1
|
Dovhyi R, Dvukhriadkina A, Ostrovska K, Rudyk M, Verhovcova I, Vaivode K, Pjanova D, Ostapchenko L, Skivka L. Bacteriophage derived dsRNA induces polarized activation of alveolar macrophages from Balb/c and C57Bl/6 mice in vitro in sex- and age-dependent manner. Cell Immunol 2025; 408:104916. [PMID: 39787694 DOI: 10.1016/j.cellimm.2025.104916] [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: 09/25/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Bacteriophage-derived dsRNA (bp-dsRNA), also known as Larifan, is a poly-functional and wide-spectrum antiviral medication with potent interferonogenic activity. In the lungs of golden Syrian hamsters infected with SARS-CoV-2, Larifan substantially reduces viral load and decreases infection-induced pathological lesion severity. Alveolar macrophages (AM) are key sentinel cells in the lung, which play an important role in antiviral innate immune responses and, at the same time, can trigger infection-associated hyper-inflammatory response. This study revealed that treatment with bp-dsRNA (Larifan) in vitro modulates the functional profile of AM from intact Balb/c and C57Bl/6 mice. The pattern of the drug response depends on the animal strain, age and sex. AM from Balb/c mice generated a weaker response to the preparation as compared to cells from C57Bl/6 mice. Most emphatic responses to the treatment with bf-dsRNA (Larifan) were registered in AM from old males of both BALB/c and C57BL/6 strains with the strongest in the latter. AM from old C57BL/6 females were less likely to be influenced by the preparation. In most cases, exposure to bf-dsRNA (Larifan) increased AM phagocytic activity and was more often accompanied by the stimulation of intracellular reactive oxygen species generation, than by its decrease. In most animal groups, treatment with bf-dsRNA (Larifan) did not affect significantly CD206 expression and down-regulated CD80 expression in AM. Taken together, our findings suggest that bf-dsRNA (Larifan) not so much stimulates the bivalent phenotype of AM, as restrains their hyper-inflammatory responses through the control of antigen-presentation while preserving functional signatures typical of patrolling tissue-resident macrophages.
Collapse
Affiliation(s)
- R Dovhyi
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine
| | - A Dvukhriadkina
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine
| | - K Ostrovska
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine
| | - M Rudyk
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine.
| | - Irina Verhovcova
- Latvian Biomedical Research and Study Centre, Rātsupītes Street 1, k-1, Riga LV-1067, Latvia
| | - Kristine Vaivode
- Latvian Biomedical Research and Study Centre, Rātsupītes Street 1, k-1, Riga LV-1067, Latvia
| | - D Pjanova
- Latvian Biomedical Research and Study Centre, Rātsupītes Street 1, k-1, Riga LV-1067, Latvia
| | - L Ostapchenko
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine
| | - L Skivka
- Educational and Scientific Centre "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, 2, Hlushkov Avenue, Kyiv 03022, Ukraine
| |
Collapse
|
2
|
Wang X, Han C, Xia J, Cui C, Min P, Meng X, Sun Y, Wen K, Feng C, Zhang Y, Li K. Ultrasound-Mediated Piezoelectric Microneedles Regulating Macrophage Polarization and Remodeling Pathological Microenvironment for Lymphedema Improvement. ACS NANO 2025; 19:1447-1462. [PMID: 39810381 DOI: 10.1021/acsnano.4c14292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Lymphedema, a severe and complex inflammatory disease caused by lymphatic system insufficiency and impeded lymphatic drainage that causes an enormous physical and psychological burden on patients and may even lead to death, has long been a challenging issue in the medical field. Clinically, conventional approaches including surgical treatment and conservative treatment have been employed for lymphedema therapy, but their curative effect is still unsatisfactory because of high operational difficulty, high cost, and long-term reliance. In this study, a novel kind of piezoelectric microneedle driven by ultrasound (US) is proposed to regulate macrophage polarization and remodel the pathological inflammatory microenvironment in a noninvasive manner, thereby promoting lymphatic regeneration and improving lymphedema. US-mediated piezoelectric microneedles can significantly enhance the anti-inflammatory M2-type polarization of macrophages while suppressing pro-inflammatory M1-type polarization in vitro. Enhancements in macrophage M2 polarization can trigger increased secretion of anti-inflammatory factors (e.g., IL-4, IL-10, and Arg-1) that promote inflammatory microenvironment remodeling and immune rebalancing. In a mouse-tail lymphedema model, a higher proportion of M2 macrophage polarization marker CD206 is observed accompanied by normal lymphangiogenesis and lymphedema subsiding following piezoelectric microneedles with US stimulation treatment. Additionally, based on RNA sequencing and mechanistic investigation, it was revealed that US-mediated piezoelectric microneedles can activate signaling pathways related to M2 macrophage polarization that regulate inflammatory responses and improve lymphatic function, consequently alleviating lymphedema. Collectively, this study provides a new strategy for lymphedema therapy in a noninvasive and drug-free manner as well as a potent tool to manipulate macrophages for other immunological diseases.
Collapse
Affiliation(s)
- Xueqian Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Chengyao Han
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Jingyi Xia
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chunxiao Cui
- Department of Burns and Plastic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Peiru Min
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Xinxian Meng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Yuhao Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Ke Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Chuanliang Feng
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yixin Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Ke Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| |
Collapse
|
3
|
Zhang Y, Li B, Gu W, Fan L, Wang X, Xu M, Zhu M, Jin C. Hepatoma cell-derived exosomal SNORD52 mediates M2 macrophage polarization by activating the JAK2/STAT6 pathway. Discov Oncol 2025; 16:36. [PMID: 39804511 PMCID: PMC11730036 DOI: 10.1007/s12672-024-01700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND A recent study revealed the oncogenic role of box C/D small nucleolar RNA 52 (SNORD52) in hepatocellular carcinoma (HCC) by facilitating the aggressive phenotypes of hepatoma cells. However, the potential role of exosomal SNORD52 in macrophage polarization during HCC progression remains poorly understood. METHODS Exosomes were isolated from hepatoma cells. Western blotting and flow cytometry were performed to determine the levels of M2 macrophage polarization markers. SNORD52 expression was assessed using qRT-PCR. The levels of JAK2/STAT6 pathway-related proteins were analyzed using western blotting. RESULTS SNORD52 was enriched in exosomes derived from hepatoma cells and in plasma samples from patients with HCC. Hepatoma cell-derived exosomal SNORD52 was internalized by THP-1 macrophages. SNORD52 overexpression increased the levels of M2 macrophage polarization markers and JAK2/STAT6 pathway-related proteins Additionally, hepatoma cell-derived exosomal SNORD52 interacted with the JAK2/STAT6 pathway to mediate M2 macrophage polarization. CONCLUSIONS Our findings revealed that hepatoma cell-derived exosomal SNORD52 induces M2 macrophage polarization by activating the JAK2/STAT6 pathway.
Collapse
Affiliation(s)
- Yaqiong Zhang
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China
| | - Bo Li
- Department of Ultrasound, Taizhou Hospital, Zhejiang University, Taizhou Enze Medical Center (Group), Taizhou, 318000, Zhejiang, China
| | - Wanhong Gu
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China
| | - Linna Fan
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiaofan Wang
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China
| | - Meifen Xu
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China
| | - Minqi Zhu
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China.
| | - Chong Jin
- Department of Hepatobiliary Pancreatic Splenic Surgery, Taizhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Taizhou, 318000, Zhejiang, China.
| |
Collapse
|
4
|
Hawthorne BC, Engel S, McCarthy MBR, Cote MC, Mazzocca AD, Coyner KJ. Biologic Adjuvants to Rotator Cuff Repairs Induce Anti-inflammatory Macrophage 2 Polarization and Reduce Inflammatory Macrophage 1 Polarization In Vitro. Arthroscopy 2025; 41:32-41. [PMID: 38735413 DOI: 10.1016/j.arthro.2024.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE To examine the effect of various biologic adjuvants on the polarization of macrophages in an in vitro model for rotator cuff tears. METHODS Tissue was harvested from 6 patients undergoing arthroscopic rotator cuff repair. An in vitro model of the supraspinatus and subacromial bursa was created and treated with control, platelet-rich plasma (PRP), autologous activated serum (AAS), or a combination of PRP+AAS. The effect of treatment on macrophage polarization between M1 proinflammatory macrophages or M2 anti-inflammatory macrophages was measured using gene expression, protein expression, flow cytometry, and nitric oxide production. RESULTS Tendon and bursa treated with PRP, AAS, and PRP+AAS significantly decreased the gene expression of M1 markers interleukin (IL)-12 and tumor necrosis factor-alpha while significantly increasing the expression of M2 markers arginase, IL-10, and transforming growth factor-β (P < .05) compared with treatment with control. Enzyme-linked immunosorbent assay analysis of protein production demonstrated that, compared with control, coculture treated with PRP, AAS, and PRP+AAS significantly decreased markers of M1-macrophages (IL-6, IL-12, and tumor necrosis factor-alpha) while significantly increasing the expression of markers of M2-macrophages (arginase, IL-10, and transforming growth factor-beta) (P < .05). Flow cytometry analysis of surface markers demonstrated that compared with control, tendon and bursa treated with PRP, AAS, and PRP+AAS significantly decreased markers of M1-macrophages (CD80, CD86, CD64, CD16) while significantly increasing the expression of markers of M2-macrophages (CD163 and CD206) (P < .05). Treatment of the coculture with PRP, AAS, and PRP+AAS consistently demonstrated a decrease in nitric oxide production (P < .05) compared with control. AAS and PRP+AAS demonstrated an increased macrophage shift to M2 compared with PRP alone, whereas there was not as uniform of a shift when comparing PRP+AAS with AAS alone. CONCLUSIONS In an in vitro model of rotator cuff tears, the treatment of supraspinatus tendon and subacromial bursa with PRP, AAS, and PRP+AAS demonstrated an increase in markers of anti-inflammatory M2-macrophages and a concomitant decrease in markers of proinflammatory M1-macrophages. AAS and PRP+AAS contributed to a large shift to macrophage polarization to the anti-inflammatory M2 compared with PRP. CLINICAL RELEVANCE The mechanism of biologic adjuvant effects on the rotator cuff remains poorly understood. This study suggests that they may contribute to polarization of macrophages for their proinflammatory (M1) state to the anti-inflammatory (M2) state.
Collapse
Affiliation(s)
| | - Sam Engel
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, U.S.A
| | - Mary Beth R McCarthy
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, U.S.A
| | - Mark C Cote
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, U.S.A
| | - Augustus D Mazzocca
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, U.S.A
| | - Katherine J Coyner
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, U.S.A..
| |
Collapse
|
5
|
Wang Y, Zhang Y, Ma M, Zhuang X, Lu Y, Miao L, Lu X, Cui Y, Cui W. Mechanisms underlying the involvement of peritoneal macrophages in the pathogenesis and novel therapeutic strategies for dialysis-induced peritoneal fibrosis. Front Immunol 2024; 15:1507265. [PMID: 39749340 PMCID: PMC11693514 DOI: 10.3389/fimmu.2024.1507265] [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: 10/07/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Long-term exposure of the peritoneum to peritoneal dialysate results in pathophysiological changes in the anatomical organization of the peritoneum and progressive development of peritoneal fibrosis. This leads to a decline in peritoneal function and ultrafiltration failure, ultimately necessitating the discontinuation of peritoneal dialysis, severely limiting the potential for long-term maintenance. Additionally, encapsulating peritoneal sclerosis, a serious consequence of peritoneal fibrosis, resulting in patients discontinuing PD and significant mortality. The causes and mechanisms underlying peritoneal fibrosis in patients undergoing peritoneal dialysis remain unknown, with no definitive treatment available. However, abnormal activation of the immune system appears to be involved in altering the structure of the peritoneum and promoting fibrotic changes. Macrophage infiltration and polarization are key contributors to pathological injury within the peritoneum, showing a strong correlation with the epithelial-to-mesenchymal transition of mesothelial cells and driving the process of fibrosis. This article discusses the role and mechanisms underlying macrophage activation-induced peritoneal fibrosis resulting from PD by analyzing relevant literature from the past decade and provides an overview of recent therapeutic approaches targeting macrophages to treat this condition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yingchun Cui
- Department of Nephrology, Second Hospital of Jilin University,
Changchun, China
| | - Wenpeng Cui
- Department of Nephrology, Second Hospital of Jilin University,
Changchun, China
| |
Collapse
|
6
|
Soboh S, Vorontsova A, Farhoud M, Barash U, Naroditsky I, Gross-Cohen M, Weissmann M, Yasuhiko N, Woolf AS, Roberts NA, Shaked Y, Ilan N, Vlodavsky I. Tumor- and host-derived heparanase-2 (Hpa2) attenuates tumorigenicity: role of Hpa2 in macrophage polarization and BRD7 nuclear localization. Cell Death Dis 2024; 15:894. [PMID: 39695102 DOI: 10.1038/s41419-024-07262-9] [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: 07/31/2024] [Revised: 11/14/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024]
Abstract
Little attention was given to heparanase 2 (Hpa2) over the last two decades, possibly because it lacks a heparan sulfate (HS)-degrading activity typical of heparanase. Emerging results suggest, nonetheless, that Hpa2 plays a role in human pathologies, including cancer progression where it functions as a tumor suppressor. Here, we examined the role of Hpa2 in cervical carcinoma. We report that high levels of Hpa2 correlate with prolonged survival of cervical carcinoma patients. Strong staining intensity of Hpa2 also correlates with low tumor grade. Overexpression of Hpa2 in SiHa cervical carcinoma cells resulted in tumor xenografts that were two-fold smaller than control tumors. Interestingly, even smaller tumor xenografts were developed by SiHa cells overexpressing the Pro140Arg and Asn543Ile Hpa2 missense mutations that were identified in patients diagnosed with urofacial syndrome (UFS). Utilizing the Ras recruitment system, we identified bromodomain-containing protein 7 (BRD7) to interact with Hpa2 and found that both BRD7 and the Hpa2 mutants are translocated to the cell nucleus in tumors developed by the Pro140Arg and Asn543Ile Hpa2 mutants. Utilizing our newly developed conditional Hpa2-KO mice, we further show that Hpa2 plays a critical role in macrophage polarization; in the absence of Hpa2, macrophages are shifted towards pro-tumorigenic, M2 phenotype. Notably, implanting SiHa cervical carcinoma cells together with Hpa2-KO macrophages promoted tumor growth. These results support, and further expand, the notion that Hpa2 functions as a tumor suppressor, co-operating with another tumor suppressor, BRD7.
Collapse
Affiliation(s)
- Soaad Soboh
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Avital Vorontsova
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Malik Farhoud
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Uri Barash
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Inna Naroditsky
- Departments of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Miriam Gross-Cohen
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Marina Weissmann
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Nishioka Yasuhiko
- Department of Respiratory Medicine and Rheumatology, Tokushima University, Tokushima, Japan
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Neil A Roberts
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Yuval Shaked
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel.
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| |
Collapse
|
7
|
Dobson GP, Morris JL, Letson HL. Traumatic brain injury: Symptoms to systems in the 21st century. Brain Res 2024; 1845:149271. [PMID: 39395646 DOI: 10.1016/j.brainres.2024.149271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024]
Abstract
Severe traumatic brain injury (TBI) is a devastating injury with a mortality of ∼ 25-30 %. Despite decades of high-quality research, no drug therapy has reduced mortality. Why is this so? We argue two contributing factors for the lack of effective drug therapies include the use of specific-pathogen free (SPF) animals for translational research and the flawed practice of single-nodal targeting for drug design. A revolution is required to better understand how the whole body responds to TBI, identify new markers of its progression, and discover new system-acting drugs to treat it. In this review, we present a brief history of TBI, discuss its system's pathophysiology and propose a new research strategy for the 21st century. TBI progression develops from injury signals radiating from the primary impact, which can cause local ischemia, hemorrhage, excitotoxicity, cellular depolarization, immune dysfunction, sympathetic hyperactivity, blood-brain barrier breach, coagulopathy and whole-body dysfunction. Metabolic reprograming of immune cells drives neuroinflammation and secondary injury processes. We propose if sympathetic hyperactivity and immune cell activation can be corrected early, cardiovascular function and endothelial-glycocalyx-mitochondrial coupling can be restored, and secondary injury minimized with improved patient outcomes. The therapeutic goal is to switch the injury phenotype to a healing phenotype by restoring homeostasis and maintaining sufficient tissue O2 delivery. We have been developing a small-volume fluid therapy comprising adenosine, lidocaine and magnesium (ALM) to treat TBI and have shown that it blunts the CNS-stress response, supports cardiovascular function and reduces secondary injury. Future research will investigate its suitability for human translation.
Collapse
Affiliation(s)
- Geoffrey P Dobson
- Heart, Sepsis and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland 4811, Australia.
| | - Jodie L Morris
- Heart, Sepsis and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland 4811, Australia.
| | - Hayley L Letson
- Heart, Sepsis and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland 4811, Australia.
| |
Collapse
|
8
|
Su J, Tu Y, Hu X, Wang Y, Chen M, Cao X, Shao M, Zhang F, Ding W. Ambient PM 2.5 orchestrates M1 polarization of alveolar macrophages via activating glutaminase 1-mediated glutaminolysis in acute lung injury. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 366:125467. [PMID: 39653263 DOI: 10.1016/j.envpol.2024.125467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
The temporary explosive growth events of atmospheric fine particulate matter (PM2.5) pollution during late autumn and winter seasons still frequently occur in China. High-concentration exposure to PM2.5 aggravates lung inflammation, leading to acute lung injury (ALI). Alveolar macrophages (AMs) participate in PM2.5-induced pulmonary inflammation and injury. The polarization of AMs is dependent on metabolic reprogramming. However, the mechanism underlying the PM2.5-induced glutaminase-mediated glutaminolysis in AM polarization is still largely obscure. In this study, we found that PM2.5-treated mice exhibited pulmonary dysfunction and inflammation. The concentrations of glutamate and succinate were increased in PM2.5-treated lungs and AMs compared with the controls, whereas glutamine and α-ketoglutarate (α-KG) levels were decreased, indicating that glutaminolysis in AMs was aberrantly activated as evidenced by increased mRNA and protein levels of GLS1 after PM2.5 exposure. Moreover, we determined that the GLS1/nuclear factor kappa-B (NF-κB)/hypoxia-inducible factor-1α (HIF-1α) pathway regulated M1 polarization of AMs upon PM2.5 exposure. Inhibition of glutaminolysis by GLS1 specific inhibitor CB-839 and GLS1 siRNA significantly decreased PM2.5-induced M1 macrophage polarization and attenuated pulmonary damage. Taken together, our findings reveal a novel mechanism by which a metabolic program regulates M1 polarization of AMs and suggest that GLS1-mediated glutaminolysis is a potential therapeutic target for treating PM2.5-induced ALI.
Collapse
Affiliation(s)
- Jingran Su
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yikun Tu
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China; Sino-Danish Center for Education and Research, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoqi Hu
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanli Wang
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mo Chen
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China; Sino-Danish Center for Education and Research, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xue Cao
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengyao Shao
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fang Zhang
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenjun Ding
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
9
|
Wang X, Liu D. Macrophage Polarization: A Novel Target and Strategy for Pathological Scarring. Tissue Eng Regen Med 2024; 21:1109-1124. [PMID: 39352458 PMCID: PMC11589044 DOI: 10.1007/s13770-024-00669-7] [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: 06/28/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Abnormal scarring imposes considerable challenges and burdens on the lives of patients and healthcare system. Macrophages at the wound site are found to be of great concern to overall wound healing. There have been many studies indicating an inextricably link between dysfunctional macrophages and fibrotic scars. Macrophages are not only related to pathogen destruction and phagocytosis of apoptotic cells, but also involved in angiogenesis, keratinization and collagen deposition. These abundant cell functions are attributed to specific heterogeneity and plasticity of macrophages, which also add an extra layer of complexity to correlational researches. METHODS This article summarizes current understanding of macrophage polarization in scar formation and several prevention and treatment strategies on pathological scarring related to regulation of macrophage behaviors by utilizing databases such as PubMed, Google Scholar and so on. RESULTS There are many studies proving that macrophages participate in the course of wound healing by converting their predominant phenotype. The potential of macrophages in managing hypertrophic scars and keloid lesions have been underscored. CONCLUSION Macrophage polarization offers new prevention strategies for pathological scarring. Learning about and targeting at macrophages may be helpful in achieving optimum wound healing.
Collapse
Affiliation(s)
- Xinyi Wang
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
- Queen Mary Academy, Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Dewu Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
10
|
Asaduzzaman M, Pavlov I, St-Jean G, Zhu Y, Castex M, Chorfi Y, Del Castillo JRE, Zhou T, Alassane-Kpembi I. Phosphorylation of Zearalenone Retains Its Toxicity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:26491-26503. [PMID: 39549027 DOI: 10.1021/acs.jafc.4c06889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2024]
Abstract
Microbial biotransformation of Zearalenone (ZEN) is a promising deactivation approach. The residual toxicity and stability of Zearalenone-14-phosphate (ZEN-14-P) and Zearalenone-16-phosphate (ZEN-16-P), two novel microbial phosphorylation products of ZEN, remain unknown. We investigated the cytotoxicity, oxidative stress, proinflammatory, and estrogenic activity of phosphorylated ZENs using porcine intestinal cells, uterine explants, and human endometrial cells and traced their metabolic fate by liquid chromatography-tandem mass spectrometry (LC-MS)/MS analysis. The phosphorylated ZENs significantly decreased the viability of the IPEC-J2 and Ishikawa cells. Similar to ZEN, phosphorylation products induced significant oxidative stress, activated the expression of proinflammatory cytokines, and demonstrated estrogenic activity through upregulation of estrogen-responsive genes, activation of alkaline phosphatase, and proliferation of endometrial glands. LC-MS/MS analysis pointed out that although phosphorylated ZENs are partially hydrolyzed to ZEN, their respective metabolic pathways differ. We conclude that phosphorylation might not be sufficient to detoxify ZEN, leaving its cytotoxic, proinflammatory, and estrogenic properties intact.
Collapse
Affiliation(s)
- Muhammad Asaduzzaman
- Département de Biomédecine vétérinaire, Faculté de Médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec J2S 2M2, Canada
| | - Ivan Pavlov
- Département de Biomédecine vétérinaire, Faculté de Médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec J2S 2M2, Canada
| | - Guillaume St-Jean
- Département de Pathologie et Microbiologie, Faculté de Médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec J2S 2M2, Canada
| | - Yan Zhu
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada
| | - Mathieu Castex
- Lallemand SAS, 19 rue des Briquetiers, BP 59, 31702 Blagnac Cedex, France
| | - Younes Chorfi
- Département de Biomédecine vétérinaire, Faculté de Médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec J2S 2M2, Canada
| | - Jérôme R E Del Castillo
- Département de Biomédecine vétérinaire, Faculté de Médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec J2S 2M2, Canada
| | - Ting Zhou
- Guelph Research and Development Centre, Agriculture and Agri-Food Canada, Guelph, ON N1G 5C9, Canada
| | - Imourana Alassane-Kpembi
- Département de Biomédecine vétérinaire, Faculté de Médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec J2S 2M2, Canada
| |
Collapse
|
11
|
Kim YJ, Nanda SS, Jiang F, Pyo SY, Han JY, Koh SS, Kang TH. Pancreatic Adenocarcinoma Up-Regulated Factor (PAUF) Transforms Human Monocytes into Alternative M2 Macrophages with Immunosuppressive Action. Int J Mol Sci 2024; 25:11545. [PMID: 39519098 PMCID: PMC11547018 DOI: 10.3390/ijms252111545] [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: 10/02/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) promote immune evasion, cancer cell proliferation, and metastasis. Ongoing research is focused on finding ways to prevent tumor growth by inhibiting TAM polarization, which has shown a correlation with unfavorable prognosis in clinical studies. Pancreatic adenocarcinoma up-regulated factor (PAUF) is a protein secreted from pancreatic cancer (PC) and acts as a TME modulator that affects the TME by acting on not only cancer cells but also stromal cells and immune cells. Tumor cells can evade the immune system by PAUF binding to Toll-like receptor (TLR) in monocytes, as this research shows. In this study, the examination centered around the recruitment of human monocytes by PAUF and the subsequent differentiation into macrophages. In an in vitro chemotaxis assay, PAUF induced chemotactic migration of TLR2-mediated monocytes. In addition, PAUF induced differentiation of monocytes into M2 macrophages, which was verified based on expressing surface markers and cytokines and morphological analysis. The inhibition of T cell proliferation and function was observed in differentiated M2 macrophages. To conclude, these findings indicate that PAUF functions as a promoter of cancer progression by regulating the recruitment and differentiation of macrophages within TMEs, ultimately causing immunosuppression.
Collapse
Affiliation(s)
- Yeon Jeong Kim
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
- Department of Biomedical Sciences, Dong-A University, Busan 49315, Republic of Korea
| | - Sitansu Sekhar Nanda
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Fen Jiang
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Seung Yeon Pyo
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Jin-Yeong Han
- Department of Laboratory Medicine, College of Medicine, Dong-A University, Busan 49201, Republic of Korea;
| | - Sang Seok Koh
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| | - Tae Heung Kang
- Prestige Biopharma IDC, Busan 46726, Republic of Korea; (Y.J.K.); (S.S.N.); (F.J.); (S.Y.P.); (S.S.K.)
| |
Collapse
|
12
|
Colaço M, Cruz MT, de Almeida LP, Borges O. Mannose and Lactobionic Acid in Nasal Vaccination: Enhancing Antigen Delivery via C-Type Lectin Receptors. Pharmaceutics 2024; 16:1308. [PMID: 39458637 PMCID: PMC11510408 DOI: 10.3390/pharmaceutics16101308] [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: 08/20/2024] [Revised: 09/24/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Nasal vaccines are a promising strategy for enhancing mucosal immune responses and preventing diseases at mucosal sites by stimulating the secretion of secretory IgA, which is crucial for early pathogen neutralization. However, designing effective nasal vaccines is challenging due to the complex immunological mechanisms in the nasal mucosa, which must balance protection and tolerance against constant exposure to inhaled pathogens. The nasal route also presents unique formulation and delivery hurdles, such as the mucous layer hindering antigen penetration and immune cell access. METHODS This review focuses on cutting-edge approaches to enhance nasal vaccine delivery, particularly those targeting C-type lectin receptors (CLRs) like the mannose receptor and macrophage galactose-type lectin (MGL) receptor. It elucidates the roles of these receptors in antigen recognition and uptake by antigen-presenting cells (APCs), providing insights into optimizing vaccine delivery. RESULTS While a comprehensive examination of targeted glycoconjugate vaccine development is outside the scope of this study, we provide key examples of glycan-based ligands, such as lactobionic acid and mannose, which can selectively target CLRs in the nasal mucosa. CONCLUSIONS With the rise of new viral infections, this review aims to facilitate the design of innovative vaccines and equip researchers, clinicians, and vaccine developers with the knowledge to enhance immune defenses against respiratory pathogens, ultimately protecting public health.
Collapse
Affiliation(s)
- Mariana Colaço
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria T. Cruz
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Olga Borges
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (M.C.); (M.T.C.); (L.P.d.A.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
13
|
Liu Z, Ting Y, Li M, Li Y, Tan Y, Long Y. From immune dysregulation to organ dysfunction: understanding the enigma of Sepsis. Front Microbiol 2024; 15:1415274. [PMID: 39252831 PMCID: PMC11381394 DOI: 10.3389/fmicb.2024.1415274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
Sepsis is a syndrome precipitated by immune dysregulation in response to infection, and represents a pivotal factor in global mortality attributed to diseases. The recent consensus delineates sepsis as a perilous state of organ dysfunction arising from the host's maladaptive reaction to infection. It masks the complexity and breadth of the immune mechanisms involved in sepsis, which is characterized by simultaneous hyperinflammation and immunosuppression. Sepsis is highly correlated with the dysregulation of immune response, which is mainly mediated by various immune cells and their interactions. This syndrome can lead to a plethora of complications, encompassing systemic inflammatory response, metabolic disturbances, infectious shock, MODS, and DIC. Furthermore, more research studies have been conducted on sepsis in the past few years. The pathological characteristics of sepsis have been improved or treated by targeting signaling pathways like NF-B, JAK-STAT, PI3K-Akt, and p38-MAPK. Combined drug therapy is better than single drug therapy for sepsis. This article will review the latest progress in the pathogenesis and treatment of sepsis.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Infectious Disease, Graduate Collaborative Training Base of Zhuzhou, Hengyang Medical School, University of South China, Hengyang, China
- Department of Infectious Disease, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Yuan Ting
- Department of Infectious Disease, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Miao Li
- Jishou University Zhuzhou Clinical College, Medical College, Jishou University, Zhuzhou, China
- Medical College, Jishou University, Xiangxi Tujia and Miao Autonomous Prefecture, Zhuzhou, China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Yingzheng Tan
- Department of Infectious Disease, Graduate Collaborative Training Base of Zhuzhou, Hengyang Medical School, University of South China, Hengyang, China
- Department of Infectious Disease, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Yunzhu Long
- Department of Infectious Disease, Graduate Collaborative Training Base of Zhuzhou, Hengyang Medical School, University of South China, Hengyang, China
- Department of Infectious Disease, Zhuzhou Central Hospital, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| |
Collapse
|
14
|
Zheng S, He H, Zheng J, Zhu X, Lin N, Wu Q, Wei E, Weng C, Chen S, Huang X, Jian C, Guan S, Yang C. Machine learning-based screening and validation of liver metastasis-specific genes in colorectal cancer. Sci Rep 2024; 14:17679. [PMID: 39085446 PMCID: PMC11291988 DOI: 10.1038/s41598-024-68706-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
Colorectal liver metastasis (CRLM) is challenging in the clinical treatment of colorectal cancer. Limited research has been conducted on how CRLM develops. RNA sequencing data were obtained from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA). Four machine learning algorithms were used to screen the hub CRLM-specific genes, including Least Absolute Shrinkage and Selection Operator (Lasso), Random forest, SVM-RFE, and XGboost. The model for identifying CRLM was developed using stepwise logistic regression and was validated using internal and independent datasets. The prognostic value of hub CRLM-specific genes was assessed using the Lasso-Cox method. The in vitro experiments were performed using SW620 cells. The CRLM identification model was developed based on four CRLM-specific genes (SPP1, ZG16, P2RY14, and PRKAR2B), and the model efficacy was validated using GSE41258 and three external cohorts. Five CRLM-specific prognostic hub genes, SPP1, ZG16, P2RY14, CYP2E1, and C5, were identified using the Lasso-Cox algorithm, and a risk score was constructed. The risk score was validated using the GSE39582 cohort. Three genes have both efficacy in identifying CRLM and prognostic value: ZG16, P2RY14, and SPP1. Immune infiltration and enrichment analyses demonstrated that SPP1 was associated with M2 macrophage polarization and extracellular matrix remodeling. In vitro experiments indicated that SPP1 may act as a cancer-promoting factor. The hub CRLM-specific gene SPP1 can help determine the diagnosis, prognosis, and immune infiltration of patients with CRLM.
Collapse
Affiliation(s)
- Shiyao Zheng
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Hongxin He
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Jianfeng Zheng
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Xingshu Zhu
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, 350025, People's Republic of China
| | - Nan Lin
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, 350025, People's Republic of China
- Fuzong Clinical Medical College of Fujian Medical University, Department of General Surgery, 900th Hospital of Joint Logistics Support Force, PLA, Fuzhou, 350025, People's Republic of China
| | - Qing Wu
- Department of Oncology, Molecular Oncology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People's Republic of China
| | - Enhao Wei
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Caiming Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350002, People's Republic of China
| | - Shuqian Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, People's Republic of China
| | - Xinxiang Huang
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Chenxing Jian
- School of Clinical Medicine, Fujian Medical University, Fuzhou, 350108, People's Republic of China.
- Department of Anorectal Surgery, Afliated Hospital of Putian University, Putian, 351106, People's Republic of China.
| | - Shen Guan
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China.
| | - Chunkang Yang
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China.
- Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, People's Republic of China.
| |
Collapse
|
15
|
Fransen LFH, Leonard MO. Mononuclear phagocyte sub-types in vitro display diverse transcriptional responses to dust mite exposure. Sci Rep 2024; 14:14187. [PMID: 38902328 PMCID: PMC11189906 DOI: 10.1038/s41598-024-64783-1] [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: 02/02/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024] Open
Abstract
Mononuclear phagocytes (MNP), including macrophages and dendritic cells form an essential component of primary responses to environmental hazards and toxic exposures. This is particularly important in disease conditions such as asthma and allergic airway disease, where many different cell types are present. In this study, we differentiated CD34+ haematopoietic stem cells towards different populations of MNP in an effort to understand how different cell subtypes present in inflammatory disease microenvironments respond to the common allergen house dust mite (HDM). Using single cell mRNA sequencing, we demonstrate that macrophage subtypes MCSPP1+ and MLCMARCO+ display different patterns of gene expression after HDM challenge, noted especially for the chemokines CXCL5, CXCL8, CCL5 and CCL15. MLCCD206Hi alternatively activated macrophages displayed the greatest changes in expression, while neutrophil and monocyte populations did not respond. Further work investigated how pollutant diesel exhaust particles could modify these transcriptional responses and revealed that CXC but not CC type chemokines were further upregulated. Through the use of diesel particles with adsorbed material removed, we suggest that soluble pollutants on these particles are the active constituents responsible for the modifying effects on HDM. This study highlights that environmental exposures may influence tissue responses dependent on which MNP cell type is present, and that these should be considerations when modelling such events in vitro. Understanding the nuanced responsiveness of different immune cell types to allergen and pollutant exposure also contributes to a better understanding of how these exposures influence the development and exacerbation of human disease.
Collapse
Affiliation(s)
- Leonie F H Fransen
- Toxicology Department, Radiation, Chemical and Environmental Hazards Directorate, UK Health Security Agency, Harwell Science and Innovation Campus, Harwell, OX11 0RQ, UK
| | - Martin O Leonard
- Toxicology Department, Radiation, Chemical and Environmental Hazards Directorate, UK Health Security Agency, Harwell Science and Innovation Campus, Harwell, OX11 0RQ, UK.
| |
Collapse
|
16
|
Singh D, Siddique HR. Epithelial-to-mesenchymal transition in cancer progression: unraveling the immunosuppressive module driving therapy resistance. Cancer Metastasis Rev 2024; 43:155-173. [PMID: 37775641 DOI: 10.1007/s10555-023-10141-y] [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: 06/14/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
Cancer cells undergo phenotypic switching (cancer cell plasticity) in response to microenvironmental cues, including exposure to therapy/treatment. Phenotypic plasticity enables the cancer cells to acquire more mesenchymal traits promoting cancer cells' growth, survival, therapy resistance, and disease recurrence. A significant program in cancer cell plasticity is epithelial-to-mesenchymal transition (EMT), wherein a comprehensive reprogramming of gene expression occurs to facilitate the translational shift from epithelial-to-mesenchymal phenotypes resulting in increased invasiveness and metastasis. In addition, EMT plays a pivotal role in facilitating cancer cells' escape from the body's immune system using several mechanisms, such as the downregulation of major histocompatibility complex-mediated antigen presentation, upregulation of immune checkpoint molecules, and recruitment of immune-suppressive cells. Cancer cells' ability to undergo phenotypic switching and EMT-driven immune escape presents a formidable obstacle in cancer management, highlighting the need to unravel the intricate mechanisms underlying these processes and develop novel therapeutic strategies. This article discusses the role of EMT in promoting immune evasion and therapy resistance. We also discuss the ongoing research on developing therapeutic approaches targeting intrinsic and induced cell plasticity within the immune suppressive microenvironment. We believe this review article will update the current research status and equip researchers, clinicians, and other healthcare professionals with valuable insights enhancing their existing knowledge and shedding light on promising directions for future cancer research. This will facilitate the development of innovative strategies for managing therapy-resistant cancers and improving patient outcomes.
Collapse
Affiliation(s)
- Deepti Singh
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India.
| |
Collapse
|
17
|
Fuchs AL, Costello SM, Schiller SM, Tripet BP, Copié V. Primary Human M2 Macrophage Subtypes Are Distinguishable by Aqueous Metabolite Profiles. Int J Mol Sci 2024; 25:2407. [PMID: 38397084 PMCID: PMC10889249 DOI: 10.3390/ijms25042407] [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: 01/01/2024] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
The complexity of macrophage (MΦ) plasticity and polarization states, which include classically activated pro-inflammatory (M1) and alternatively activated anti-inflammatory (M2) MΦ phenotypes, is becoming increasingly appreciated. Within the M2 MΦ polarization state, M2a, M2b, M2c, and M2d MΦ subcategories have been defined based on their expression of specific cell surface receptors, secreted cytokines, and specialized immune effector functions. The importance of immunometabolic networks in mediating the function and regulation of MΦ immune responses is also being increasingly recognized, although the exact mechanisms and extent of metabolic modulation of MΦ subtype phenotypes and functions remain incompletely understood. In this study, proton (1H) nuclear magnetic resonance (NMR) metabolomics was employed to determine the polar metabolomes of M2 MΦ subtypes and to investigate the relationship between aqueous metabolite profiles and M2 MΦ functional phenotypes. Results from this study demonstrate that M2a MΦs are most distinct from M2b, M2c, and M2d MΦ subtypes, and that M2b MΦs display several metabolic traits associated with an M1-like MΦ phenotype. The significance of metabolome differences for metabolites implicated in glycolysis, the tricarboxylic acid (TCA) cycle, phospholipid metabolism, and creatine-phosphocreatine cycling is discussed. Altogether, this study provides biochemical insights into the role of metabolism in mediating the specialized effector functions of distinct M2 MΦ subtypes and supports the concept of a continuum of macrophage activation states rather than two well-separated and functionally distinct M1/M2 MΦ classes, as originally proposed within a classical M1/M2 MΦ framework.
Collapse
Affiliation(s)
| | | | | | | | - Valérie Copié
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| |
Collapse
|
18
|
Mihaila AC, Ciortan L, Tucureanu MM, Simionescu M, Butoi E. Anti-Inflammatory Neutrophils Reprogram Macrophages toward a Pro-Healing Phenotype with Increased Efferocytosis Capacity. Cells 2024; 13:208. [PMID: 38334600 PMCID: PMC10854927 DOI: 10.3390/cells13030208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/11/2024] [Accepted: 01/20/2024] [Indexed: 02/10/2024] Open
Abstract
Following myocardial infarction (MI), blood neutrophils quickly and extensively infiltrate the heart, where they are temporally polarized into pro-inflammatory (N1) and anti-inflammatory (N2) subpopulations. Neutrophil transmigration is rapidly followed by the accrual of macrophages (MACs), which are believed to undergo local phenotypic transformations from pro-inflammatory to pro-healing MACs that mediate inflammation resolution. We hypothesized that N2 neutrophils can reprogram MACs toward a healing phenotype with increased efferocytosis capacity. To examine this, human neutrophils isolated from healthy subjects were polarized in N1 and N2 neutrophils, and their secretome was added to human MACs derived from THP monocytes. The impact of neutrophil factors on macrophages was investigated using qPCR, ELISA, Western blot, immunofluorescence, or an efferocytosis assay. The results show that the MACs exposed to N2 neutrophil secretome exhibited (i) increased expression of the anti-inflammatory molecules CD206, TGF-β, and IL-10 and the nuclear factors associated with reparatory macrophages (PPARγ, Nur77, and KLF4); (ii) enhanced expression of efferocytosis receptors (MerTK, CD36, CX3CR1, and integrins αv/β5) and of the bridge molecules Mfage8 and Gas6; and (iii) enhanced efferocytosis. In conclusion, factors released by N2 neutrophils induce a pro-healing phenotype of MACs by upregulating anti-inflammatory molecules and efferocytosis receptors and ensuing the efferocytosis capacity. The data suggest that molecular therapy to foster N2 polarization, which boosts macrophages' pro-healing phenotype, could be a promising strategy to speed up inflammation resolution and tissue repair.
Collapse
Affiliation(s)
| | | | | | | | - Elena Butoi
- Biopathology and Therapy of Inflammation, Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania; (A.C.M.); (L.C.); (M.S.)
| |
Collapse
|
19
|
Li C, Liu C, Zhang J, Lu Y, Jiang B, Xiong H, Li C. Pyruvate dehydrogenase kinase regulates macrophage polarization in metabolic and inflammatory diseases. Front Immunol 2023; 14:1296687. [PMID: 38193078 PMCID: PMC10773690 DOI: 10.3389/fimmu.2023.1296687] [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: 09/19/2023] [Accepted: 11/13/2023] [Indexed: 01/10/2024] Open
Abstract
Macrophages are highly heterogeneous and plastic, and have two main polarized phenotypes that are determined by their microenvironment, namely pro- and anti-inflammatory macrophages. Activation of pro-inflammatory macrophages is closely associated with metabolic reprogramming, especially that of aerobic glycolysis. Mitochondrial pyruvate dehydrogenase kinase (PDK) negatively regulates pyruvate dehydrogenase complex activity through reversible phosphorylation and further links glycolysis to the tricarboxylic acid cycle and ATP production. PDK is commonly associated with the metabolism and polarization of macrophages in metabolic and inflammatory diseases. This review examines the relationship between PDK and macrophage metabolism and discusses the mechanisms by which PDK regulates macrophage polarization, migration, and inflammatory cytokine secretion in metabolic and inflammatory diseases. Elucidating the relationships between the metabolism and polarization of macrophages under physiological and pathological conditions, as well as the regulatory pathways involved, may provide valuable insights into the etiology and treatment of macrophage-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Chenyu Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Chuanbin Liu
- Department of Pediatric Dentistry, Jining Stomatological Hospital, Jining, Shandong, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Yanyu Lu
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Bingtong Jiang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| | - Chunxia Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
20
|
Soliman AH, Youness RA, Sebak AA, Handoussa H. Phytochemical-derived tumor-associated macrophage remodeling strategy using Phoenix dactylifera L. boosted photodynamic therapy in melanoma via H19/iNOS/PD-L1 axis. Photodiagnosis Photodyn Ther 2023; 44:103792. [PMID: 37689125 DOI: 10.1016/j.pdpdt.2023.103792] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND The tumor microenvironment (TME) represents a barrier to PDT efficacy among melanoma patients. The aim of this study is to employ a novel muti-tactic TME-remodeling strategy via repolarization of tumor-associated macrophages (TAMs), the main TME immune cells in melanoma, from the pro-tumor M2 into the antitumor M1 phenotype using Phoenix dactylifera L. (date palm) in combination with PDT. METHODS Screening of different date cultivars was employed to choose extracts of selective toxicity to melanoma and TAMs, not normal macrophages. Potential extracts were then fractionated and characterized by gas chromatography-mass spectrometry (GC-MS). Finally, the efficacy and the potential molecular mechanism of the co-treatment were portrayed via quantitative real-time polymerase chain reaction (qRT-PCR) analysis. RESULTS Initial screening resulted in the selection of the two Phoenix dactylifera L. cultivars Safawi and Sukkari methanolic extracts. Sukkari showed superior capacity to revert TAM phenotype into M1 as well as more prominent upregulation of M1 markers and repression of melanoma immunosuppressive markers relative to positive control (resiquimod). Molecularly, it was shown that PDT of melanoma cells in the presence of the secretome of repolarized TAMs surpassed the monotherapy via the modulation of the H19/iNOS/PD-L1immune-regulatory axis. CONCLUSION This study highlights the potential utilization of nutraceuticals in combination with PDT in the treatment of melanoma to provide a dual activity through alleviating the immune suppressive TME and potentiating the anti-tumor responses.
Collapse
Affiliation(s)
- Aya H Soliman
- Department of Pharmaceutical Biology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, Main Entrance El Tagamoa El Khames, New Cairo 11511, Egypt.
| | - Rana A Youness
- Department of Pharmaceutical Biology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, Main Entrance El Tagamoa El Khames, New Cairo 11511, Egypt; Department of Biology and Biochemistry, Faculty of Biotechnology, German International University, New Administrative Capital, New Cairo 11835, Egypt
| | - Aya A Sebak
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11511, Egypt.
| | - Heba Handoussa
- Department of Pharmaceutical Biology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, Main Entrance El Tagamoa El Khames, New Cairo 11511, Egypt
| |
Collapse
|
21
|
Kobayashi Y, Arai H, Hamamoto Y, Yoshida K, Shimizu S, Yasuhara Y, Ichimaru N. High Infiltration of CD163-Positive Macrophages in Intratumor Compartment Predicts Poor Prognosis in Patients With Upper Urinary Tract Urothelial Carcinoma and Radical Nephroureterectomy. Clin Genitourin Cancer 2023; 21:e386-e393. [PMID: 37244798 DOI: 10.1016/j.clgc.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/29/2023]
Abstract
OBJECTIVES To investigate the prognostic value of CD68- and CD163-positive macrophages in patients with upper urinary tract urothelial carcinoma (UTUC). PATIENTS AND METHODS This retrospective study enrolled 50 patients (34 men and 16 women) with UTUC who received radical nephroureterectomy (RNU). We evaluated the expression of CD68 and CD163 in the intratumor compartment by immunohistochemistry. The Kaplan-Meier method and Cox proportional hazards regression model were used to evaluate overall survival (OS), cancer-specific survival (CSS), recurrence-free survival (RFS), and bladder recurrence-free survival (BRFS). RESULTS High infiltration of CD163-positive macrophages in patients with UTUC was significantly correlated with worse OS, CSS, and RFS (P < .05 for all). Multivariate analysis showed that high infiltration of CD163-positive macrophages was an independent negative prognostic factor of OS and CSS in patients with UTUC who received RNU. Lymphovascular invasion was an independent negative prognostic factor of RFS, and high infiltration of CD68-positive macrophages was an independent positive prognostic factor of BRFS. CONCLUSION This study indicated that high infiltration of CD163-positive macrophages in the intratumor compartment might be a useful prognostic marker for survival in patients with UTUC who receive RNU. Further, high infiltration of CD68-positive macrophages in the intratumoral compartment might be a useful prognostic marker for bladder recurrence in these patients.
Collapse
Affiliation(s)
- Yasuyuki Kobayashi
- Department of Urology, Kinki Central Hospital of Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan.
| | - Hiroki Arai
- Department of Urology, Kinki Central Hospital of Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan
| | - Yuichiro Hamamoto
- Department of Diagnostic Pathology, Kinki Central Hospital of Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan
| | - Kyotaro Yoshida
- Department of Clinical Laboratory, Kinki Central Hospital of Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan
| | - Shigeki Shimizu
- Department of Laboratory Medicine and Pathology, Kinki-Chuo Chest Medical Center, Sakai, Osaka, Japan
| | - Yumiko Yasuhara
- Department of Pathology, Sakai City Medical Center, Sakai, Osaka, Japan
| | - Naotsugu Ichimaru
- Department of Urology, Kinki Central Hospital of Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan
| |
Collapse
|
22
|
Hung CN, Chen M, DeArmond DT, Chiu CHL, Limboy CA, Tan X, Kusi M, Chou CW, Lin LL, Zhang Z, Wang CM, Chen CL, Mitsuya K, Osmulski PA, Gaczynska ME, Kirma NB, Vadlamudi RK, Gibbons DL, Warner S, Brenner AJ, Mahadevan D, Michalek JE, Huang THM, Taverna JA. AXL-initiated paracrine activation of pSTAT3 enhances mesenchymal and vasculogenic supportive features of tumor-associated macrophages. Cell Rep 2023; 42:113067. [PMID: 37659081 PMCID: PMC10577802 DOI: 10.1016/j.celrep.2023.113067] [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: 12/09/2022] [Revised: 07/14/2023] [Accepted: 08/18/2023] [Indexed: 09/04/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are integral to the development of complex tumor microenvironments (TMEs) and can execute disparate cellular programs in response to extracellular cues. However, upstream signaling processes underpinning this phenotypic plasticity remain to be elucidated. Here, we report that concordant AXL-STAT3 signaling in TAMs is triggered by lung cancer cells or cancer-associated fibroblasts in the cytokine milieu. This paracrine action drives TAM differentiation toward a tumor-promoting "M2-like" phenotype with upregulation of CD163 and putative mesenchymal markers, contributing to TAM heterogeneity and diverse cellular functions. One of the upregulated markers, CD44, mediated by AXL-IL-11-pSTAT3 signaling cascade, enhances macrophage ability to interact with endothelial cells and facilitate formation of primitive vascular networks. We also found that AXL-STAT3 inhibition can impede the recruitment of TAMs in a xenograft mouse model, thereby suppressing tumor growth. These findings suggest the potential application of AXL-STAT3-related markers to quantitatively assess metastatic potential and inform therapeutic strategies in lung cancer.
Collapse
Affiliation(s)
- Chia-Nung Hung
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Meizhen Chen
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Daniel T DeArmond
- Department of Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX, USA
| | - Cheryl H-L Chiu
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Catherine A Limboy
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Xi Tan
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Meena Kusi
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Chih-Wei Chou
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Li-Ling Lin
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Zhao Zhang
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Chiou-Miin Wang
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Chun-Liang Chen
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA; Office of Nursing Research & Scholarship, School of Nursing, University of Texas Health Science Center, San Antonio, TX, USA
| | - Kohzoh Mitsuya
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Pawel A Osmulski
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Maria E Gaczynska
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Nameer B Kirma
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Ratna K Vadlamudi
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Don L Gibbons
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Andrew J Brenner
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Division of Hematology and Oncology, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Daruka Mahadevan
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Division of Hematology and Oncology, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Joel E Michalek
- Department of Population Health Sciences, University of Texas Health Science Center, San Antonio, TX, USA
| | - Tim H-M Huang
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA; Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA.
| | - Josephine A Taverna
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX, USA; Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, USA; Division of Hematology and Oncology, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
23
|
Nakamura T, Fujiwara M. Comparative Immunohistochemical Analysis of Macrophage Phenotypes in Cutaneous Sarcoid Granuloma, Suture Granuloma, and Lipogranuloma. Am J Dermatopathol 2023; 45:371-377. [PMID: 37130221 DOI: 10.1097/dad.0000000000002446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
ABSTRACT Granulomas are composed of a heterogeneous population of resident and recruited macrophages according to the type of lesion, extent of injury, and local tissue environment (eg, involved site and interaction with infiltrating lymphocytes). Although macrophage phenotypes in various types of granulomas have been previously described, the experimental conditions varied across studies, precluding a comparative and comprehensive understanding of granulomas. This study was conducted to comparatively analyze the expression of markers of the M1 and M2 phenotypes in macrophages that compose various types of granulomas, including epithelioid lesions, under strict conditions. Surgical specimens of cutaneous sarcoidosis (11 lesions), suture granuloma (10 lesions), and subcutaneous lipogranuloma (12 lesions) were immunohistochemically stained for CD11c, CD206, CD163, and CD10. The expression of these markers in macrophages composing each type of granuloma was scored and statistically analyzed. Granuloma macrophages were mostly immunoreactive for CD11c and CD206 in all the examined cases, although many intermingling CD206-negative cells were observed in 5 cases of lipogranuloma. CD163 and CD10 were diffusely expressed in macrophages composing suture granuloma and lipogranuloma, whereas they were not expressed in epithelioid cells in cutaneous sarcoidosis. Meanwhile, "interstitial" macrophages around epithelioid granulomas revealed moderate to marked CD163 expression in 7 lesions of cutaneous sarcoidosis. These results indicate significant differences of expression of CD163 and CD10 between cutaneous sarcoidosis and suture granuloma/lipogranuloma; CD163 and CD10 are downregulated after the epithelioid transformation of macrophages in cutaneous sarcoidosis.
Collapse
|
24
|
Yang H, Chelariu-Raicu A, Makri D, Ori C, Ribeiro PCP, Kolben T, Kuhn C, Kessler M, Schulz C, Mahner S, Jeschke U, von Schönfeldt V. Updates of placental macrophages: Origins, molecular markers, functions, and related diseases. J Reprod Immunol 2023; 157:103942. [PMID: 36989681 DOI: 10.1016/j.jri.2023.103942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023]
Abstract
Placental macrophages are highly heterogeneous cells with differential phenotypes and functions defined by differential origins and modulated by the changing placental environment. During pregnancy, placental macrophages play a critical role in embryo implantation, placenta formation and homeostasis, fetal development and parturition. This review summarizes recent findings on the cellular origin of placental macrophages, and provide a comprehensive description of their phenotypes, corresponding molecular markers and functions in human placenta. Finally, alterations of placental macrophages in pregnancy-related diseases are discussed.
Collapse
|
25
|
Gui Y, Zeng Y, Chen B, Yang Y, Ma J, Li C. A smart pathogen detector engineered from intracellular hydrogelation of DNA-decorated macrophages. Nat Commun 2023; 14:2927. [PMID: 37217531 DOI: 10.1038/s41467-023-38733-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 05/12/2023] [Indexed: 05/24/2023] Open
Abstract
Bacterial infection is a major threat to global public health, which urgently requires useful tools to rapidly analyze pathogens in the early stages of infection. Herein, we develop a smart macrophage (Mø)-based bacteria detector, which can recognize, capture, enrich and detect different bacteria and their secreted exotoxins. We transform the fragile native Møs into robust gelated cell particles (GMøs) using photo-activated crosslinking chemistry, which retains membrane integrity and recognition capacity for different microbes. Meanwhile, these GMøs equipped with magnetic nanoparticles and DNA sensing elements can not only respond to an external magnet for facile bacteria collection, but allow the detection of multiple types of bacteria in a single assay. Additionally, we design a propidium iodide-based staining assay to rapidly detect pathogen-associated exotoxins at ultralow concentrations. Overall, these nanoengineered cell particles have broad applicability in the analysis of bacteria, and could potentially be used for the management and diagnosis of infectious diseases.
Collapse
Affiliation(s)
- Yueyue Gui
- School of Food and Biological Engineering, Hefei University of Technology, 230009, Hefei, P. R. China
| | - Yujing Zeng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, 210023, Nanjing, P. R. China
| | - Binrui Chen
- School of Food and Biological Engineering, Hefei University of Technology, 230009, Hefei, P. R. China
| | - Yueping Yang
- School of Food and Biological Engineering, Hefei University of Technology, 230009, Hefei, P. R. China
| | - Jiehua Ma
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, Shanghai, P. R. China
| | - Chao Li
- School of Food and Biological Engineering, Hefei University of Technology, 230009, Hefei, P. R. China.
| |
Collapse
|
26
|
Tada A, Minami T, Kitai H, Higashiguchi Y, Tokuda M, Higashiyama T, Negi Y, Horio D, Nakajima Y, Otsuki T, Mikami K, Takahashi R, Nakamura A, Kitajima K, Ohmuraya M, Kuribayashi K, Kijima T. Combination therapy with anti-programmed cell death 1 antibody plus angiokinase inhibitor exerts synergistic antitumor effect against malignant mesothelioma via tumor microenvironment modulation. Lung Cancer 2023; 180:107219. [PMID: 37146474 DOI: 10.1016/j.lungcan.2023.107219] [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: 03/16/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/07/2023]
Abstract
Malignant pleural mesothelioma (MPM) is an asbestos-related fatal malignant neoplasm. Although there has been no reliable chemotherapeutic regimen other than combination therapy of cisplatin and pemetrexed for two decades, combination of ipilimumab plus nivolumab brought about a better outcome in patients with MPM. Thus, cancer immunotherapy using immune checkpoint inhibitor (ICI) is expected to play a central role in the treatment of MPM. To maximize the antitumor effect of ICI, we evaluated whether nintedanib, an antiangiogenic agent, could augment the antitumor effect of anti-programmed cell death 1 (PD-1) antibody (Ab). Although nintedanib could not inhibit the proliferation of mesothelioma cells in vitro, it significantly suppressed the growth of mesothelioma allografts in mice. Moreover, combination therapy with anti-PD-1 Ab plus nintedanib reduced tumor burden more dramatically compared with nintedanib monotherapy via inducing remarkable necrosis in MPM allografts. Nintedanib did not promote the infiltration of CD8+ T cells within the tumor when used alone or in combination with anti-PD-1 Ab but it independently decreased the infiltration of tumor-associated macrophages (TAMs). Moreover, immunohistochemical analysis and ex vivo study using bone marrow-derived macrophages (BMDMs) showed that nintedanib could polarize TAMs from M2 to M1 phenotype. These results indicated that nintedanib had a potential to suppress protumor activity of TAMs both numerically and functionally. On the other hand, ex vivo study revealed that nintedanib upregulated the expression of PD-1 and PD-ligand 1 (PD-L1) in BMDMs and mesothelioma cells, respectively, and exhibited the impairment of phagocytic activity of BMDMs against mesothelioma cells. Co-administration of anti-PD-1 Ab may reactivate phagocytic activity of BMDMs by disrupting nintedanib-induced immunosuppressive signal via binding between PD-1 on BMDMs and PD-L1 on mesothelioma cells. Collectively, combination therapy of anti-PD-1 Ab plus nintedanib enhances the antitumor activity compared with respective monotherapy and can become a novel therapeutic option for patients with MPM.
Collapse
Affiliation(s)
- Akio Tada
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Toshiyuki Minami
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan; Department of Thoracic Oncology, Hyogo Medical University, Nishinomiya, Hyogo, Japan.
| | - Hidemi Kitai
- Department of Thoracic Oncology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Yoko Higashiguchi
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Mayuko Tokuda
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Tomoki Higashiyama
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Yoshiki Negi
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan; Department of Thoracic Oncology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Daisuke Horio
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan; Department of Thoracic Oncology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Yasuhiro Nakajima
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Taiichiro Otsuki
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan; Department of Thoracic Oncology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Koji Mikami
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan; Department of Thoracic Oncology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Ryo Takahashi
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan; Department of Thoracic Oncology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Akifumi Nakamura
- Department of Thoracic Surgery, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Kazuhiro Kitajima
- Department of Radiology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Masaki Ohmuraya
- Department of Genetics, Hyogo Medical University, Nishinomiya, Japan
| | - Kozo Kuribayashi
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan; Department of Thoracic Oncology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Takashi Kijima
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan; Department of Thoracic Oncology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
27
|
Feng L, Luo G, Li Y, Zhang C, Liu Y, Liu Y, Chen H, He D, Zhu Y, Gan L. Curcumin-dependent phenotypic transformation of microglia mediates resistance to pseudorabies-induced encephalitis. Vet Res 2023; 54:25. [PMID: 36918933 PMCID: PMC10015794 DOI: 10.1186/s13567-023-01149-x] [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: 10/19/2022] [Accepted: 01/10/2023] [Indexed: 03/15/2023] Open
Abstract
Pseudorabies virus (PRV) causes viral encephalitis, a devastating disease with high mortality worldwide. Curcumin (CUR) can reduce inflammatory damage by altering the phenotype of microglia; however, whether and how these changes mediate resistance to PRV-induced encephalitis is still unclear. In this study, BV2 cells were infected with/without PRV for 24 h and further treated with/without CUR for 24 h. The results indicated that CUR promoted the polarization of PRV-infected BV2 cells from the M1 phenotype to the M2 phenotype and reversed PRV-induced mitochondrial dysfunction. Furthermore, M1 BV2 cell secretions induced signalling pathways leading to apoptosis in PC-12 neuronal cells, and this effect was abrogated by the secretions of M2 BV2 cells. RNA sequencing and bioinformatics analysis predicted that this phenotypic shift may be due to changes in energy metabolism. Furthermore, Western blot analysis showed that CUR inhibited the increase in AMP-activated protein kinase (AMPK) phosphorylation, glycolysis, and triacylglycerol synthesis and the reduction in oxidative phosphorylation induced by PRV infection. Moreover, the ATP levels in M2 BV2 cells were higher than those in M1 cells. Furthermore, CUR prevented the increase in mortality, elevated body temperature, slowed growth, nervous system excitation, brain tissue congestion, vascular cuffing, and other symptoms of PRV-induced encephalitis in vivo. Thus, this study demonstrated that CUR protected against PRV-induced viral encephalitis by switching the phenotype of BV2 cells, thereby protecting neurons from inflammatory injury, and this effect was mediated by improving mitochondrial function and the AMPK/NF-κB p65-energy metabolism-related pathway.
Collapse
Affiliation(s)
- Luqiu Feng
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Guodong Luo
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Yuhang Li
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Chen Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Yuxuan Liu
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Yanqing Liu
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China
| | - Hongyue Chen
- Chongqing General Station of Animal Husbandry Technology Promotion, Chongqing, 401120, China
| | - Daoling He
- Chongqing General Station of Animal Husbandry Technology Promotion, Chongqing, 401120, China
| | - Yan Zhu
- Chongqing General Station of Animal Husbandry Technology Promotion, Chongqing, 401120, China
| | - Ling Gan
- College of Veterinary Medicine, Southwest University, Chongqing, 402460, China.
| |
Collapse
|
28
|
Tong P, Chen L, Sun X, Li H, Feng Y, Li J, Guan S. Surface modification of biodegradable magnesium alloy with poly (L-lactic acid) and sulfonated hyaluronic acid nanoparticles for cardiovascular application. Int J Biol Macromol 2023; 237:124191. [PMID: 36990404 DOI: 10.1016/j.ijbiomac.2023.124191] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/07/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
Magnesium (Mg) and its alloys have attracted extensive attention of researchers in the field of cardiovascular implants due to their good mechanical properties and biosafety. Constructing a multifunctional hybrid coating seems to be an effective strategy to address the insufficient endothelialization and poor corrosion resistance of Mg alloy vascular stents. In this study, a dense layer of magnesium fluoride (MgF2) was prepared on the surface of Mg alloy aiming at better corrosion resistance; Thereafter, sulfonated hyaluronic acid (S-HA) was made into small sized nanoparticles (NP) which were deposited on the MgF2 surface by self-assembly method, followed with poly-L-lactic acid (PLLA) coating preparation by one-step pulling method. The blood and cell tests showed that the composite coating had good blood compatibility, pro-endothelial, anti-hyperplasia and anti-inflammatory functions. Compared to current clinical PLLA@ Rapamycin coating, our PLLA/NP@S-HA coating showed better functions of promoting endothelial cells growth. These results strongly furnished a promising and feasible strategy for the surface modification of Mg-based degradable cardiovascular stents.
Collapse
Affiliation(s)
- Peiduo Tong
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold Technology (Ministry of Education), Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Lan Chen
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold Technology (Ministry of Education), Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Xiaojing Sun
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold Technology (Ministry of Education), Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Hang Li
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold Technology (Ministry of Education), Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Yashan Feng
- Zhengzhou Railway Vocational & Technical College, Zhengzhou 450000, China
| | - Jingan Li
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold Technology (Ministry of Education), Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China..
| | - Shaokang Guan
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold Technology (Ministry of Education), Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China..
| |
Collapse
|
29
|
Li T, Yan Z, Fan Y, Fan X, Li A, Qi Z, Zhang J. Cardiac repair after myocardial infarction: A two-sided role of inflammation-mediated. Front Cardiovasc Med 2023; 9:1077290. [PMID: 36698953 PMCID: PMC9868426 DOI: 10.3389/fcvm.2022.1077290] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Myocardial infarction is the leading cause of death and disability worldwide, and the development of new treatments can help reduce the size of myocardial infarction and prevent adverse cardiovascular events. Cardiac repair after myocardial infarction can effectively remove necrotic tissue, induce neovascularization, and ultimately replace granulation tissue. Cardiac inflammation is the primary determinant of whether beneficial cardiac repair occurs after myocardial infarction. Immune cells mediate inflammatory responses and play a dual role in injury and protection during cardiac repair. After myocardial infarction, genetic ablation or blocking of anti-inflammatory pathways is often harmful. However, enhancing endogenous anti-inflammatory pathways or blocking endogenous pro-inflammatory pathways may improve cardiac repair after myocardial infarction. A deficiency of neutrophils or monocytes does not improve overall cardiac function after myocardial infarction but worsens it and aggravates cardiac fibrosis. Several factors are critical in regulating inflammatory genes and immune cells' phenotypes, including DNA methylation, histone modifications, and non-coding RNAs. Therefore, strict control and timely suppression of the inflammatory response, finding a balance between inflammatory cells, preventing excessive tissue degradation, and avoiding infarct expansion can effectively reduce the occurrence of adverse cardiovascular events after myocardial infarction. This article reviews the involvement of neutrophils, monocytes, macrophages, and regulatory T cells in cardiac repair after myocardial infarction. After myocardial infarction, neutrophils are the first to be recruited to the damaged site to engulf necrotic cell debris and secrete chemokines that enhance monocyte recruitment. Monocytes then infiltrate the infarct site and differentiate into macrophages and they release proteases and cytokines that are harmful to surviving myocardial cells in the pre-infarct period. As time progresses, apoptotic neutrophils are cleared, the recruitment of anti-inflammatory monocyte subsets, the polarization of macrophages toward the repair phenotype, and infiltration of regulatory T cells, which secrete anti-inflammatory factors that stimulate angiogenesis and granulation tissue formation for cardiac repair. We also explored how epigenetic modifications regulate the phenotype of inflammatory genes and immune cells to promote cardiac repair after myocardial infarction. This paper also elucidates the roles of alarmin S100A8/A9, secreted frizzled-related protein 1, and podoplanin in the inflammatory response and cardiac repair after myocardial infarction.
Collapse
Affiliation(s)
- Tingting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhipeng Yan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yajie Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinbiao Fan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Aolin Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhongwen Qi
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Zhongwen Qi,
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China,Junping Zhang,
| |
Collapse
|
30
|
Henshaw J, Tremellen K. Intralipid infusion therapy as an adjunct treatment in women experiencing adenomyosis-related infertility. Ther Adv Reprod Health 2023; 17:26334941231181258. [PMID: 37362235 PMCID: PMC10288412 DOI: 10.1177/26334941231181258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
Background Currently, there is some evidence that adenomyosis patients using gonadotropin-releasing hormone (GnRH) agonist long downregulation (LDR) prior to embryo transfer may improve in vitro fertilization (IVF) success rate, but not to the baseline expected success where there is no adenomyosis. Given the association between adenomyosis and an aberrant endometrial immune environment, many physicians also use prednisolone or Intralipid adjuvant treatments in combination with GnRH agonist therapy, despite neither being of proven benefit. Objective The purpose of this study was to investigate whether the addition of prednisolone or Intralipid immune therapy to GnRH agonist LDR improves fertility outcomes in patients with adenomyosis. Methods This is a retrospective cohort study of 116 consecutive adenomyosis patients who underwent their first transfer of a genetically screened euploid embryo between January 2019 and December 2020 at a private IVF clinic. Results There was no difference in maternal age, body mass index, number of embryo's transferred and gravidity or parity among the three treatment groups. Patients who received Intralipid had a poorer prognosis with a longer duration of infertility (4 years) and a higher number of previous embryo transfers (ETs, 5 previous ETs) compared to the comparison groups. Logistic regression analysis adjustment for all covariates revealed that LDR plus Intralipid therapy produced significantly higher live birth rates (LBRs; 60%) compared to LDR alone (40% LBR); yet, the addition of prednisolone to GnRH agonist LDR (30% LBR) provided no additional live birth benefit. Conclusion In this retrospective analysis, we showed Intralipid adjuvant treatment in combination with GnRH agonist therapy in adenomyosis patients undergoing IVF resulted in a LBR expected in women without adenomyosis using preimplantation genetic testing screened embryos. This benefit was not seen when using prednisolone as an adjuvant to GnRH agonist LDR. Future randomized clinical trials will be required to confirm the therapeutic benefit of Intralipid in combination with GnRH agonist therapy.
Collapse
Affiliation(s)
- James Henshaw
- Department of Obstetrics and Gynaecology, Royal Hospital for Women, Randwick, NSW, Australia
| | - Kelton Tremellen
- Department of Obstetrics Gynaecology and Reproductive Medicine, Flinders University, Sturt Rd, Bedford Park, SA 5042, Australia
- Repromed, Dulwich, SA, Australia
| |
Collapse
|
31
|
Tim-3: An inhibitory immune checkpoint is associated with maternal-fetal tolerance and recurrent spontaneous abortion. Clin Immunol 2022; 245:109185. [DOI: 10.1016/j.clim.2022.109185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/27/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
|
32
|
Emerging Effects of IL-33 on COVID-19. Int J Mol Sci 2022; 23:ijms232113656. [PMID: 36362440 PMCID: PMC9658128 DOI: 10.3390/ijms232113656] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Since the start of COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), more than 6 million people have lost their lives worldwide directly or indirectly. Despite intensified efforts to clarify the immunopathology of COVID-19, the key factors and processes that trigger an inflammatory storm and lead to severe clinical outcomes in patients remain unclear. As an inflammatory storm factor, IL-33 is an alarmin cytokine, which plays an important role in cell damage or infection. Recent studies have shown that serum IL-33 is upregulated in COVID-19 patients and is strongly associated with poor outcomes. Increased IL-33 levels in severe infections may result from an inflammatory storm caused by strong interactions between activated immune cells. However, the effects of IL-33 in COVID-19 and the underlying mechanisms remain to be fully elucidated. In this review, we systematically discuss the biological properties of IL-33 under pathophysiological conditions and its regulation of immune cells, including neutrophils, innate lymphocytes (ILCs), dendritic cells, macrophages, CD4+ T cells, Th17/Treg cells, and CD8+ T cells, in COVID-19 phagocytosis. The aim of this review is to explore the potential value of the IL-33/immune cell pathway as a new target for early diagnosis, monitoring of severe cases, and clinical treatment of COVID-19.
Collapse
|
33
|
He TR, Tang XY, Yan Q, Wu XY, Shi B, Lin Y. All-trans Retinoic Acid-incorporated Glycol Chitosan Nanoparticles Regulate Macrophage Polarization in Pg-LPS-Induced Inflammation. Curr Med Sci 2022; 42:974-980. [PMID: 36245026 DOI: 10.1007/s11596-022-2602-8] [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: 03/22/2021] [Accepted: 10/27/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE The occurrence and development of inflammation are closely correlated to the polarization of macrophages. All-trans retinoic acid (ATRA) has been proven to promote the polarization of macrophages from M1 to M2, but this lacks an effective carrier to participate in the biological response. The present study aims to determine whether retinoic acid-incorporated glycol chitosan (RA-GC) nanoparticles can regulate macrophage polarization in Porphyromonas gingivalis-lipopolysaccharide (Pg-LPS)-induced inflammation. METHODS Mouse 264.7 cell lines were treated with 1 µg/mL Pg-LPS to induce inflammation. After the effects of ATRA and RA-GC on the activity of macrophages were detected by CCK-8 assay, cells induced with Pg-LPS were assigned to the blank control group (GC) nanoparticles without ATRA, and experimental groups (GC nanoparticles loaded with different concentrations of ATRA: 1, 10 and 100 µg/mL). The effects of RA-GC on inflammatory cytokines tumor necrosis factor-α, interleukin (IL)-10 and IL-12 in macrophages were detected by enzyme-linked immunosorbent assay (ELISA). Subsequently, the effects of GC nanoparticles loaded with/without ATRA on macrophage polarization in an inflammatory environment were detected by RT-PCR and Western blotting. RESULTS The results revealed that RA-GC had no significant effect on macrophage activity. However, RA-GC could effectively inhibit the Pg-LPS-induced inflammatory factor expression in macrophages. Meanwhile, the experimental results confirmed that RA-GC could downregulate the expression of inducible nitric oxide synthase (iNOS) (a marker of M1 macrophages) and upregulate the expression of mannose receptor and Arginase-1 (a marker of M2 macrophages) in a dose-dependent manner. CONCLUSION The present study confirms that RA-GC can promote the M2 polarization of macrophages in an inflammatory environment, and proposes this as a promising target for the clinical treatment of Pg-LPS-related diseases.
Collapse
Affiliation(s)
- Tian-Rong He
- Fujian Provincial Hospital, Shengli Clinical College of Fujian Medical University, Fuzhou, 350001, China
| | - Xiu-Yu Tang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedical Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430075, China
| | - Qi Yan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedical Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430075, China
| | - Xin-Yu Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedical Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430075, China
| | - Bin Shi
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory for Oral Biomedical Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430075, China.
| | - Yi Lin
- Fujian Provincial Hospital, Shengli Clinical College of Fujian Medical University, Fuzhou, 350001, China.
| |
Collapse
|
34
|
Rui Y, Han X, Jiang A, Hu J, Li M, Liu B, Qian F, Huang L. Eucalyptol prevents bleomycin-induced pulmonary fibrosis and M2 macrophage polarization. Eur J Pharmacol 2022; 931:175184. [PMID: 35964659 DOI: 10.1016/j.ejphar.2022.175184] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/23/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrosing interstitial pneumonia with limited therapeutic options. Eucalyptol, a terpenoid oxide isolated from eucalyptus species, reportedly exhibits various biological activities such as anti-inflammatory and antioxidant effects. In the present study, we aimed to determine whether eucalyptol could alleviate bleomycin (BLM)-induced pulmonary fibrosis and inhibit interleukin (IL)-13-induced M2 macrophage polarization. Upon treatment with eucalyptol, BLM-induced pulmonary fibrosis and lung inflammation were significantly reduced. The pulmonary neutrophil accumulation and pulmonary permeability were inhibited and the expression of hydroxyproline, alpha-smooth muscle actin, and fibronectin was significantly down-regulated. Eucalyptol also markedly inhibited the expression of arginase-1, Ym-1, IL-13, and transforming growth factor (TGF)-β1, reduced the production of IL-13, IL-6, tumor necrosis factor (TNF)-α, and attenuated the activity of TGF-β1 in bronchoalveolar lavage fluid (BALF). Furthermore, the in vitro assay revealed that eucalyptol disturbed M2 macrophage polarization and reduced the macrophage-mediated secretion of the profibrotic factor TGF-β1. Eucalyptol inhibited the nuclear location of signal transducer and activator of transcription 6 (STAT6) and the phosphorylation of STAT6 and p38 mitogen-activated protein kinase (p38 MAPK), and reduced the expression of their downstream transcription factors, krupple-like factor 4 (KLF4) and peroxisome proliferator-activated receptor gamma (PPAR-γ). These findings indicated that eucalyptol alleviates BLM-induced pulmonary fibrosis by regulating M2 macrophage polarization, which, in turn, inhibits the activation of signaling molecules (e.g., STAT6 and p38 MAPK) and the expression of transcription factors (e.g., KLF4 and PPAR-γ). Thus, eucalyptol might be a potential therapeutic agent for IPF.
Collapse
Affiliation(s)
- Yan Rui
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Xiaojing Han
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Anbang Jiang
- Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Junfeng Hu
- Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Miao Li
- Department of General Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Bangzhu Liu
- Department of Respiratory Medicine, The Second People's Hospital of Anhui, Wuhu, Anhui, 233000, China
| | - Feng Qian
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui, 233000, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Linian Huang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China.
| |
Collapse
|
35
|
The Regulating Effect of CII-3 and Its Active Components from Periplaneta americana on M1/M2 Macrophage Polarization. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27144416. [PMID: 35889289 PMCID: PMC9323847 DOI: 10.3390/molecules27144416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/26/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022]
Abstract
CII-3 is the effective part of Periplaneta americana for application in oncotherapy. This study investigated its main chemical components for macrophage polarization regulation activity. Compounds were separated and purified, and their structures were elucidated based on NMR and HR-ESI-MS analyses. After inducing the M1 and M2 phenotype macrophages, CII-3 and testing components were added and co-incubated to evaluate their effects on the relevant markers of macrophages. Then, gradient concentrations of CII-3 and active monomers were further investigated for their effects on M2 macrophages. The effects were detected by RT-PCR, ELISA, flow cytometry, and immunofluorescence. Twelve compounds were identified from CII-3. CII-3 and pericanaside (5) had no obvious effect on M1 macrophages, while they significantly reduced the expression levels of M2 macrophage markers. Specifically, they significantly reduced the levels of TGF-β and IL-10 and the mRNA expression levels of ARG-1 and CD206 in the M2 phenotypes of RAW264.7 and Ana-1 macrophages. The conditioned medium of CII-3 and pericanaside (5) could inhibit the migration capacity of CT26.WT tumor cells. Macrophage M1/M2 polarization is a dynamic equilibrium, and the M2 phenotype, which can promote the growth of tumor cells, is relatively highly expressed in the tumor microenvironment. CII-3 and pericanaside could significantly reduce the phenotype of M2-type macrophages, indicating that the anti-tumor activity of CII-3 could be related to the inhibitory effect on M2 polarization, and pericanaside was one of the active components.
Collapse
|
36
|
Klimov V, Cherevko N, Klimov A, Novikov P. Neuronal-Immune Cell Units in Allergic Inflammation in the Nose. Int J Mol Sci 2022; 23:6938. [PMID: 35805946 PMCID: PMC9266453 DOI: 10.3390/ijms23136938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022] Open
Abstract
Immune cells and immune-derived molecules, endocrine glands and hormones, the nervous system and neuro molecules form the combined tridirectional neuroimmune network, which plays a significant role in the communication pathways and regulation at the level of the whole organism and local levels, in both healthy persons and patients with allergic rhinitis based on an allergic inflammatory process. This review focuses on a new research paradigm devoted to neuronal-immune cell units, which are involved in allergic inflammation in the nose and neuroimmune control of the nasal mucociliary immunologically active epithelial barrier. The categorization, cellular sources of neurotransmitters and neuropeptides, and their prevalent profiles in constituting allergen tolerance maintenance or its breakdown are discussed. Novel data on the functional structure of the nasal epithelium based on a transcriptomic technology, single-cell RNA-sequencing results, are considered in terms of neuroimmune regulation. Notably, the research of pathogenesis and therapy for atopic allergic diseases, including recently identified local forms, from the viewpoint of the tridirectional interaction of the neuroimmune network and discrete neuronal-immune cell units is at the cutting-edge.
Collapse
Affiliation(s)
- Vladimir Klimov
- Immunology & Allergy Dept, Siberian State Medical University, 634041 Tomsk, Russia; (N.C.); (A.K.); (P.N.)
| | - Natalia Cherevko
- Immunology & Allergy Dept, Siberian State Medical University, 634041 Tomsk, Russia; (N.C.); (A.K.); (P.N.)
| | - Andrew Klimov
- Immunology & Allergy Dept, Siberian State Medical University, 634041 Tomsk, Russia; (N.C.); (A.K.); (P.N.)
| | - Pavel Novikov
- Immunology & Allergy Dept, Siberian State Medical University, 634041 Tomsk, Russia; (N.C.); (A.K.); (P.N.)
- Medical Association “Center for Family Medicine”, 634050 Tomsk, Russia
| |
Collapse
|
37
|
Ma D, Zhou X, Wang Y, Dai L, Yuan J, Peng J, Zhang X, Wang C. Changes in the Small Noncoding RNAome During M1 and M2 Macrophage Polarization. Front Immunol 2022; 13:799733. [PMID: 35619693 PMCID: PMC9127141 DOI: 10.3389/fimmu.2022.799733] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/01/2022] [Indexed: 12/14/2022] Open
Abstract
Macrophages belong to a special phagocytic subgroup of human leukocytes and are one of the important cells of the human immune system. Small noncoding RNAs are a group of small RNA molecules that can be transcribed without the ability to encode proteins but could play a specific function in cells. SncRNAs mainly include microRNAs (miRNAs) and piwi-interacting RNAs (piRNAs), small nucleolar RNAs (snoRNAs), small nuclear RNAs (snRNAs) and repeat RNAs. We used high-throughput sequencing analysis and qPCR to detect the expression changes of the small noncoding RNAome during macrophage polarization. Our results showed that 84 miRNAs and 47 miRNAs with were downregulated during M1 macrophage polarization and that 11 miRNAs were upregulated and 19 miRNAs were downregulated during M2 macrophage polarization. MiR-novel-3-nature and miR-27b-5p could promote expression of TNF-α which was marker gene of M1 macrophages. The piRNA analysis results showed that 69 piRNAs were upregulated and 61 piRNAs were downregulated during M1 macrophage polarization and that 3 piRNAs were upregulated and 10 piRNAs were downregulated during M2 macrophage polarization. DQ551351 and DQ551308 could promote the mRNA expression of TNF-α and DQ551351overexpression promoted the antitumor activity of M1 macrophages. SnoRNA results showed that 62 snoRNAs were upregulated and 59 snoRNAs were downregulated during M1 macrophage polarization, whereas 6 snoRNAs were upregulated and 10 snoRNAs were downregulated during M2 macrophage polarization. Overexpression of snoRNA ENSMUST00000158683.2 could inhibit expression of TNF-α. For snRNA, we found that 12 snRNAs were upregulated and 15 snRNAs were downregulated during M1 macrophage polarization and that 2 snRNAs were upregulated during M2 macrophage polarization. ENSMUSG00000096786 could promote expression of IL-1 and iNOS and ENSMUSG00000096786 overexpression promoted the antitumor activity of M1 macrophages. Analysis of repeat RNAs showed that 7 repeat RNAs were upregulated and 9 repeat RNAs were downregulated during M1 macrophage polarization and that 2 repeat RNAs were downregulated during M2 macrophage polarization. We first reported the expression changes of piRNA, snoRNA, snRNA and repeat RNA during macrophage polarization, and preliminarily confirmed that piRNA, snoRNA and snRNA can regulate the function of macrophages.
Collapse
Affiliation(s)
- Ding Ma
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Xing Zhou
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, China
| | - Yu Wang
- Department of Cardiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Liming Dai
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Jie Yuan
- Department of Orthopaedic Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jianping Peng
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Chuandong Wang
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| |
Collapse
|
38
|
Li T, Zhang YS, Wan M, Wu W, Yao YF, Li WJ. Ganoderma atrum polysaccharide modulates the M1/M2 polarization of macrophages linked to the Notch signaling pathway. Food Funct 2022; 13:4216-4228. [PMID: 35332895 DOI: 10.1039/d1fo04309a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Macrophages could be polarized into two major sub-populations including classically activated (M1) and alternatively activated (M2) macrophages. The present study aimed to investigate the effects of Ganoderma atrum polysaccharide (PSG-1) on the regulation of macrophage polarization and further explored the associated molecular mechanisms. In this work, a lipopolysaccharide (LPS) plus IFN-γ and IL-4 were used to establish an in vitro model of two extreme states, namely pro-inflammatory M1 and anti-inflammatory M2. The results showed that PSG-1 had effects on the behavior modification of macrophage polarization by reducing CD80 expression in LPS plus IFN-γ-induced M1 macrophages, and attenuating CD23 expression in IL-4-induced M2 macrophages. Further study revealed that PSG-1-modulated M1 and M2 macrophage polarization was associated with controlling phagocytosis, reactive oxygen species generation, NO and cytokines (IL-1β, IL-6 and IL-10). Subsequently, the treatment of M1 macrophages with a combination of PSG-1 and a Notch-response inhibitor (DAPT) did not alter CD80 expression compared with DAPT alone, while several pro-inflammatory parameters were considerably decreased, suggesting that the Notch signaling pathway partly mediated the effects of PSG-1 on modulating macrophage polarization. Together, our findings suggested that PSG-1 could repair the chaos in the polarization of M1/M2 macrophages and the molecular mechanism linked to the Notch signaling pathway.
Collapse
Affiliation(s)
- Teng Li
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Yan-Song Zhang
- Research Institute of Food Science & Engineering Technology, Hezhou University, Hezhou 542899, China
| | - Min Wan
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Wei Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| | - Yu-Fei Yao
- Department of Critical Care Medicine, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China.
| | - Wen-Juan Li
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| |
Collapse
|
39
|
Activation of the Innate Immune Checkpoint CLEC5A on Myeloid Cells in the Absence of Danger Signals Modulates Macrophages’ Function but Does Not Trigger the Adaptive T Cell Immune Response. J Immunol Res 2022; 2022:9926305. [PMID: 35252461 PMCID: PMC8896916 DOI: 10.1155/2022/9926305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 11/30/2021] [Accepted: 02/09/2022] [Indexed: 12/19/2022] Open
Abstract
C-Type lectin receptor 5A (CLEC5A) is a spleen tyrosine kinase- (Syk-) coupled pattern recognition receptor expressed on myeloid cells and involved in the innate immune response to viral and bacterial infections. Activation of the CLEC5A receptor with pathogen-derived antigens leads to a secretion of proinflammatory mediators such as TNF-α and IL-6 that may provoke a systemic cytokine storm, and CLEC5A gene polymorphisms are associated with the severity of DV infection. In addition, the CLEC5A receptor was mentioned in the context of noninfectious disorders like chronic obstructive pulmonary disease (COPD) or arthritis. Altogether, CLEC5A may be considered as an innate immune checkpoint capable to amplify proinflammatory signals, and this way contributes to infection or to aseptic inflammation. In this study, we determined CLEC5A receptor expression on different macrophage subsets (in vitro and ex vivo) and the functional consequences of its activation in aseptic conditions. The CLEC5A surface expression appeared the highest on proinflammatory M1 macrophages while intermediate on tumor-associated phenotypes (M2c or TAM). In contrast, the CLEC5A expression on ex vivo-derived alveolar macrophages from healthy donors or macrophages from ovarian cancer patients was hardly detectable. Targeting CLEC5A on noninflammatory macrophages with an agonistic α-CLEC5A antibody triggered a release of proinflammatory cytokines, resembling a response to dengue virus, and led to phenotypic changes in myeloid cells that may suggest their reprogramming towards a proinflammatory phenotype, e.g., upregulation of CD80 and downregulation of CD163. Interestingly, the CLEC5A agonist upregulated immune-regulatory molecules like CD206, PD-L1, and cytokines like IL-10, macrophage-derived chemokine (MDC/CCL22), and thymus and activation chemokine (TARC/CCL17) which are associated with an anti-inflammatory or a protumorigenic macrophage phenotype. In the absence of concomitant pathogenic or endogenous danger signals, the CLEC5A receptor activation did not amplify an autologous T cell response, which may represent a protective innate mechanism to avoid an undesirable autoimmune adaptive response.
Collapse
|
40
|
Ahamada MM, Jia Y, Wu X. Macrophage Polarization and Plasticity in Systemic Lupus Erythematosus. Front Immunol 2022; 12:734008. [PMID: 34987500 PMCID: PMC8721097 DOI: 10.3389/fimmu.2021.734008] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that attacks almost every organ. The condition mostly happens to adults but is also found in children, and the latter have the most severe manifestations. Among adults, females, especially non-Caucasian, are mostly affected. Even if the etiology of SLE remains unclear, studies show a close relation between this disease and both genetics and environment. Despite the large number of published articles about SLE, we still do not have a clear picture of its pathogenesis, and no specific drug has been found to treat this condition effectively. The implication of macrophages in SLE development is gaining ground, and studying it could answer these gaps. Indeed, both in vivo and in vitro studies increasingly report a strong link between this disease and macrophages. Hence, this review aims to explore the role of macrophages polarization and plasticity in SLE development. Understanding this role is of paramount importance because in-depth knowledge of the connection between macrophages and this systemic disease could clarify its pathogenesis and provide a foundation for macrophage-centered therapeutic approaches.
Collapse
Affiliation(s)
- Mariame Mohamed Ahamada
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yang Jia
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaochuan Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
41
|
Chen H, Agrawal DK, Thankam FG. Biomaterials-Driven Sterile Inflammation. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:22-34. [PMID: 33213285 PMCID: PMC8892963 DOI: 10.1089/ten.teb.2020.0253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Performance of the biomaterials used for regenerative medicine largely depends on biocompatibility; however, the biological mechanisms underlying biocompatibility of a biomaterial within the host system is poorly understood. In addition to the classical immune response against non-self-entities, the sterile inflammatory response could limit the compatibility of biological scaffolds. Whereas the immediate to short-term host response to a biomaterial implant have been characterized, the long-term progression of host-biomaterial relationship has not been described. This article explores the novel concept of biomaterials-driven sterile inflammation (BSI) in long-term biodegradable implants and throws light for possible explanation for the onset of BSI and the associated damage-associated molecular patterns. The understanding of BSI would advance the current strategies to improve biomaterial-host tissue integration and open novel translational avenues in biomaterials-based tissue regeneration. Impact statement Understanding the novel concept of biomaterials-driven sterile inflammation and associated damage-associated molecular patterns in long-term biodegradable implants would determine their success and improves the tissue engineering and regenerative strategies.
Collapse
Affiliation(s)
- Henry Chen
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - Devendra K. Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - Finosh G. Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
- Address correspondence to: Finosh G. Thankam, PhD, Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA 91766-1854, USA
| |
Collapse
|
42
|
Kuntzel T, Bagnard D. Manipulating Macrophage/Microglia Polarization to Treat Glioblastoma or Multiple Sclerosis. Pharmaceutics 2022; 14:344. [PMID: 35214076 PMCID: PMC8877500 DOI: 10.3390/pharmaceutics14020344] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Macrophages and microglia are implicated in several diseases with divergent roles in physiopathology. This discrepancy can be explained by their capacity to endorse different polarization states. Theoretical extremes of these states are called M1 and M2. M1 are pro-inflammatory, microbicidal, and cytotoxic whereas M2 are anti-inflammatory, immunoregulatory cells in favor of tumor progression. In pathological states, these polarizations are dysregulated, thus restoring phenotypes could be an interesting treatment approach against diseases. In this review, we will focus on compounds targeting macrophages and microglia polarization in two very distinctive pathologies: multiple sclerosis and glioblastoma. Multiple sclerosis is an inflammatory disease characterized by demyelination and axon degradation. In this case, macrophages and microglia endorse a M1-like phenotype inducing inflammation. Promoting the opposite M2-like polarization could be an interesting treatment strategy. Glioblastoma is a brain tumor in which macrophages and microglia facilitate tumor progression, spreading, and angiogenesis. They are part of the tumor associated macrophages displaying an anti-inflammatory phenotype, thereby inhibiting anti-tumoral immunity. Re-activating them could be a method to limit and reduce tumor progression. These two pathologies will be used to exemplify that targeting the polarization of macrophages and microglia is a promising approach with a broad spectrum of applications deserving more attention.
Collapse
Affiliation(s)
- Thomas Kuntzel
- UMR7242 Biotechnology and Cell Signaling, Centre National de la Recherche Scientifique, Strasbourg Drug Discovery and Development Institute (IMS), University of Strasbourg, 67400 Illkirch-Graffenstaden, France;
| | - Dominique Bagnard
- UMR7242 Biotechnology and Cell Signaling, Centre National de la Recherche Scientifique, Strasbourg Drug Discovery and Development Institute (IMS), University of Strasbourg, 67400 Illkirch-Graffenstaden, France;
- Ecole Supérieure de Biotechnologie de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
43
|
Cutolo M, Soldano S, Gotelli E, Montagna P, Campitiello R, Paolino S, Pizzorni C, Sulli A, Smith V, Tardito S. CTLA4-Ig treatment induces M1-M2 shift in cultured monocyte-derived macrophages from healthy subjects and rheumatoid arthritis patients. Arthritis Res Ther 2021; 23:306. [PMID: 34952630 PMCID: PMC8709961 DOI: 10.1186/s13075-021-02691-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/03/2021] [Indexed: 12/29/2022] Open
Abstract
Background In rheumatoid arthritis (RA), macrophages play an important role in modulating the immunoinflammatory response through their polarisation into “classically” (M1) or “alternatively activated” (M2) phenotypes. In RA, CTLA4-Ig (abatacept) reduces the inflammatory activity of macrophages by interacting with the costimulatory molecule CD86. The study aimed to investigate the efficacy of CTLA4-Ig treatment to induce an M2 phenotype both in M1-polarised monocyte-derived macrophages (MDMs) obtained from healthy subjects (HS) and in cultured MDMs obtained from active RA patients. Methods Cultured MDMs were obtained from peripheral blood mononuclear cells of 7 active RA patients and from 10 HS after stimulation with phorbol myristate acetate (5 ng/mL) for 24 h. HS-MDMs were then stimulated with lipopolysaccharide (LPS, 1 mg/mL) for 4 h to induce M1-MDMs. M1-MDMs and RA-MDMs were treated with CTLA4-Ig (100 μM and 500 μM) for 3, 12, 24, and 48 h. The gene expression of CD80, CD86, and TLR4 (M1 markers); CD163, CD204, and CD206 (surface M2 markers); and MerTK (functional M2 marker) was evaluated by qRT-PCR. The protein synthesis of surface M2 markers was investigated by Western blotting. The statistical analysis was performed by the Wilcoxon t-test. Results In LPS-induced HS-M1-MDMs, CTLA4-Ig 100 μM and 500 μM significantly downregulated the gene expression of M1 markers (3 h p<0.01 for all molecules; 12 h p<0.05 for TLR4 and CD86) and significantly upregulated that of M2 markers, primarily after 12 h of treatment (CD163: p < 0.01 and p < 0.05; CD206: p < 0.05 and p < 0.01; CD204: p < 0.05 by 100 mg/mL). Moreover, in these cells, CTLA4-Ig 500 μM increased the protein synthesis of surface M2 markers (p < 0.05). Similarly, in RA-MDMs, the CTLA4-Ig treatment significantly downregulated the gene expression of M1 markers at both concentrations primarily after 12 h (p < 0.05). Furthermore, both concentrations of CTLA4-Ig significantly upregulated the gene expression of CD206 (after 3 h of treatment; p < 0.05), CD163, and MerTK (after 12 h of treatment, p < 0.05), whereas CD204 gene expression was significantly upregulated by the high concentration of CTLA4-Ig (p < 0.05). The protein synthesis of all surface markers was increased primarily by CTLA4-Ig 500 μM, significantly for CD204 and CD206 after 24 h of treatment (p < 0.05). Conclusions CTLA4-Ig treatment seems to induce the in vitro shift from M1 to M2 macrophages, of both HS-M1-MDMs and RA-MDMs, as observed by the significant downregulation exerted on selected M1 markers and the upregulation of selected M2 markers suggesting an additional mechanism for its modulation of the RA inflammatory process. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02691-9.
Collapse
Affiliation(s)
- Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy.
| | - Stefano Soldano
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Emanuele Gotelli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Paola Montagna
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Rosanna Campitiello
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Sabrina Paolino
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Carmen Pizzorni
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Alberto Sulli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| | - Vanessa Smith
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium.,Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Center (IRC), Ghent, Belgium
| | - Samuele Tardito
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine, IRCCS San Martino Polyclinic Hospital, University of Genova, Genoa, Italy
| |
Collapse
|
44
|
Ma X, Gao Y, Chen Y, Liu J, Yang C, Bao C, Wang Y, Feng Y, Song X, Qiao S. M2-Type Macrophages Induce Tregs Generation by Activating the TGF-β/Smad Signalling Pathway to Promote Colorectal Cancer Development. Onco Targets Ther 2021; 14:5391-5402. [PMID: 34908844 PMCID: PMC8665883 DOI: 10.2147/ott.s336548] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose The increase of both M2-type macrophages and Tregs is closely associated with the development of colorectal cancer. However, the mechanism of their interaction is still unclear. In this study, we investigated the correlation of M2-type macrophages with Tregs and the possible mechanisms between them. Methods Using immunohistochemistry, we analysed Smad3 (a key protein in the TGF-β/Smad signalling pathway) expression in colorectal cells, as well as infiltrating numbers of CD163 (a marker for M2-type macrophages), Foxp3 (a marker for Tregs) in 250 surgically resected colorectal cancer tissues, matched normal and paracancerous tissues. The relation of CD163 and Foxp3 was investigated in CRC with clinicopathological characteristics and preoperative tumour markers. Results CD163, Foxp3 and Smad3 were upregulated in CRC tissues compared to matched normal and paracancerous tissues. Interestingly, CD163 and Foxp3 were significantly positively correlated in CRC, and both were significantly positively correlated with Smad3. Both CD163 and Foxp3 were upregulated with increasing tumour TNM staging, increasing number of lymph node metastases and increasing vascular invasion. Additionally, CD163 was upregulated with increasing depth of infiltration. The number of M2-type macrophages and the expression levels of preoperative CEA, CA19-9 and CA72-4 were significantly positively correlated. The number of Tregs was significantly positively correlated with the expression levels of preoperative CEA and CA19-9. Conclusion M2-type macrophages may induce Tregs generation through activation of the TGF-β/Smad signalling pathway, which can promote the development of colorectal cancer.
Collapse
Affiliation(s)
- Xueqian Ma
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, People's Republic of China
| | - Yu Gao
- Computer Teaching and Research Section, Jinzhou Medical University, Jinzhou, 121000, Liaoning, People's Republic of China
| | - Yanlei Chen
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, People's Republic of China
| | - Jinhao Liu
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, People's Republic of China
| | - Chunyu Yang
- Department of Pathology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, People's Republic of China
| | - Cuifen Bao
- Basic Medical Experimental Teaching Center, Jinzhou Medical University, Jinzhou, 121000, Liaoning, People's Republic of China
| | - Yanping Wang
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, People's Republic of China
| | - Yang Feng
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, People's Republic of China
| | - Xiaoyu Song
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, People's Republic of China
| | - Shifeng Qiao
- The Second Ward of Colorectal Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, People's Republic of China
| |
Collapse
|
45
|
Tian X, Aiyer KTS, Kapsenberg JM, Roelen DL, van der Hoorn ML, Eikmans M. Uncomplicated oocyte donation pregnancies display an elevated CD163-positive type 2 macrophage load in the decidua, which is associated with fetal-maternal HLA mismatches. Am J Reprod Immunol 2021; 87:e13511. [PMID: 34738274 PMCID: PMC9286476 DOI: 10.1111/aji.13511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/07/2021] [Accepted: 11/01/2021] [Indexed: 11/27/2022] Open
Abstract
PROBLEM The embryo of an oocyte donation (OD) pregnancy is completely allogeneic to the mother, which may challenge the maternal immune system to tolerize the fetus. Decidual macrophages are essential in maintaining a healthy pregnancy, and type 2 macrophages may exhibit immune suppressive activity. We hypothesized that the composition of decidual macrophages is different between uncomplicated OD pregnancies and non-OD in vitro fertilization (IVF) pregnancies, and is related to fetal-maternal incompatibility. METHOD OF STUDY Women with uncomplicated pregnancy were enrolled: 25 singleton OD pregnancies and 17 non-OD IVF pregnancies. The extent of immunohistochemical staining of CD14 (pan-macrophage marker) and CD163 (type 2 macrophage marker) in both decidua basalis and parietalis was quantitated by digital image analysis. Maternal and fetal DNA was typed for human leukocyte antigen (HLA)-A, -B, C, -DRB1, and -DQB1, and fetal-maternal HLA mismatches were calculated. RESULTS OD pregnancies showed a higher percentage of CD163+ staining (P = .040) and higher CD163/CD14 ratio (P = .032) in the parietalis than non-OD IVF. The OD group was separated into a semi-allogeneic group (≤5 fetal maternal HLA mismatches) and a fully allogeneic group (> 5 mismatches). The HLA-fully-allogeneic OD group, but not the HLA-semi-allogeneic OD group, showed significantly elevated CD163/CD14 ratio in the parietalis compared with the non-OD IVF group (P < .05). CONCLUSIONS Uncomplicated OD pregnancies display a higher CD163-positive cell fraction in the total decidual macrophage population compared to autologous pregnancies, which may suggest that a local type 2 macrophage-related mechanism is needed to compensate for the higher fetal-maternal HLA mismatch load.
Collapse
Affiliation(s)
- Xuezi Tian
- Department of Obstetrics and Gynecology, Leiden University Medical Center, Leiden, Netherlands
| | - Kaveri T S Aiyer
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Dave L Roelen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
46
|
Wang L, Lu Q, Gao W, Yu S. Recent advancement on development of drug-induced macrophage polarization in control of human diseases. Life Sci 2021; 284:119914. [PMID: 34453949 DOI: 10.1016/j.lfs.2021.119914] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
Macrophages, an important part of human immune system, possess a high plasticity and heterogeneity (macrophage polarization) as classically activated macrophages (M1) and alternatively activated macrophages (M2), which exert pro-inflammatory/anti-tumor and anti-inflammatory/pro-tumor effects, respectively. Thus, drug development in induction of macrophage polarization could be used to treat different human diseases. This review summarizes the recent advancement on modulation of macrophage polarization and its related molecular mechanisms induced by a number of agents. Research on the anti-inflammatory drugs to regulate the macrophage polarization accounts for a large proportion in the field and types of diseases investigated could include atherosclerosis, enteritis, nephritis, and the nervous system and skeletal diseases, while study of the anti-tumor agents to modify macrophage polarization is a novel area of research. Future study of the molecular mechanisms by which the different agents regulate the macrophage polarization could lead to an effective control of various human diseases, including inflammation and cancers.
Collapse
Affiliation(s)
- Lu Wang
- Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, China; School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qi Lu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacy, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, China
| | - Wenwen Gao
- Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, China
| | - Shuwen Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacy, Qilu Hospital of Shandong University, Clinical Trial Center, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
47
|
Criado RFJ, Criado PR, Pagliari C, Sotto MN, Machado Filho CD, Bianco B. M2 macrophage polarization in chronic spontaneous urticaria refractory to antihistamine treatment. Allergol Int 2021; 70:504-506. [PMID: 33994101 DOI: 10.1016/j.alit.2021.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022] Open
Affiliation(s)
- Roberta F J Criado
- Department of Dermatology, Faculdade de Medicina do ABC, Santo André, SP, Brazil.
| | - Paulo Ricardo Criado
- Department of Dermatology, Faculdade de Medicina do ABC, Santo André, SP, Brazil
| | - Carla Pagliari
- Department of Pathology, Faculdade de Medicina da Universidade de São Paulo, Brazil
| | - Mirian N Sotto
- Department of Pathology, Faculdade de Medicina da Universidade de São Paulo, Brazil
| | | | - Bianca Bianco
- Department of Collective Health, Discipline of Sexual and Reproductive Health and Population Genetics, Faculdade de Medicina do ABC, Santo André, Brazil
| |
Collapse
|
48
|
Ouyang A, Wang H, Su J, Liu X. Mannose Receptor Mediates the Activation of Chitooligosaccharides on Blunt Snout Bream ( Megalobrama amblycephala) Macrophages. Front Immunol 2021; 12:686846. [PMID: 34408745 PMCID: PMC8365301 DOI: 10.3389/fimmu.2021.686846] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/16/2021] [Indexed: 01/23/2023] Open
Abstract
Chitooligosaccharide (COS) is an important immune enhancer and has been proven to have a variety of biological activities. Our previous research has established an M1 polarization mode by COS in blunt snout bream (Megalobrama amblycephala) macrophages, but the mechanism of COS activation of blunt snout bream macrophages remains unclear. In this study, we further explored the internalization mechanism and signal transduction pathway of chitooligosaccharide hexamer (COS6) in blunt snout bream macrophages. The results showed that mannose receptor C-type lectin-like domain 4-8 of M. amblycephala (MaMR CTLD4-8) could recognize and bind to COS6 and mediate COS6 into macrophages by both clathrin-dependent and caveolin-dependent pathways. In the inflammatory response of macrophages activated by COS6, the gene expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and nitric oxide synthase 2 (NOS2) was significantly inhibited after MaMR CTLD4-8-specific antibody blockade. However, even if it was blocked, the expression of these inflammation-related genes was still relatively upregulated, which suggested that there are other receptors involved in immune regulation. Further studies indicated that MaMR CTLD4-8 and Toll-like receptor 4 (TLR4) cooperated to regulate the pro-inflammatory response of macrophages caused by COS6. Taken together, these results revealed that mannose receptor (MR) CTLD4-8 is indispensable in the process of recognition, binding, internalization, and immunoregulation of COS in macrophages of blunt snout bream.
Collapse
Affiliation(s)
- Aotian Ouyang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Huabing Wang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jianguo Su
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, China
| | - Xiaoling Liu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
| |
Collapse
|
49
|
Osmulski PA, Cunsolo A, Chen M, Qian Y, Lin CL, Hung CN, Mahalingam D, Kirma NB, Chen CL, Taverna JA, Liss MA, Thompson IM, Huang THM, Gaczynska ME. Contacts with Macrophages Promote an Aggressive Nanomechanical Phenotype of Circulating Tumor Cells in Prostate Cancer. Cancer Res 2021; 81:4110-4123. [PMID: 34045187 PMCID: PMC8367292 DOI: 10.1158/0008-5472.can-20-3595] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 04/06/2021] [Accepted: 05/25/2021] [Indexed: 01/07/2023]
Abstract
Aggressive tumors of epithelial origin shed cells that intravasate and become circulating tumor cells (CTC). The CTCs that are able to survive the stresses encountered in the bloodstream can then seed metastases. We demonstrated previously that CTCs isolated from the blood of prostate cancer patients display specific nanomechanical phenotypes characteristic of cell endurance and invasiveness and patient sensitivity to androgen deprivation therapy. Here we report that patient-isolated CTCs are nanomechanically distinct from cells randomly shed from the tumor, with high adhesion as the most distinguishing biophysical marker. CTCs uniquely coisolated with macrophage-like cells bearing the markers of tumor-associated macrophages (TAM). The presence of these immune cells was indicative of a survival-promoting phenotype of "mechanical fitness" in CTCs based on high softness and high adhesion as determined by atomic force microscopy. Correlations between enumeration of macrophages and mechanical fitness of CTCs were strong in patients before the start of hormonal therapy. Single-cell proteomic analysis and nanomechanical phenotyping of tumor cell-macrophage cocultures revealed that macrophages promoted epithelial-mesenchymal plasticity in prostate cancer cells, manifesting in their mechanical fitness. The resulting softness and adhesiveness of the mechanically fit CTCs confer resistance to shear stress and enable protective cell clustering. These findings suggest that selected tumor cells are coached by TAMs and accompanied by them to acquire intermediate epithelial/mesenchymal status, thereby facilitating survival during the critical early stage leading to metastasis. SIGNIFICANCE: The interaction between macrophages and circulating tumor cells increases the capacity of tumor cells to initiate metastasis and may constitute a new set of blood-based targets for pharmacologic intervention.
Collapse
Affiliation(s)
- Pawel A Osmulski
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas.
| | - Alessandra Cunsolo
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Meizhen Chen
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Yusheng Qian
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Chun-Lin Lin
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Chia-Nung Hung
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Devalingam Mahalingam
- Department of Hematology and Oncology, University of Texas Health Science Center at San Antonio/Mays Cancer Center, San Antonio, Texas
| | - Nameer B Kirma
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Chun-Liang Chen
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Josephine A Taverna
- Department of Hematology and Oncology, University of Texas Health Science Center at San Antonio/Mays Cancer Center, San Antonio, Texas
| | - Michael A Liss
- Department of Urology, University of Texas Health Science Center/Mays Cancer Center, San Antonio, Texas
| | - Ian M Thompson
- Department of Urology, University of Texas Health Science Center/Mays Cancer Center, San Antonio, Texas
| | - Tim H-M Huang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Maria E Gaczynska
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas.
| |
Collapse
|
50
|
Vojtová L, Pavliňáková V, Muchová J, Kacvinská K, Brtníková J, Knoz M, Lipový B, Faldyna M, Göpfert E, Holoubek J, Pavlovský Z, Vícenová M, Blahnová VH, Hearnden V, Filová E. Healing and Angiogenic Properties of Collagen/Chitosan Scaffolds Enriched with Hyperstable FGF2-STAB ® Protein: In Vitro, Ex Ovo and In Vivo Comprehensive Evaluation. Biomedicines 2021; 9:590. [PMID: 34067330 PMCID: PMC8224647 DOI: 10.3390/biomedicines9060590] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Wound healing is a process regulated by a complex interaction of multiple growth factors including fibroblast growth factor 2 (FGF2). Although FGF2 appears in several tissue engineered studies, its applications are limited due to its low stability both in vitro and in vivo. Here, this shortcoming is overcome by a unique nine-point mutant of the low molecular weight isoform FGF2 retaining full biological activity even after twenty days at 37 °C. Crosslinked freeze-dried 3D porous collagen/chitosan scaffolds enriched with this hyper stable recombinant human protein named FGF2-STAB® were tested for in vitro biocompatibility and cytotoxicity using murine 3T3-A31 fibroblasts, for angiogenic potential using an ex ovo chick chorioallantoic membrane assay and for wound healing in vivo with 3-month old white New Zealand rabbits. Metabolic activity assays indicated the positive effect of FGF2-STAB® already at very low concentrations (0.01 µg/mL). The angiogenic properties examined ex ovo showed enhanced vascularization of the tested scaffolds. Histological evaluation and gene expression analysis by RT-qPCR proved newly formed granulation tissue at the place of a previous skin defect without significant inflammation infiltration in vivo. This work highlights the safety and biocompatibility of newly developed crosslinked collagen/chitosan scaffolds involving FGF2-STAB® protein. Moreover, these sponges could be used as scaffolds for growing cells for dermis replacement, where neovascularization is a crucial parameter for successful skin regeneration.
Collapse
Affiliation(s)
- Lucy Vojtová
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
| | - Veronika Pavliňáková
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
| | - Johana Muchová
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
| | - Katarína Kacvinská
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
| | - Jana Brtníková
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
| | - Martin Knoz
- Faculty of Medicine, Department of Burns and Plastic Surgery, Institution Shared with the University Hospital Brno, 625 00 Brno, Czech Republic; (M.K.); (J.H.)
- Clinic of Plastic and Esthetic Surgery, St Anne’s University Hospital, 602 00 Brno, Czech Republic
| | - Břetislav Lipový
- CEITEC–Central European Institute of Technology, Brno University of Technology, 612 00 Brno, Czech Republic; (L.V.); (J.M.); (K.K.); (J.B.); (B.L.)
- Faculty of Medicine, Department of Burns and Plastic Surgery, Institution Shared with the University Hospital Brno, 625 00 Brno, Czech Republic; (M.K.); (J.H.)
| | - Martin Faldyna
- Veterinary Research Institute, 621 00 Brno, Czech Republic; (M.F.); (E.G.); (M.V.)
| | - Eduard Göpfert
- Veterinary Research Institute, 621 00 Brno, Czech Republic; (M.F.); (E.G.); (M.V.)
| | - Jakub Holoubek
- Faculty of Medicine, Department of Burns and Plastic Surgery, Institution Shared with the University Hospital Brno, 625 00 Brno, Czech Republic; (M.K.); (J.H.)
| | - Zdeněk Pavlovský
- Faculty of Medicine, Institute of Pathology, University Hospital Brno, Masaryk University, 625 00 Brno, Czech Republic;
| | - Monika Vícenová
- Veterinary Research Institute, 621 00 Brno, Czech Republic; (M.F.); (E.G.); (M.V.)
| | - Veronika Hefka Blahnová
- Institute of Experimental Medicine of the Czech Academy of Science, 142 20 Prague, Czech Republic; (V.H.B.); (E.F.)
| | - Vanessa Hearnden
- Department of Materials Science and Engineering, Kroto Research Institute, North Campus, University of Sheffield, Broad Lane, Sheffield S3 7HQ, UK;
| | - Eva Filová
- Institute of Experimental Medicine of the Czech Academy of Science, 142 20 Prague, Czech Republic; (V.H.B.); (E.F.)
| |
Collapse
|