long-term benefits. Nociceptors release neuropeptides, such as calcitonin gene-related peptide (CGRP), which are known to shape immunity through neuroimmune communication. Given the comorbidity between endometriosis and migraine and the integral role of immune cells and inflammation in endometriosis, we investigated the role of CGRP-mediated neuroimmune communication in endometriosis. Using samples from eight patients with endometriosis and a nonsurgical mouse model of the disease, we found that mouse and human endometriosis lesions contain both CGRP and its coreceptor, receptor activity modifying protein 1 (RAMP1). In mice, nociceptor ablation reduced pain, monocyte recruitment, and lesion size, suggesting that nociceptor activation and neuropeptide release contribute to endometriosis lesion growth and pain. Mechanistically, CGRP changed the phenotype of macrophages to a pro-endometriosis phenotype. CGRP-stimulated macrophages demonstrated impaired efferocytosis and supported increased endometrial cell growth in a RAMP1-dependent manner. Treatment of lesion-bearing mice with US Food and Drug Administration-approved drugs that block CGRP-RAMP1 signaling reduced mechanical hyperalgesia, spontaneous pain, and lesion size. Together, our data demonstrated the effectiveness and underlying cellular mechanisms of nonhormonal and nonopioid CGRP/RAMP1 blockade in a mouse model of endometriosis, suggesting that targeting this axis may lead to clinical benefit for patients with endometriosis.
Collapse
Affiliation(s)
- Victor Fattori
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Tiago H Zaninelli
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology, and General Pathology, Center of Biological Sciences, Londrina State University, Londrina, PR 86057-970, Brazil
| | - Fernanda S Rasquel-Oliveira
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology, and General Pathology, Center of Biological Sciences, Londrina State University, Londrina, PR 86057-970, Brazil
| | - Olivia K Heintz
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Ashish Jain
- Research Computing, Information Technology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Liang Sun
- Research Computing, Information Technology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Maya L Seshan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniëlle Peterse
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Anne E Lindholm
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Raymond M Anchan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology, and General Pathology, Center of Biological Sciences, Londrina State University, Londrina, PR 86057-970, Brazil
| | - Michael S Rogers
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
21
|
Chen YJ, Chang TY, Chen CH. Unraveling the association between chronic inflammatory demyelinating polyradiculoneuropathy and peritoneal Dialysis. BMC Nephrol 2024; 25:383. [PMID: 39468467 PMCID: PMC11514778 DOI: 10.1186/s12882-024-03830-5] [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: 12/26/2023] [Accepted: 10/21/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is a rare disease seen in the general population and has been reported as showing an increased incidence in the peritoneal dialysis (PD) population, as documented in case reports. METHODS We conducted a case-control study using data from the Taichung Veterans General Hospital electric medical record database from the years 2010 to 2023. We defined cases as CIDP with End-stage kidney disease (ESKD) and controls as without CIDP. A logistic regression analysis was used to investigate the association between CIDP and dialysis modality, age, gender, dialysis duration, plasma potassium > 5.5 mEq/L and < 2.5 mEq/L, and intact parathyroid hormone (i-PTH) > 613 pg/mL. RESULTS Our findings suggest that PD may be a risk factor in the ESKD population (Odds ratio: 5.125, C.I.: 1.078 ~ 24.372, p = 0.040) according to logistic regression analysis. Dialysis duration, gender, diabetes mellitus, HbA1c > 7%, hypokalemia, hyperkalemia, and hyperparathyroidism did not show an association with CIDP. CONCLUSION There seems to be an association between PD and CIDP in this case-control study. Possible mechanisms may involve systemic inflammation induced by peritoneal dialysate exchange or the content of the dialysate. Further studies are still needed.
Collapse
Affiliation(s)
- Yu-Jen Chen
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ting-Ya Chang
- Department of Neurology, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Cheng-Hsu Chen
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan.
- Department of Life Science, Tunghai University, Taichung, Taiwan.
| |
Collapse
|
22
|
Gallerand A, Dolfi B, Stunault MI, Caillot Z, Castiglione A, Strazzulla A, Chen C, Heo GS, Luehmann H, Batoul F, Vaillant N, Dumont A, Pilot T, Merlin J, Zair FN, Gilleron J, Bertola A, Carmeliet P, Williams JW, Arguello RJ, Masson D, Dombrowicz D, Yvan-Charvet L, Doyen D, Haschemi A, Liu Y, Guinamard RR, Ivanov S. Glucose metabolism controls monocyte homeostasis and migration but has no impact on atherosclerosis development in mice. Nat Commun 2024; 15:9027. [PMID: 39424804 PMCID: PMC11489573 DOI: 10.1038/s41467-024-53267-5] [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: 12/05/2023] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
Monocytes directly contribute to atherosclerosis development by their recruitment to plaques in which they differentiate into macrophages. In the present study, we ask how modulating monocyte glucose metabolism could affect their homeostasis and their impact on atherosclerosis. Here we investigate how circulating metabolites control monocyte behavior in blood, bone marrow and peripheral tissues of mice. We find that serum glucose concentrations correlate with monocyte numbers. In diet-restricted mice, monocytes fail to metabolically reprogram from glycolysis to fatty acid oxidation, leading to reduced monocyte numbers in the blood. Mechanistically, Glut1-dependent glucose metabolism helps maintain CD115 membrane expression on monocytes and their progenitors, and regulates monocyte migratory capacity by modulating CCR2 expression. Results from genetic models and pharmacological inhibitors further depict the relative contribution of different metabolic pathways to the regulation of CD115 and CCR2 expression. Meanwhile, Glut1 inhibition does not impact atherosclerotic plaque development in mouse models despite dramatically reducing blood monocyte numbers, potentially due to the remaining monocytes having increased migratory capacity. Together, these data emphasize the role of glucose uptake and intracellular glucose metabolism in controlling monocyte homeostasis and functions.
Collapse
Affiliation(s)
- Alexandre Gallerand
- Université Côte d'Azur, CNRS, LP2M, Nice, France.
- Université Côte d'Azur, INSERM, C3M, Nice, France.
| | - Bastien Dolfi
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | | | - Alexia Castiglione
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | - Chuqiao Chen
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Flora Batoul
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | | | | | - Thomas Pilot
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France
| | | | | | | | | | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB, Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, 3000, Belgium
| | - Jesse W Williams
- Center for Immunology, Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Rafael J Arguello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - David Masson
- Université Bourgogne Franche-Comté, LNC UMR1231, F-21000, Dijon, France
| | - David Dombrowicz
- Univ.Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | | | - Denis Doyen
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Médecine Intensive Réanimation, Hôpital Pasteur, CHU de Nice, Nice, France
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rodolphe R Guinamard
- Université Côte d'Azur, CNRS, LP2M, Nice, France
- Université Côte d'Azur, INSERM, C3M, Nice, France
| | - Stoyan Ivanov
- Université Côte d'Azur, CNRS, LP2M, Nice, France.
- Université Côte d'Azur, INSERM, C3M, Nice, France.
| |
Collapse
|
23
|
Wu X, Wang Z, Shern T, Zhang H. Efferocytosis assay to quantify the engulfment and acidification of apoptotic cells by macrophages using flow cytometry. STAR Protoc 2024; 5:103215. [PMID: 39068649 PMCID: PMC11338188 DOI: 10.1016/j.xpro.2024.103215] [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: 05/07/2024] [Revised: 06/06/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Efficient macrophage efferocytosis maintains homeostasis and resolves inflammation. Here, we provide a protocol to assess the engulfment and acidification of apoptotic cells (ACs) by macrophages. We describe steps for preparing bone marrow-derived macrophages (BMDMs) and peritoneal macrophages (PMs), fluorescent labeling of ACs using both a pH-sensitive dye, pHrodo-Red succinimidyl ester, and a pH-insensitive dye, Hoechst, and subsequent incubation with macrophages for efferocytosis. We then detail procedures for flow cytometry-based quantification of engulfment and acidification. For complete details on the use and execution of this protocol, please refer to Shi and Wu et al.1.
Collapse
Affiliation(s)
- Xun Wu
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Ziyi Wang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Tyler Shern
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Hanrui Zhang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
24
|
Henlon Y, Panir K, McIntyre I, Hogg C, Dhami P, Cuff AO, Senior A, Moolchandani-Adwani N, Courtois ET, Horne AW, Rosser M, Ott S, Greaves E. Single-cell analysis identifies distinct macrophage phenotypes associated with prodisease and proresolving functions in the endometriotic niche. Proc Natl Acad Sci U S A 2024; 121:e2405474121. [PMID: 39255000 PMCID: PMC11420174 DOI: 10.1073/pnas.2405474121] [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: 03/15/2024] [Accepted: 07/24/2024] [Indexed: 09/11/2024] Open
Abstract
Endometriosis negatively impacts the health-related quality of life of 190 million women worldwide. Novel advances in nonhormonal treatments for this debilitating condition are desperately needed. Macrophages play a vital role in the pathophysiology of endometriosis and represent a promising therapeutic target. In the current study, we revealed the full transcriptomic complexity of endometriosis-associated macrophage subpopulations using single-cell analyses in a preclinical mouse model of experimental endometriosis. We have identified two key lesion-resident populations that resemble i) tumor-associated macrophages (characterized by expression of Folr2, Mrc1, Gas6, and Ccl8+) that promoted expression of Col1a1 and Tgfb1 in human endometrial stromal cells and increased angiogenic meshes in human umbilical vein endothelial cells, and ii) scar-associated macrophages (Mmp12, Cd9, Spp1, Trem2+) that exhibited a phenotype associated with fibrosis and matrix remodeling. We also described a population of proresolving large peritoneal macrophages that align with a lipid-associated macrophage phenotype (Apoe, Saa3, Pid1) concomitant with altered lipid metabolism and cholesterol efflux. Gain of function experiments using an Apoe mimetic resulted in decreased lesion size and fibrosis, and modification of peritoneal macrophage populations in the preclinical model. Using cross-species analysis of mouse and human single-cell datasets, we determined the concordance of peritoneal and lesion-resident macrophage subpopulations, identifying key similarities and differences in transcriptomic phenotypes. Ultimately, we envisage that these findings will inform the design and use of specific macrophage-targeted therapies and open broad avenues for the treatment of endometriosis.
Collapse
Affiliation(s)
- Yasmin Henlon
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Kavita Panir
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Iona McIntyre
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Chloe Hogg
- Centre for Reproductive Health, Institute of Regeneration and Repair, The University of Edinburgh, EdinburghEH16 4UU, United Kingdom
| | - Priya Dhami
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Antonia O. Cuff
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Anna Senior
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Niky Moolchandani-Adwani
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Elise T. Courtois
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT06032
| | - Andrew W. Horne
- Centre for Reproductive Health, Institute of Regeneration and Repair, The University of Edinburgh, EdinburghEH16 4UU, United Kingdom
| | - Matthew Rosser
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Sascha Ott
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Erin Greaves
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CoventryCV4 7AL, United Kingdom
- Centre for Early Life, University of Warwick, CoventryCV4 7AL, United Kingdom
| |
Collapse
|
25
|
Zhang WB, Chen ZX, Liu Z, Qian XY, Ge YZ, Zhang HY, Xu WT, Shan LT, Zhao DB. PBMC-mediated modulation of macrophage polarization in RAW264.7 cells through STAT1/STAT6 signaling cascades. Int Immunopharmacol 2024; 138:112651. [PMID: 38986303 DOI: 10.1016/j.intimp.2024.112651] [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: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
Peripheral blood mononuclear cells (PBMC), sourced autologously, offer numerous advantages when procured: easier acquisition process, no in vitro amplification needed, decreased intervention and overall increased acceptability make PBMC an attractive candidate for cell therapy treatment. However, the exact mechanism by which PBMC treat diseases remains poorly understood. Immune imbalance is the pathological basis of many diseases, with macrophages playing a crucial role in this process. However, research on the role and mechanisms of PBMC in regulating macrophages remains scarce. This study employed an in vitro co-culture model of PBMC and RAW264.7 macrophages to explore the role and mechanisms of PBMC in regulating macrophages. The results showed that the co-culturing led to decreased expression of inflammatory cytokines and increased expression of anti-inflammatory cytokines in RAW264.7 or in the culture supernatant. Additionally, the pro-inflammatory, tissue matrix-degrading M1 macrophages decreased, while the anti-inflammatory, matrix-synthesizing, regenerative M2 macrophages increased in both RAW264.7 and monocytes within PBMC. Moreover, co-cultured macrophages exhibited a significantly decreased p-STAT1/STAT1 ratio, while the p-STAT6/STAT6 ratio significantly increased. This suggests that PBMC may inhibit M1 macrophage polarization by blocking STAT1 signaling cascades and may promote M2 macrophage polarization through the activation of STAT6 signaling cascades. Overall, this study sheds light on the role and mechanism of PBMC in regulating macrophages. Moreover, it was found that monocytes within co-cultured PBMC differentiated into M2 macrophages in the presence of macrophages. This finding provides experimental evidence for the use of PBMC in treating inflammatory diseases, especially macrophage-depleting inflammatory diseases such as osteoarthritis.
Collapse
Affiliation(s)
- Wen-Bo Zhang
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zu-Xiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Zhen Liu
- Department of Anatomy, Naval Medical University, Shanghai 200433, China
| | - Xin-Yu Qian
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan-Zhi Ge
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Hai-Yan Zhang
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Wen-Ting Xu
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Le-Tian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| | - Dong-Bao Zhao
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
26
|
Li X, Liu Y, Zou Y, Zhang J, Wang Y, Ding Y, Shi Z, Guo X, Zhang S, Yin H, Guo A, Wang S. Echinococcus multilocularis serpin regulates macrophage polarization and reduces gut dysbiosis in colitis. Infect Immun 2024; 92:e0023224. [PMID: 39037247 PMCID: PMC11320943 DOI: 10.1128/iai.00232-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024] Open
Abstract
Helminths serve as principal regulators in modulating host immune responses, and their excretory-secretory proteins are recognized as potential therapeutic agents for inflammatory bowel disease. Nevertheless, our comprehension of the mechanisms underlying immunoregulation remains restricted. This investigation delves into the immunomodulatory role of a secretory protein serpin (Emu-serpin), within the larval stage of Echinococcus multilocularis. Our observations indicate that Emu-serpin effectively alleviates dextran sulfate sodium-induced colitis, yielding a substantial reduction in immunopathology and an augmentation of anti-inflammatory cytokines. Furthermore, this suppressive regulatory effect is concomitant with the reduction of gut microbiota dysbiosis linked to colitis, as evidenced by a marked impediment to the expansion of the pathobiont taxa Enterobacteriaceae. In vivo experiments demonstrate that Emu-serpin facilitates the expansion of M2 phenotype macrophages while concurrently diminishing M1 phenotype macrophages, alongside an elevation in anti-inflammatory cytokine levels. Subsequent in vitro investigations involving RAW264.7 and bone marrow macrophages reveal that Emu-serpin induces a conversion of M2 macrophage populations from a pro-inflammatory to an anti-inflammatory phenotype through direct inhibition. Adoptive transfer experiments reveal the peritoneal macrophages induced by Emu-serpin alleviate colitis and gut microbiota dysbiosis. In summary, these findings propose that Emu-serpin holds the potential to regulate macrophage polarization and maintain gut microbiota homeostasis in colitis, establishing it as a promising candidate for developing helminth therapy for preventing inflammatory diseases.
Collapse
Affiliation(s)
- Xiaolu Li
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
| | - Yihui Liu
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
| | - Yang Zou
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
| | - Jiayun Zhang
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
| | - Yugui Wang
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
| | - Yingying Ding
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
| | - Zhiqi Shi
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
| | - Xiaola Guo
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
| | - Shaohua Zhang
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
| | - Hong Yin
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| | - Aijiang Guo
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| | - Shuai Wang
- State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou, Gansu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
| |
Collapse
|
27
|
Shi M, MacLean JA, Hayashi K. The involvement of peritoneal GATA6 + macrophages in the pathogenesis of endometriosis. Front Immunol 2024; 15:1396000. [PMID: 39192982 PMCID: PMC11348394 DOI: 10.3389/fimmu.2024.1396000] [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: 03/05/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Endometriosis is a chronic inflammatory disease that causes debilitating pelvic pain in women. Macrophages are considered to be key players in promoting disease progression, as abundant macrophages are present in ectopic lesions and elevated in the peritoneum. In the present study, we examined the role of GATA6+ peritoneal macrophages on endometriosis-associated hyperalgesia using mice with a specific myeloid deficiency of GATA6. Lesion induction induced the disappearance of TIM4hi MHCIIlo residential macrophages and the influx of increased Ly6C+ monocytes and TIM4lo MHCIIhi macrophages. The recruitment of MHCIIhi inflammatory macrophages was extensive in Mac Gata6 KO mice due to the severe disappearance of TIM4hi MHCIIlo residential macrophages. Ki67 expression confirmed GATA6-dependent proliferative ability, showing different proliferative phenotypes of TIM4+ residential macrophages in Gata6f/f and Mac Gata6 KO mice. Peritoneal proinflammatory cytokines were elevated after lesion induction. When cytokine levels were compared between Gata6f/f and Mac Gata6 KO mice, TNFα at day 21 in Gata6f/f mice was higher than in Mac Gata6 KO mice. Lesion induction increased both abdominal and hind paw sensitivities. Gata6f/f mice tended to show higher sensitivity in the abdomen after day 21. Elevated expression of TRPV1 and CGRP was observed in the dorsal root ganglia after ELL induction in Gata6f/f mice until days 21 and 42, respectively. These results support that peritoneal GATA6+ macrophages are involved in the recruitment and reprogramming of monocyte-derived macrophages. The extensive recruitment of monocyte-derived macrophages in Mac Gata6 KO mice might protect against inflammatory stimuli during the resolution phase, whereas GATA6 deficiency did not affect lesion initiation and establishment at the acute phase of inflammation. GATA6+ residential macrophages act to sustain local inflammation in the peritoneum and sensitivities in the neurons, reflecting endometriosis-associated hyperalgesia.
Collapse
Affiliation(s)
| | | | - Kanako Hayashi
- School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, Pullman, WA, United States
| |
Collapse
|
28
|
Yip JLK, Balasuriya GK, Hill-Yardin EL, Spencer SJ. The gut-brain and gut-macrophage contribution to gastrointestinal dysfunction with systemic inflammation. Brain Behav Immun 2024; 119:867-877. [PMID: 38750700 DOI: 10.1016/j.bbi.2024.05.017] [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: 01/08/2024] [Revised: 04/24/2024] [Accepted: 05/12/2024] [Indexed: 05/19/2024] Open
Abstract
The gastrointestinal tract is one of the main organs affected during systemic inflammation and disrupted gastrointestinal motility is a major clinical manifestation. Many studies have investigated the involvement of neuroimmune interactions in regulating colonic motility during localized colonic inflammation, i.e., colitis. However, little is known about how the enteric nervous system and intestinal macrophages contribute to dysregulated motility during systemic inflammation. Given that systemic inflammation commonly results from the innate immune response against bacterial infection, we mimicked bacterial infection by administering lipopolysaccharide (LPS) to rats and assessed colonic motility using ex vivo video imaging techniques. We utilized the Cx3cr1-Dtr rat model of transient depletion of macrophages to investigate the role of intestinal macrophages in regulating colonic motility during LPS infection. To investigate the role of inhibitory enteric neurotransmission on colonic motility following LPS, we applied the nitric oxide synthase inhibitor, Nω-nitro-L-arginine (NOLA). Our results confirmed an increase in colonic contraction frequency during LPS-induced systemic inflammation. However, neither the depletion of intestinal macrophages, nor the suppression of inhibitory enteric nervous system activity impacted colonic motility disruption during inflammation. This implies that the interplay between the enteric nervous system and intestinal macrophages is nuanced, and complex, and further investigation is needed to clarify their joint roles in colonic motility.
Collapse
Affiliation(s)
- Jackson L K Yip
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
| | - Gayathri K Balasuriya
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia; Department of Physiology and Cell Biology, Kobe University School of Medicine, Kobe, Japan
| | - Elisa L Hill-Yardin
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC, Australia.
| |
Collapse
|
29
|
Kim JE, Han D, Kim KH, Seo A, Moon JJ, Jeong JS, Kim JH, Kang E, Bae E, Kim YC, Lee JW, Cha RH, Kim DK, Oh KH, Kim YS, Jung HY, Yang SH. Protective effect of Cyclo(His-Pro) on peritoneal fibrosis through regulation of HDAC3 expression. FASEB J 2024; 38:e23819. [PMID: 38984942 DOI: 10.1096/fj.202400854r] [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: 04/15/2024] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Peritoneal dialysis is a common treatment for end-stage renal disease, but complications often force its discontinuation. Preventive treatments for peritoneal inflammation and fibrosis are currently lacking. Cyclo(His-Pro) (CHP), a naturally occurring cyclic dipeptide, has demonstrated protective effects in various fibrotic diseases, yet its potential role in peritoneal fibrosis (PF) remains uncertain. In a mouse model of induced PF, CHP was administered, and quantitative proteomic analysis using liquid chromatography-tandem mass spectrometry was employed to identify PF-related protein signaling pathways. The results were further validated using human primary cultured mesothelial cells. This analysis revealed the involvement of histone deacetylase 3 (HDAC3) in the PF signaling pathway. CHP administration effectively mitigated PF in both peritoneal tissue and human primary cultured mesothelial cells, concurrently regulating fibrosis-related markers and HDAC3 expression. Moreover, CHP enhanced the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) while suppressing forkhead box protein M1 (FOXM1), known to inhibit Nrf2 transcription through its interaction with HDAC3. CHP also displayed an impact on spleen myeloid-derived suppressor cells, suggesting an immunomodulatory effect. Notably, CHP improved mitochondrial function in peritoneal tissue, resulting in increased mitochondrial membrane potential and adenosine triphosphate production. This study suggests that CHP can significantly prevent PF in peritoneal dialysis patients by modulating HDAC3 expression and associated signaling pathways, reducing fibrosis and inflammation markers, and improving mitochondrial function.
Collapse
Affiliation(s)
- Ji Eun Kim
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Dohyun Han
- Proteomics Core Facility, Seoul National University Hospital, Seoul, Korea
| | - Kyu Hong Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Areum Seo
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Jong Joo Moon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jin Seon Jeong
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - Ji Hye Kim
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Eunjeong Kang
- Transplantation Center, Seoul National University Hospital, Seoul, Korea
| | - Eunjin Bae
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Changwon, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Wook Lee
- Nephrology Clinic, National Cancer Center, Goyang, Korea
| | - Ran-Hui Cha
- Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hoe-Yune Jung
- R&D Center, NovMetaPharma Co., Ltd, Pohang, Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Seung Hee Yang
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Tighe RM, Birukova A, Malakhau Y, Kobayashi Y, Vose AT, Chandramohan V, Cyphert-Daly JM, Cumming RI, Fradin Kirshner H, Tata PR, Ingram JL, Gunn MD, Que LG, Yu YRA. Altered ontogeny and transcriptomic signatures of tissue-resident pulmonary interstitial macrophages ameliorate allergic airway hyperresponsiveness. Front Immunol 2024; 15:1371764. [PMID: 38983858 PMCID: PMC11231371 DOI: 10.3389/fimmu.2024.1371764] [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: 01/16/2024] [Accepted: 06/05/2024] [Indexed: 07/11/2024] Open
Abstract
Introduction Environmental exposures and experimental manipulations can alter the ontogenetic composition of tissue-resident macrophages. However, the impact of these alterations on subsequent immune responses, particularly in allergic airway diseases, remains poorly understood. This study aims to elucidate the significance of modified macrophage ontogeny resulting from environmental exposures on allergic airway responses to house dust mite (HDM) allergen. Methods We utilized embryonic lineage labeling to delineate the ontogenetic profile of tissue-resident macrophages at baseline and following the resolution of repeated lipopolysaccharide (LPS)-induced lung injury. We investigated differences in house dust mite (HDM)-induced allergy to assess the influence of macrophage ontogeny on allergic airway responses. Additionally, we employed single-cell RNA sequencing (scRNAseq) and immunofluorescent staining to characterize the pulmonary macrophage composition, associated pathways, and tissue localization. Results Our findings demonstrate that the ontogeny of homeostatic alveolar and interstitial macrophages is altered after the resolution from repeated LPS-induced lung injury, leading to the replacement of embryonic-derived by bone marrow-derived macrophages. This shift in macrophage ontogeny is associated with reduced HDM-induced allergic airway responses. Through scRNAseq and immunofluorescent staining, we identified a distinct subset of resident-derived interstitial macrophages expressing genes associated with allergic airway diseases, localized adjacent to terminal bronchi, and diminished by prior LPS exposure. Discussion These results suggest a pivotal role for pulmonary macrophage ontogeny in modulating allergic airway responses. Moreover, our findings highlight the implications of prior environmental exposures in shaping future immune responses and influencing the development of allergies. By elucidating the mechanisms underlying these phenomena, this study provides valuable insights into potential therapeutic targets for allergic airway diseases and avenues for further research into immune modulation and allergic disease prevention.
Collapse
Affiliation(s)
- Robert M. Tighe
- Department of Medicine, Duke University, Durham, NC, United States
| | | | - Yuryi Malakhau
- Department of Medicine, Duke University, Durham, NC, United States
| | - Yoshihiko Kobayashi
- Department of Cell Biology, Duke University, Durham, NC, United States
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Aaron T. Vose
- Department of Medicine, Duke University, Durham, NC, United States
| | | | | | - R. Ian Cumming
- Department of Medicine, Duke University, Durham, NC, United States
| | | | | | | | - Michael D. Gunn
- Department of Medicine, Duke University, Durham, NC, United States
| | - Loretta G. Que
- Department of Medicine, Duke University, Durham, NC, United States
| | - Yen-Rei A. Yu
- Department of Medicine, Duke University, Durham, NC, United States
- Department of Medicine, University of Colorado Anschutz School of Medicine, Aurora, CO, United States
| |
Collapse
|
31
|
Purvis GSD, McNeill E, Wright B, Channon KM, Greaves DR. Ly6C hi Monocytes Are Metabolically Reprogrammed in the Blood during Inflammatory Stimulation and Require Intact OxPhos for Chemotaxis and Monocyte to Macrophage Differentiation. Cells 2024; 13:916. [PMID: 38891050 PMCID: PMC11171939 DOI: 10.3390/cells13110916] [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: 04/29/2024] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Acute inflammation is a rapid and dynamic process involving the recruitment and activation of multiple cell types in a coordinated and precise manner. Here, we investigate the origin and transcriptional reprogramming of monocytes using a model of acute inflammation, zymosan-induced peritonitis. Monocyte trafficking and adoptive transfer experiments confirmed that monocytes undergo rapid phenotypic change as they exit the blood and give rise to monocyte-derived macrophages that persist during the resolution of inflammation. Single-cell transcriptomics revealed significant heterogeneity within the surface marker-defined CD11b+Ly6G-Ly6Chi monocyte populations within the blood and at the site of inflammation. We show that two major transcriptional reprogramming events occur during the initial six hours of Ly6Chi monocyte mobilisation, one in the blood priming monocytes for migration and a second at the site of inflammation. Pathway analysis revealed an important role for oxidative phosphorylation (OxPhos) during both these reprogramming events. Experimentally, we demonstrate that OxPhos via the intact mitochondrial electron transport chain is essential for murine and human monocyte chemotaxis. Moreover, OxPhos is needed for monocyte-to-macrophage differentiation and macrophage M(IL-4) polarisation. These new findings from transcriptional profiling open up the possibility that shifting monocyte metabolic capacity towards OxPhos could facilitate enhanced macrophage M2-like polarisation to aid inflammation resolution and tissue repair.
Collapse
Affiliation(s)
- Gareth S. D. Purvis
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK;
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (E.M.); (B.W.); (K.M.C.)
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Eileen McNeill
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (E.M.); (B.W.); (K.M.C.)
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Benjamin Wright
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (E.M.); (B.W.); (K.M.C.)
| | - Keith M. Channon
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (E.M.); (B.W.); (K.M.C.)
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK
| | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK;
- British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
32
|
Ryu S, Lee EK. The Pivotal Role of Macrophages in the Pathogenesis of Pancreatic Diseases. Int J Mol Sci 2024; 25:5765. [PMID: 38891952 PMCID: PMC11171839 DOI: 10.3390/ijms25115765] [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: 04/25/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
The pancreas is an organ with both exocrine and endocrine functions, comprising a highly organized and complex tissue microenvironment composed of diverse cellular and non-cellular components. The impairment of microenvironmental homeostasis, mediated by the dysregulation of cell-to-cell crosstalk, can lead to pancreatic diseases such as pancreatitis, diabetes, and pancreatic cancer. Macrophages, key immune effector cells, can dynamically modulate their polarization status between pro-inflammatory (M1) and anti-inflammatory (M2) modes, critically influencing the homeostasis of the pancreatic microenvironment and thus playing a pivotal role in the pathogenesis of the pancreatic disease. This review aims to summarize current findings and provide detailed mechanistic insights into how alterations mediated by macrophage polarization contribute to the pathogenesis of pancreatic disorders. By analyzing current research comprehensively, this article endeavors to deepen our mechanistic understanding of regulatory molecules that affect macrophage polarity and the intricate crosstalk that regulates pancreatic function within the microenvironment, thereby facilitating the development of innovative therapeutic strategies that target perturbations in the pancreatic microenvironment.
Collapse
Affiliation(s)
- Seungyeon Ryu
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eun Kyung Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
33
|
Grezzi L, González C, Díaz Á, Casaravilla C. The Acute Inflammatory Potential of Particles From the Echinococcus granulosus Laminated Layer Is Moderated by Its Calcium Inositol Hexakisphosphate Component. Parasite Immunol 2024; 46:e13040. [PMID: 38801355 DOI: 10.1111/pim.13040] [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: 12/15/2023] [Revised: 04/11/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024]
Abstract
Cystic echinococcosis is caused by the tissue-dwelling larva (hydatid) of Echinococcus granulosus sensu lato. A salient feature is that this larva is protected by the acellular laminated layer (LL). As the parasite grows, the LL sheds abundant particles that can accumulate in the parasite's vicinity. The potential of LL particles to induce inflammation in vivo has not been specifically analysed. It is not known how each of its two major components, namely highly glycosylated mucins and calcium inositol hexakisphosphate (InsP6) deposits, impacts inflammation induced by the LL as a whole. In this work, we show that LL particles injected intraperitoneally cause infiltration of eosinophils, neutrophils and monocytes/macrophages as well as the disappearance of resident (large peritoneal) macrophages. Strikingly, the absence of calcium InsP6 enhanced the recruitment of all the inflammatory cell types analysed. In contrast, oxidation of the mucin carbohydrates caused decreased recruitment of neutrophils. The carbohydrate-oxidised particles caused cell influx nonetheless, which may be explained by possible receptor-independent effects of LL particles on innate immune cells, as suggested by previous works from our group. In summary, LL particles can induce acute inflammatory cell recruitment partly dependent on its mucin glycans, and this recruitment is attenuated by the calcium InsP6 component.
Collapse
Affiliation(s)
- Leticia Grezzi
- Área Inmunología, Departamento de Biociencias (Facultad de Química), Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Inmunología, Instituto de Química Biológica (Facultad de Ciencias), Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | | | - Álvaro Díaz
- Área Inmunología, Departamento de Biociencias (Facultad de Química), Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Inmunología, Instituto de Química Biológica (Facultad de Ciencias), Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | - Cecilia Casaravilla
- Área Inmunología, Departamento de Biociencias (Facultad de Química), Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Inmunología, Instituto de Química Biológica (Facultad de Ciencias), Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
34
|
Joshi S, López L, Morosi LG, Amadio R, Pachauri M, Bestagno M, Ogar IP, Giacca M, Piperno GM, Vorselen D, Benvenuti F. Tim4 enables large peritoneal macrophages to cross-present tumor antigens at early stages of tumorigenesis. Cell Rep 2024; 43:114096. [PMID: 38607919 DOI: 10.1016/j.celrep.2024.114096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/20/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Receptors controlling the cross-presentation of tumor antigens by macrophage subsets in cancer tissues are poorly explored. Here, we show that TIM4+ large peritoneal macrophages efficiently capture and cross-present tumor-associated antigens at early stages of peritoneal infiltration by ovarian cancer cells. The phosphatidylserine (PS) receptor TIM4 promotes maximal uptake of dead cells or PS-coated artificial targets and triggers inflammatory and metabolic gene programs in combination with cytoskeletal remodeling and upregulation of transcriptional signatures related to antigen processing. At the cellular level, TIM4-mediated engulfment induces nucleation of F-actin around nascent phagosomes, delaying the recruitment of vacuolar ATPase, acidification, and cargo degradation. In vivo, TIM4 deletion blunts induction of early anti-tumoral effector CD8 T cells and accelerates the progression of ovarian tumors. We conclude that TIM4-mediated uptake drives the formation of specialized phagosomes that prolong the integrity of ingested antigens and facilitate cross-presentation, contributing to immune surveillance of the peritoneum.
Collapse
Affiliation(s)
- Sonal Joshi
- Cellular Immunology, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Lucía López
- Cellular Immunology, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Luciano Gastón Morosi
- Cellular Immunology, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Roberto Amadio
- Cellular Immunology, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Manendra Pachauri
- Department of Medical, Surgical, and Health Sciences, University of Trieste and International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Marco Bestagno
- Cellular Immunology, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Ironya Paul Ogar
- Cellular Immunology, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy; Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, University of Calabar, P.M.B. 1115 Calabar, Cross River State, Nigeria
| | - Mauro Giacca
- Department of Medical, Surgical, and Health Sciences, University of Trieste and International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy; King's College London, British Heart Foundation Center of Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Giulia Maria Piperno
- Cellular Immunology, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Daan Vorselen
- Department of Cell Biology & Immunology, Wageningen University & Research, 6708 PD Wageningen, the Netherlands
| | - Federica Benvenuti
- Cellular Immunology, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.
| |
Collapse
|
35
|
Castellano M, Blanco V, Calzi ML, Costa B, Witwer K, Hill M, Cayota A, Segovia M, Tosar JP. Ribonuclease activity undermines immune sensing of naked extracellular RNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590771. [PMID: 38712104 PMCID: PMC11071435 DOI: 10.1101/2024.04.23.590771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The plasma membrane and the membrane of endosomal vesicles are considered physical barriers preventing extracellular RNA uptake. While naked RNA can be spontaneously internalized by certain cells types, functional delivery of naked RNA into the cytosol has been rarely observed. Here we show that extracellular ribonucleases, mainly derived from cell culture supplements, have so far hindered the study of extracellular RNA functionality. In the presence of active ribonuclease inhibitors (RI), naked bacterial RNA is pro-inflammatory when spiked in the media of dendritic cells and macrophages. In murine cells, this response mainly depends on the action of endosomal Toll-like receptors. However, we also show that naked RNA can perform endosomal escape and engage with cytosolic RNA sensors and ribosomes. For example, naked mRNAs encoding reporter proteins can be spontaneously internalized and translated by a variety of cell types, in an RI-dependent manner. In vivo, RI co-injection enhances the activation induced by naked extracellular RNA on splenic lymphocytes and myeloid-derived leukocytes. Furthermore, naked extracellular RNA is inherently pro-inflammatory in ribonuclease-poor compartments such as the peritoneal cavity. Overall, these results demonstrate that naked RNA is bioactive and does not need encapsulation inside synthetic or biological lipid vesicles for functional uptake, making a case for nonvesicular extracellular RNA-mediated intercellular communication.
Collapse
Affiliation(s)
- Mauricio Castellano
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Immunoregulation and Inflammation Laboratory, Institut Pasteur Montevideo, Uruguay
| | - Valentina Blanco
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
| | - Marco Li Calzi
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
| | - Bruno Costa
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Analytical Biochemistry Unit, School of Science, Universidad de la República, Uruguay
| | - Kenneth Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- EV Core Facility “EXCEL”, Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcelo Hill
- Immunoregulation and Inflammation Laboratory, Institut Pasteur Montevideo, Uruguay
- Academic Unit of Immunobiology, School of Medicine, Universidad de la República, Uruguay
| | - Alfonso Cayota
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Hospital de Clínicas, Universidad de la República, Uruguay
| | - Mercedes Segovia
- Immunoregulation and Inflammation Laboratory, Institut Pasteur Montevideo, Uruguay
- Academic Unit of Immunobiology, School of Medicine, Universidad de la República, Uruguay
| | - Juan Pablo Tosar
- Functional Genomics Laboratory, Institut Pasteur Montevideo, Uruguay
- Analytical Biochemistry Unit, School of Science, Universidad de la República, Uruguay
| |
Collapse
|
36
|
Hopkins JW, Sulka KB, Sawden M, Carroll KA, Brown RD, Bunnell SC, Poltorak A, Tai A, Reed ER, Sharma S. STING promotes homeostatic maintenance of tissues and confers longevity with aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588107. [PMID: 38645182 PMCID: PMC11030237 DOI: 10.1101/2024.04.04.588107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Local immune processes within aging tissues are a significant driver of aging associated dysfunction, but tissue-autonomous pathways and cell types that modulate these responses remain poorly characterized. The cytosolic DNA sensing pathway, acting through cyclic GMP-AMP synthase (cGAS) and Stimulator of Interferon Genes (STING), is broadly expressed in tissues, and is poised to regulate local type I interferon (IFN-I)-dependent and independent inflammatory processes within tissues. Recent studies suggest that the cGAS/STING pathway may drive pathology in various in vitro and in vivo models of accelerated aging. To date, however, the role of the cGAS/STING pathway in physiological aging processes, in the absence of genetic drivers, has remained unexplored. This remains a relevant gap, as STING is ubiquitously expressed, implicated in multitudinous disorders, and loss of function polymorphisms of STING are highly prevalent in the human population (>50%). Here we reveal that, during physiological aging, STING-deficiency leads to a significant shortening of murine lifespan, increased pro-inflammatory serum cytokines and tissue infiltrates, as well as salient changes in histological composition and organization. We note that aging hearts, livers, and kidneys express distinct subsets of inflammatory, interferon-stimulated gene (ISG), and senescence genes, collectively comprising an immune fingerprint for each tissue. These distinctive patterns are largely imprinted by tissue-specific stromal and myeloid cells. Using cellular interaction network analyses, immunofluorescence, and histopathology data, we show that these immune fingerprints shape the tissue architecture and the landscape of cell-cell interactions in aging tissues. These age-associated immune fingerprints are grossly dysregulated with STING-deficiency, with key genes that define aging STING-sufficient tissues greatly diminished in the absence of STING. Changes in immune signatures are concomitant with a restructuring of the stromal and myeloid fractions, whereby cell:cell interactions are grossly altered and resulting in disorganization of tissue architecture in STING-deficient organs. This altered homeostasis in aging STING-deficient tissues is associated with a cross-tissue loss of homeostatic tissue-resident macrophage (TRM) populations in these tissues. Ex vivo analyses reveal that basal STING-signaling limits the susceptibility of TRMs to death-inducing stimuli and determines their in situ localization in tissue niches, thereby promoting tissue homeostasis. Collectively, these data upend the paradigm that cGAS/STING signaling is primarily pathological in aging and instead indicate that basal STING signaling sustains tissue function and supports organismal longevity. Critically, our study urges caution in the indiscriminate targeting of these pathways, which may result in unpredictable and pathological consequences for health during aging.
Collapse
Affiliation(s)
- Jacob W. Hopkins
- Department of Immunology, Tufts University, Boston, MA 02111
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111
| | - Katherine B. Sulka
- Department of Immunology, Tufts University, Boston, MA 02111
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111
| | - Machlan Sawden
- Department of Immunology, Tufts University, Boston, MA 02111
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111
| | - Kimberly A. Carroll
- Department of Immunology, Tufts University, Boston, MA 02111
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111
| | - Ronald D. Brown
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, 12853
| | | | | | - Albert Tai
- Department of Immunology, Tufts University, Boston, MA 02111
- Data Intensive Studies Center, Tufts University, Medford, MA, 02155
| | - Eric R. Reed
- Data Intensive Studies Center, Tufts University, Medford, MA, 02155
| | - Shruti Sharma
- Department of Immunology, Tufts University, Boston, MA 02111
| |
Collapse
|
37
|
McBride MA, Stothers CL, Fensterheim BA, Caja KR, Owen AM, Hernandez A, Bohannon JK, Patil NK, Ali S, Dalal S, Rahim M, Trenary IA, Young JD, Williams DL, Sherwood ER. Bacteria- and fungus-derived PAMPs induce innate immune memory via similar functional, metabolic, and transcriptional adaptations. J Leukoc Biol 2024; 115:358-373. [PMID: 37793181 PMCID: PMC10872320 DOI: 10.1093/jleuko/qiad120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/28/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023] Open
Abstract
Exposure to pathogen-associated molecular patterns (PAMPs) induces an augmented, broad-spectrum antimicrobial response to subsequent infection, a phenomenon termed innate immune memory. This study examined the effects of treatment with β-glucan, a fungus-derived dectin-1 ligand, or monophosphoryl lipid A (MPLA), a bacteria-derived Toll-like receptor 4 ligand, on innate immune memory with a focus on identifying common cellular and molecular pathways activated by these diverse PAMPs. Treatment with either PAMP prepared the innate immune system to respond more robustly to Pseudomonas aeruginosa infection in vivo by facilitating mobilization of innate leukocytes into blood, recruitment of leukocytes to the site of infection, augmentation of microbial clearance, and attenuation of cytokine production. Examination of macrophages ex vivo showed amplification of metabolism, phagocytosis, and respiratory burst after treatment with either agent, although MPLA more robustly augmented these activities and more effectively facilitated killing of bacteria. Both agents activated gene expression pathways in macrophages that control inflammation, antimicrobial functions, and protein synthesis and suppressed pathways regulating cell division. β-glucan treatment minimally altered macrophage differential gene expression in response to lipopolysaccharide (LPS) challenge, whereas MPLA attenuated the magnitude of the LPS-induced transcriptional response, especially cytokine gene expression. These results show that β-glucan and MPLA similarly augment the innate response to infection in vivo. Yet, MPLA more potently induces alterations in macrophage metabolism, antimicrobial functions, gene transcription and the response to LPS.
Collapse
Affiliation(s)
- Margaret A. McBride
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Cody L. Stothers
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Benjamin A. Fensterheim
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Katherine R. Caja
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Allison M. Owen
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Julia K. Bohannon
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Naeem K. Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Sabah Ali
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Sujata Dalal
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
| | - Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville 37235, Tennessee
| | - Irina A. Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville 37235, Tennessee
| | - Jamey D. Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, 2301 Vanderbilt Place, Nashville 37235, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2215 Garland Avenue, Nashville 37232, Tennessee
| | - David L. Williams
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, 325 North State of Franklin Road, Johnson City 37604, Tennessee
- Center for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, 325 North State of Franklin Road, Johnson City 37604, Tennessee
| | - Edward R. Sherwood
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville 37232, Tennessee
- Department of Surgery, Quillen College of Medicine, East Tennessee State University, 325 North State of Franklin Road, Johnson City 37604, Tennessee
- Center for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, 325 North State of Franklin Road, Johnson City 37604, Tennessee
| |
Collapse
|
38
|
Majumder N, Roy S, Sharma A, Arora S, Vaishya R, Bandyopadhyay A, Ghosh S. Assessing the advantages of 3D bioprinting and 3D spheroids in deciphering the osteoarthritis healing mechanism using human chondrocytes and polarized macrophages. Biomed Mater 2024; 19:025005. [PMID: 38198731 DOI: 10.1088/1748-605x/ad1d18] [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: 07/03/2023] [Accepted: 01/10/2024] [Indexed: 01/12/2024]
Abstract
The molecular niche of an osteoarthritic microenvironment comprises the native chondrocytes, the circulatory immune cells, and their respective inflammatory mediators. Although M2 macrophages infiltrate the joint tissue during osteoarthritis (OA) to initiate cartilage repair, the mechanistic crosstalk that dwells underneath is still unknown. Our study established a co-culture system of human OA chondrocytes and M2 macrophages in 3D spheroids and 3D bioprinted silk-gelatin constructs. It is already well established that Silk fibroin-gelatin bioink supports chondrogenic differentiation due to upregulation of the Wnt/β-catenin pathway. Additionally, the presence of anti-inflammatory M2 macrophages significantly upregulated the expression of chondrogenic biomarkers (COL-II, ACAN) with an attenuated expression of the chondrocyte hypertrophy (COL-X), chondrocyte dedifferentiation (COL-I) and matrix catabolism (MMP-1 and MMP-13) genes even in the absence of the interleukins. Furthermore, the 3D bioprinted co-culture model displayed an upper hand in stimulating cartilage regeneration and OA inhibition than the spheroid model, underlining the role of silk fibroin-gelatin in encouraging chondrogenesis. Additionally, the 3D bioprinted silk-gelatin constructs further supported the maintenance of stable anti-inflammatory phenotype of M2 macrophage. Thus, the direct interaction between the primary OAC and M2 macrophages in the 3D context, along with the release of the soluble anti-inflammatory factors by the M2 cells, significantly contributed to a better understanding of the molecular mechanisms responsible for immune cell-mediated OA healing.
Collapse
Affiliation(s)
- Nilotpal Majumder
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Subhadeep Roy
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Aarushi Sharma
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Shuchi Arora
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Raju Vaishya
- Indraprastha Apollo Hospitals Delhi, New Delhi 110076, India
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi 110016, India
| |
Collapse
|
39
|
Cao M, Wang Z, Lan W, Xiang B, Liao W, Zhou J, Liu X, Wang Y, Zhang S, Lu S, Lang J, Zhao Y. The roles of tissue resident macrophages in health and cancer. Exp Hematol Oncol 2024; 13:3. [PMID: 38229178 PMCID: PMC10790434 DOI: 10.1186/s40164-023-00469-0] [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: 10/08/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024] Open
Abstract
As integral components of the immune microenvironment, tissue resident macrophages (TRMs) represent a self-renewing and long-lived cell population that plays crucial roles in maintaining homeostasis, promoting tissue remodeling after damage, defending against inflammation and even orchestrating cancer progression. However, the exact functions and roles of TRMs in cancer are not yet well understood. TRMs exhibit either pro-tumorigenic or anti-tumorigenic effects by engaging in phagocytosis and secreting diverse cytokines, chemokines, and growth factors to modulate the adaptive immune system. The life-span, turnover kinetics and monocyte replenishment of TRMs vary among different organs, adding to the complexity and controversial findings in TRMs studies. Considering the complexity of tissue associated macrophage origin, macrophages targeting strategy of each ontogeny should be carefully evaluated. Consequently, acquiring a comprehensive understanding of TRMs' origin, function, homeostasis, characteristics, and their roles in cancer for each specific organ holds significant research value. In this review, we aim to provide an outline of homeostasis and characteristics of resident macrophages in the lung, liver, brain, skin and intestinal, as well as their roles in modulating primary and metastatic cancer, which may inform and serve the future design of targeted therapies.
Collapse
Affiliation(s)
- Minmin Cao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zihao Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wanying Lan
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- Guixi Community Health Center of the Chengdu High-Tech Zone, Chengdu, China
| | - Binghua Xiang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjun Liao
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jie Zhou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaomeng Liu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yiling Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shichuan Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shun Lu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jinyi Lang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Zhao
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
40
|
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
|
41
|
Balog S, Jeong S, Asahina K. Recruitment of large peritoneal macrophages to capsular fibrosis developed on the liver surface. FASEB J 2024; 38:e23327. [PMID: 38019178 DOI: 10.1096/fj.202301187r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/15/2023] [Accepted: 11/08/2023] [Indexed: 11/30/2023]
Abstract
Upon injury to Glisson's capsule, mesothelial cells covering the liver surface differentiate into myofibroblasts and participate in capsular fibrosis. In the fibrotic area, infiltrating macrophages are present, but their origin and role in capsular fibrosis remain elusive. In the present study, we examined whether macrophages in the peritoneal cavity migrate to the liver and participate in capsular fibrosis. Capsular fibrosis was induced by intraperitoneal injection of chlorhexidine gluconate. Chlorhexidine gluconate treatment induced disappearance of CD11bHigh F4/80High large peritoneal macrophages from the peritoneal cavity. Transplantation of TIMD4+ large peritoneal macrophages to the mouse peritoneal cavity resulted in their recruitment to the fibrotic area of the liver. Bone marrow-derived monocytes were also recruited to the chlorhexidine gluconate-induced fibrotic area upon their transplantation to the peritoneal cavity. However, bone marrow-derived macrophages, Kupffer cells, peritoneal B cells, and small peritoneal macrophages prepared from chlorhexidine gluconate-treated mice did not exhibit such potential. In the hepatic fibrotic area, peritoneal macrophages lost expression of unique markers (Gata6, Timd4) and increased expression of genes involved in inflammation (Il1b, Il6, Tnf) and extracellular matrix remodeling (Mmp13, Timp1). Depletion of peritoneal macrophages by clodronate liposomes reduced capsular fibrosis. Our data indicate that large peritoneal macrophages are recruited to the injured liver surface and promote capsular fibrosis by inducing inflammation and extracellular matrix remodeling. Modulating the function of peritoneal macrophages might be a new approach for suppressing capsular fibrosis.
Collapse
Affiliation(s)
- Steven Balog
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Soi Jeong
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Kinji Asahina
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
- Central Research Laboratory, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
42
|
Stumpff JP, Kim SY, McFadden MI, Nishida A, Shirazi R, Steuerman Y, Gat-Viks I, Forero A, Nair MG, Morrison J. Pleural macrophages translocate to the lung during infection to promote improved influenza outcomes. Proc Natl Acad Sci U S A 2023; 120:e2300474120. [PMID: 38100417 PMCID: PMC10743374 DOI: 10.1073/pnas.2300474120] [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: 01/10/2023] [Accepted: 10/30/2023] [Indexed: 12/17/2023] Open
Abstract
Seasonal influenza results in 3 to 5 million cases of severe disease and 250,000 to 500,000 deaths annually. Macrophages have been implicated in both the resolution and progression of the disease, but the drivers of these outcomes are poorly understood. We probed mouse lung transcriptomic datasets using the Digital Cell Quantifier algorithm to predict immune cell subsets that correlated with mild or severe influenza A virus (IAV) infection outcomes. We identified a unique lung macrophage population that transcriptionally resembled small serosal cavity macrophages and whose presence correlated with mild disease. Until now, the study of serosal macrophage translocation in the context of viral infections has been neglected. Here, we show that pleural macrophages (PMs) migrate from the pleural cavity to the lung after infection with IAV. We found that the depletion of PMs increased morbidity and pulmonary inflammation. There were increased proinflammatory cytokines in the pleural cavity and an influx of neutrophils within the lung. Our results show that PMs are recruited to the lung during IAV infection and contribute to recovery from influenza. This study expands our knowledge of PM plasticity and identifies a source of lung macrophages independent of monocyte recruitment and local proliferation.
Collapse
Affiliation(s)
- James P. Stumpff
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA92521
| | - Sang Yong Kim
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA92521
| | - Matthew I. McFadden
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH43210
- Infectious Diseases Institute, The Ohio State University, Columbus, OH43210
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, WA98109
| | - Roksana Shirazi
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA92521
| | - Yael Steuerman
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv6997801, Israel
| | - Irit Gat-Viks
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv6997801, Israel
| | - Adriana Forero
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH43210
- Infectious Diseases Institute, The Ohio State University, Columbus, OH43210
| | - Meera G. Nair
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA92521
| | - Juliet Morrison
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA92521
| |
Collapse
|
43
|
Rigamonti A, Villar J, Segura E. Monocyte differentiation within tissues: a renewed outlook. Trends Immunol 2023; 44:999-1013. [PMID: 37949783 DOI: 10.1016/j.it.2023.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
When recruited to mammalian tissues, monocytes differentiate into macrophages or dendritic cells (DCs). In the past few years, the existence of monocyte-derived DCs (moDCs) was questioned by the discovery of new DC populations with overlapping phenotypes. Here, we critically review the evidence for monocyte differentiation into DCs in tissues and highlight their specific functions. Recent studies have shown that monocyte-derived macrophages (moMacs) with distinct life cycles coexist in tissues, both at steady state and upon inflammation. Integrating studies in mice and humans, we highlight specific features of moMacs during inflammation and tissue repair. We also discuss the notion of monocyte differentiation occurring via a binary fate decision. Deciphering monocyte-derived cell properties is essential for understanding their role in nonresolving inflammation and how they might be targeted for therapies.
Collapse
Affiliation(s)
| | - Javiera Villar
- Institut Curie, PSL University, INSERM, U932, 26 Rue d'Ulm, Paris 75005, France
| | - Elodie Segura
- Institut Curie, PSL University, INSERM, U932, 26 Rue d'Ulm, Paris 75005, France.
| |
Collapse
|
44
|
Pang J, Koh TJ. Proliferation of monocytes and macrophages in homeostasis, infection, injury, and disease. J Leukoc Biol 2023; 114:532-546. [PMID: 37555460 PMCID: PMC10673715 DOI: 10.1093/jleuko/qiad093] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/30/2023] [Accepted: 07/31/2023] [Indexed: 08/10/2023] Open
Abstract
Monocytes (Mo) and macrophages (Mφ) play important roles in the function of tissues, organs, and systems of all animals during homeostasis, infection, injury, and disease. For decades, conventional wisdom has dictated that Mo and Mφ are end-stage cells that do not proliferate and that Mφ accumulation in tissues is the result of infiltration of Mo from the blood and subsequent differentiation to Mφ. However, reports from the early 1900s to the present describe evidence of Mo and Mφ proliferation in different tissues and contexts. The purpose of this review is to summarize both historical and current evidence for the contribution of Mφ proliferation to their accumulation in different tissues during homeostasis, infection, injury, and disease. Mφ proliferate in different organs and tissues, including skin, peritoneum, lung, heart, aorta, kidney, liver, pancreas, brain, spinal cord, eye, adipose tissue, and uterus, and in different species including mouse, rat, rabbit, and human. Mφ can proliferate at different stages of differentiation with infiltrating Mo-like cells proliferating in certain inflammatory contexts (e.g. skin wounding, kidney injury, bladder and liver infection) and mature resident Mφ proliferating in other inflammatory contexts (e.g. nematode infection, acetaminophen liver injury) and during homeostasis. The pathways involved in stimulating Mφ proliferation also may be context dependent, with different cytokines and transcription factors implicated in different studies. Although Mφ are known to proliferate in health, injury, and disease, much remains to be learned about the regulation of Mφ proliferation in different contexts and its impact on the homeostasis, injury, and repair of different organs and tissues.
Collapse
Affiliation(s)
- Jingbo Pang
- Center for Wound Healing and Tissue Regeneration, Department of Kinesiology and Nutrition, University of Illinois at Chicago, 1919 West Taylor Street, Chicago, IL 60612-7246, United States
| | - Timothy J Koh
- Center for Wound Healing and Tissue Regeneration, Department of Kinesiology and Nutrition, University of Illinois at Chicago, 1919 West Taylor Street, Chicago, IL 60612-7246, United States
| |
Collapse
|
45
|
Kim KH, Park D, Cho SY, Cho Y, Lee B, Jeong H, Lee Y, Lee Y, Nam KT. Role of histamine-mediated macrophage differentiation in clearance of metastatic bacterial infection. Front Immunol 2023; 14:1290191. [PMID: 38035074 PMCID: PMC10682073 DOI: 10.3389/fimmu.2023.1290191] [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: 09/07/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023] Open
Abstract
Macrophages are highly heterogeneous immune cells with a role in maintaining tissue homeostasis, especially in activating the defense response to bacterial infection. Using flow cytometric and single-cell RNA-sequencing analyses of peritoneal cells, we here show that small peritoneal macrophage and immature macrophage populations are enriched in histamine-deficient (Hdc -/-) mice, characterized by a CD11bmiF4/80loCCR2+MHCIIhi and CD11bloF4/80miTHBS1+IL-1α+ phenotype, respectively. Molecular characterization revealed that immature macrophages represent an abnormally differentiated form of large peritoneal macrophages with strong inflammatory properties. Furthermore, deficiency in histamine signaling resulted in significant impairment of the phagocytic activity of peritoneal macrophage populations, conferring high susceptibility to bacterial infection. Collectively, this study reveals the importance of histamine signaling in macrophage differentiation at the molecular level to maintain tissue homeostasis, offering a potential therapeutic target for bacterial infection-mediated diseases.
Collapse
Affiliation(s)
- Kwang H. Kim
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Donghwan Park
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soo Young Cho
- Department of Molecular and Life Science, Hanyang University College of Science and Convergence Technology, Ansan, Republic of Korea
| | - Yejin Cho
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Buhyun Lee
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Haengdueng Jeong
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yura Lee
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yourim Lee
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ki Taek Nam
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
46
|
Chen P, Pan Y, Ning X, Shi X, Zhong J, Fan X, Li W, Teng Y, Liu X, Yu B, Yang Y, Li H, Ou C. Targeted heart repair by Tβ4-loaded cardiac-resident macrophage-derived extracellular vesicles modified with monocyte membranes. Acta Biomater 2023; 169:372-386. [PMID: 37597679 DOI: 10.1016/j.actbio.2023.08.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023]
Abstract
Recent studies have demonstrated the critical role of cardiac-resident macrophages (cMacs) in the maintenance of physiological homeostasis. However, recruitment of circulating monocyte-derived macrophages decreases cMac levels post-myocardial infarction (MI). Transplanting cMacs is not an ideal option due to their low survival rates and the risk of immunological rejection. However, extracellular vesicle therapy has the potential to provide a feasible and safe alternative for cardiac repair. In this study, cell membrane-modified extracellular vesicles (MmEVs) were developed for heart repair by modifying cMac-derived extracellular vesicles (mEVs) with monocyte membranes, resulting in immune evasion and sequential targeted localization to damaged regions through expression of CD47 on MmEVs and strong affinity between monocyte membrane proteins and CCL2. Additionally, to fully exploit the potential clinical application of MmEVs and achieve a better curative effect, thymosin β4 (Tβ4) was loaded into the nanoparticles, resulting in Tβ4-MmEVs. In vitro experiments indicated that both the MmEVs and Tβ4-MmEVs promoted cardiomyocyte proliferation and endothelial cell migration. Animal experiments suggested that MI mice treated with MmEVs and Tβ4-MmEVs exhibited reduced myocardial fibrosis and increased vascular density compared to the control group. Thus, we posit that these targeted nanoparticles hold significant potential for MI adjuvant therapy and may open new avenues for cardiac repair and regeneration. STATEMENT OF SIGNIFICANCE: Extracellular vesicles (EVs) derived from bioactive parent cell sources involved in pathological and repair processes for cardiovascular disease have emerged as a compelling strategy for regenerative therapy. In this study, we constructed monocyte membrane-modified extracellular vesicles loaded with a drug (Tβ4-MmEVs) for heart repair that exhibit extraordinary abilities of immune evasion and sequential localization to damaged regions owing to the presence of CD47 and the strong affinity between monocytes and damaged cardiomyocytes and endothelial cells. The bioactivities of Tβ4-MmEVs on enhancing cardiomyocyte and endothelial cell proliferation were validated both in vitro and in vivo. Effective development and implementation of therapeutically membrane-modified nanoparticles from homologous origins can provide a reference for adjuvant therapy in clinical MI management.
Collapse
Affiliation(s)
- Peier Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan 523018, China
| | - Yuxuan Pan
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan 523018, China
| | - Xiaodong Ning
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan 523018, China
| | - Xu Shi
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan 523018, China
| | - Jianfeng Zhong
- Department of Cardiology, The Affiliated Hospital of Guangdong Medical University, Zhanjiang 524003, China
| | - Xianglin Fan
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan 523018, China
| | - Weirun Li
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan 523018, China
| | - Yintong Teng
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan 523018, China
| | - Xueting Liu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan 523018, China
| | - Bin Yu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yanhua Yang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan 523018, China.
| | - Hekai Li
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Caiwen Ou
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan 523018, China.
| |
Collapse
|
47
|
Xie MM, Dai B, Hackney JA, Sun T, Zhang J, Jackman JK, Jeet S, Irizarry-Caro RA, Fu Y, Liang Y, Bender H, Shamir ER, Keir ME, Bevers J, Nakamura G, Townsend MJ, Fox DA, Scherl A, Lee WP, Martin F, Godowski PJ, Pappu R, Yi T. An agonistic anti-signal regulatory protein α antibody for chronic inflammatory diseases. Cell Rep Med 2023; 4:101130. [PMID: 37490914 PMCID: PMC10439247 DOI: 10.1016/j.xcrm.2023.101130] [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: 12/24/2022] [Revised: 05/23/2023] [Accepted: 06/30/2023] [Indexed: 07/27/2023]
Abstract
Signal regulatory protein (SIRPα) is an immune inhibitory receptor expressed by myeloid cells to inhibit immune cell phagocytosis, migration, and activation. Despite the progress of SIRPα and CD47 antagonist antibodies to promote anti-cancer immunity, it is not yet known whether SIRPα receptor agonism could restrain excessive autoimmune tissue inflammation. Here, we report that neutrophil- and monocyte-associated genes including SIRPA are increased in inflamed tissue biopsies from patients with rheumatoid arthritis and inflammatory bowel diseases, and elevated SIRPA is associated with treatment-refractory ulcerative colitis. We next identify an agonistic anti-SIRPα antibody that exhibits potent anti-inflammatory effects in reducing neutrophil and monocyte chemotaxis and tissue infiltration. In preclinical models of arthritis and colitis, anti-SIRPα agonistic antibody ameliorates autoimmune joint inflammation and inflammatory colitis by reducing neutrophils and monocytes in tissues. Our work provides a proof of concept for SIRPα receptor agonism for suppressing excessive innate immune activation and chronic inflammatory disease treatment.
Collapse
Affiliation(s)
- Markus M Xie
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Bingbing Dai
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Jason A Hackney
- Department of OMNI Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | - Tianhe Sun
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Juan Zhang
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Janet K Jackman
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Surinder Jeet
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Ricardo A Irizarry-Caro
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA; Department of Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, CA, USA
| | - Yongyao Fu
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, CA, USA
| | - Yuxin Liang
- Department of Microchemistry, Proteomics, and Lipidomics and Next Generation Sequencing, Genentech, Inc., South San Francisco, CA, USA
| | - Hannah Bender
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Eliah R Shamir
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Mary E Keir
- Department of Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, CA, USA
| | - Jack Bevers
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, CA, USA
| | - Gerald Nakamura
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, CA, USA
| | - Michael J Townsend
- Department of Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, CA, USA
| | - David A Fox
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alexis Scherl
- Department of Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Wyne P Lee
- Department of Translational Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Flavius Martin
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA
| | - Paul J Godowski
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA.
| | - Rajita Pappu
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA.
| | - Tangsheng Yi
- Department of Immunology Discovery, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
48
|
McGrath-Morrow SA, Venezia J, Ndeh R, Michki N, Perez J, Singer BD, Cimbro R, Soloski M, Scott AL. Cellular and molecular dynamics in the lungs of neonatal and juvenile mice in response to E. coli. eLife 2023; 12:e82933. [PMID: 37266566 PMCID: PMC10264069 DOI: 10.7554/elife.82933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 06/01/2023] [Indexed: 06/03/2023] Open
Abstract
Bacterial pneumonia in neonates can cause significant morbidity and mortality when compared to other childhood age groups. To understand the immune mechanisms that underlie these age-related differences, we employed a mouse model of Escherichia coli pneumonia to determine the dynamic cellular and molecular differences in immune responsiveness between neonates (PND 3-5) and juveniles (PND 12-18), at 24, 48, and 72 hr. Cytokine gene expression from whole lung extracts was also quantified at these time points, using quantitative RT-PCR. E. coli challenge resulted in rapid and significant increases in neutrophils, monocytes, and γδT cells, along with significant decreases in dendritic cells and alveolar macrophages in the lungs of both neonates and juveniles. E. coli-challenged juvenile lung had significant increases in interstitial macrophages and recruited monocytes that were not observed in neonatal lungs. Expression of IFNγ-responsive genes was positively correlated with the levels and dynamics of MHCII-expressing innate cells in neonatal and juvenile lungs. Several facets of immune responsiveness in the wild-type neonates were recapitulated in juvenile MHCII-/- juveniles. Employing a pre-clinical model of E. coli pneumonia, we identified significant differences in the early cellular and molecular dynamics in the lungs that likely contribute to the elevated susceptibility of neonates to bacterial pneumonia and could represent targets for intervention to improve respiratory outcomes and survivability of neonates.
Collapse
Affiliation(s)
- Sharon A McGrath-Morrow
- Children's Hospital of Philadelphia Division of Pulmonary Medicine and SleepPhiladelphiaUnited States
| | - Jarrett Venezia
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public HealthBaltimoreUnited States
| | - Roland Ndeh
- Children's Hospital of Philadelphia Division of Pulmonary Medicine and SleepPhiladelphiaUnited States
| | - Nigel Michki
- Children's Hospital of Philadelphia Division of Pulmonary Medicine and SleepPhiladelphiaUnited States
| | - Javier Perez
- Children's Hospital of Philadelphia Division of Pulmonary Medicine and SleepPhiladelphiaUnited States
| | - Benjamin David Singer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine Northwestern, University Feinberg School of MedicineChicagoUnited States
| | - Raffaello Cimbro
- Department of Medicine, Division of Rheumatology, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Mark Soloski
- Department of Medicine, Division of Rheumatology, Johns Hopkins University, School of MedicineBaltimoreUnited States
| | - Alan L Scott
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public HealthBaltimoreUnited States
| |
Collapse
|
49
|
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
|
50
|
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
|