1
|
Yu Y, Mao N, Yu L, Lin F, Shi X, Lu X, Yang Y, Lu Y, Wang D. YiMu-QingGong san alleviates lipopolysaccharide-induced endometritis in mice via inhibiting inflammation and oxidative stress through regulating macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118992. [PMID: 39454706 DOI: 10.1016/j.jep.2024.118992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 10/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Endometritis is a common reproductive disease in dairy cattle after calving, characterized by persistent inflammation of the endometrium. YiMu-QingGong San (YMQGS), a patented Chinese medicine formulation, has demonstrated efficacy in the treatment of clinical endometritis in dairy cattle. However, the potential mechanisms by which YMQGS alleviates endometritis remain unclear. AIM OF THE STUDY The objective of this study is to elucidate the underlying therapeutic mechanisms of YMQGS in the context of endometritis. MATERIALS AND METHODS The chemical components of YMQGS were analyzed using UHPLC/MS. The anti-inflammatory capabilities of YMQGS were assessed by inducing an inflammatory response in RAW264.7 cells with lipopolysaccharide (LPS). Moreover, a clinical model of endometritis was simulated via vaginal injection of LPS into the uterine horns to investigate the therapeutic mechanisms of YMQGS. RESULTS YMQGS could downregulate the expression of inflammatory mediators (TNF-α, IL-6, IL-1β) by suppressing the activation of the TLRs/MyD88/TRAF6/NF-κB signaling pathway in RAW264.7 cells. Additionally, YMQGS facilitated the polarization of RAW264.7 cells towards M2-like macrophages (Mφs) while inhibiting M1-like Mφs polarization, thereby exerting anti-inflammatory effects. Further investigation revealed that YMQGS could diminish ROS production, augment cellular antioxidant capacity, preserve mitochondrial membrane potential, and reduce intracellular [Ca2+]i accumulation, thereby minimizing cellular damage. In the mouse model of endometritis, YMQGS similarly inhibited the activation of the NF-κB signaling pathway in uterine tissues and modulated Mφ polarization in the endometrium, leading to a decrease in the expression of inflammatory factors and subsidence of the inflammatory response in the endometrium. Histological staining results showed that YMQGS reduced endometrial fibrosis, increased the expression of endometrial ZO-1, Occludin and glycoprotein, protect the endometrial barrier function, and prevent the further development of endometritis. CONCLUSION Collectively, the present study confirms that YMQGS possesses notable anti-inflammatory and antioxidant properties. The suppression of the NF-κB signaling pathway and the modulation of Mφ polarization emerge as the pivotal mechanisms by which YMQGS alleviates endometritis.
Collapse
Affiliation(s)
- Yaming Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ningning Mao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lin Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Fangzhu Lin
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaofeng Shi
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xuanqi Lu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yang Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yu Lu
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
2
|
Yang X, Li J, Xu C, Zhang G, Che X, Yang J. Potential mechanisms of rheumatoid arthritis therapy: Focus on macrophage polarization. Int Immunopharmacol 2024; 142:113058. [PMID: 39236455 DOI: 10.1016/j.intimp.2024.113058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that affects multiple organs and systems in the human body, often leading to disability. Its pathogenesis is complex, and the long-term use of traditional anti-rheumatic drugs frequently results in severe toxic side effects. Therefore, the search for a safer and more effective antirheumatic drug is extremely important for the treatment of RA. As important immune cells in the body, macrophages are polarized. Under pathological conditions, macrophages undergo proliferation and are recruited to diseased tissues upon stimulation. In the local microenvironment, they polarize into different types of macrophages in response to specific factors and perform unique functions and roles. Previous studies have shown that there is a link between macrophage polarization and RA, indicating that certain active ingredients can ameliorate RA symptoms through macrophage polarization. Notably, Traditional Chinese medicine (TCM) monomer component and compounds demonstrate a particular advantage in this process. Building upon this insight, we reviewed and analyzed recent studies to offer valuable and meaningful insights and directions for the development and application of anti-rheumatic drugs.
Collapse
Affiliation(s)
- Xinyu Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinling Li
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengchao Xu
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinzhen Che
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiguo Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
3
|
Chang JW, Tang CH. The role of macrophage polarization in rheumatoid arthritis and osteoarthritis: Pathogenesis and therapeutic strategies. Int Immunopharmacol 2024; 142:113056. [PMID: 39217882 DOI: 10.1016/j.intimp.2024.113056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/17/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are common and debilitating joint disorders affecting millions of individuals worldwide. Despite their distinct pathological features, both conditions share a crucial role of macrophages in disease progression. Macrophages exhibit remarkable plasticity, polarizing into pro-inflammatory M1 or anti-inflammatory M2 phenotypes in response to environmental cues. An imbalance in macrophage polarization, particularly a shift towards the M1 phenotype, contributes to chronic inflammation and joint damage in RA and OA. This review explores the complex interplay between macrophages and various cell types, including T cells, B cells, synovial fibroblasts, osteoclasts, chondrocytes, and adipocytes, in the pathogenesis of these diseases. We discuss the current understanding of macrophage polarization in RA and OA, highlighting the molecular mechanisms involved. Furthermore, we provide an overview of potential therapeutic strategies targeting macrophage polarization, such as disease-modifying anti-rheumatic drugs, traditional Chinese medicine, nanomedicines, proteins, chemical compounds, and physical therapies. By elucidating the precise mechanisms governing macrophage polarization and its interactions with other cells in the joint microenvironment, researchers can identify novel therapeutic targets and develop targeted interventions to alleviate disease progression and improve patient outcomes in RA and OA.
Collapse
Affiliation(s)
- Jun-Way Chang
- The Ph.D. Program of Biotechnology and Biomedical Industry, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan; Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu, Taiwan.
| |
Collapse
|
4
|
Li H, Chen C, Huang W, Shi L, Zhang Q, Zhou L, Huang H, Zhou S. Long-term expanded hepatic progenitor cells ameliorate D-GalN/LPS-induced acute liver failure through repolarizing M1 macrophage to M2-Like phenotype via activation of the IL-10/JAK2/STAT3 signaling pathway. Int Immunopharmacol 2024; 142:113127. [PMID: 39276457 DOI: 10.1016/j.intimp.2024.113127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/25/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
Acute liver failure (ALF) is a devastating liver disease characterized by the rapid deterioration of hepatocytes, which causes a series of clinical complications, including hepatic dysfunction, coagulopathy, encephalopathy, and multiorgan failure. Cell-based therapy is a promising alternative as it can bridge patients until their livers regenerate, releasing immunomodulatory molecules to suppress inflammation. This study reports an iPSCs-derived long-term expanded hepatic progenitor cell (LTHepPCs), which can differentiate into hepatocyte-like cells (HLCs) in vivo. When introduced into drug-induced ALF models, LTHepPCs mitigate liver damage by modulating the local immune microenvironment. This is achieved by shifting macrophages/Kupffer cells towards an anti-inflammatory state, resulting in a decrease in the expression of inflammatory cytokines such as TNF-a, IL-1β, and IL-8, and an increase in the expression of anti-inflammatory cytokines such as IL-10 and ARG-1. In vitro co-culturing of THP-1 or mBMDMs with LTHepPCs suggested that LTHepPCs could activate the anti-inflammatory state of macrophages/Kupffer cells via the IL-10/JAK2/STAT3 signaling pathway. Therefore, LTHepPC transplantation is a promising therapy for ALF patients.
Collapse
Affiliation(s)
- Hongsheng Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Chen Chen
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Weijian Huang
- Celliver Biotechnology Inc., Shanghai, PR China; Department of Anesthesiology and Critical Care Medicine, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Lei Shi
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qin Zhang
- Celliver Biotechnology Inc., Shanghai, PR China
| | - Li Zhou
- Celliver Biotechnology Inc., Shanghai, PR China
| | - Hai Huang
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Road No.2, Shanghai, PR China.
| | - Shen'ao Zhou
- Celliver Biotechnology Inc., Shanghai, PR China; State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, CAS. Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, PR China.
| |
Collapse
|
5
|
Zhou X, Guo YL, Xu C, Wang J. Macrophages: Key players in diabetic wound healing. World J Diabetes 2024; 15:2177-2181. [DOI: 10.4239/wjd.v15.i11.2177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/03/2024] [Accepted: 09/18/2024] [Indexed: 10/16/2024] Open
Abstract
In this editorial, we discuss the article by Wen et al published. Diabetic foot ulcers are prevalent and serious complications of diabetes, significantly impacting patients’ quality of life and often leading to disability or death, thereby placing a heavy burden on society. Effective diabetic wound healing is hindered by an imbalance in macrophage polarization; many macrophages fail to transition from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, which is crucial for tissue remodelling and repair. The wound healing process is both dynamic and complex. Healthy M1 macrophages, which have strong phagocytic abilities, are vital during the inflammatory phase of diabetic wound healing. However, the failure to transition to M2 macrophages during the proliferative phase hinders wound healing. We anticipate the development of new therapies that can repair damaged M1 macrophages during the inflammatory phase and promote M2 macrophage polarization during the proliferative phase, thereby enhancing the overall healing process.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Science and Education, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Huzhou 313000, Zhejiang Province, China
| | - Yan-Ling Guo
- Department of Ulcers and Peripheral Vascular Surgery, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture Moxibustion, Tianjin 300381, China
| | - Chuan Xu
- Department of Pharmacy, Huzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Huzhou 313000, Zhejiang Province, China
| | - Jun Wang
- Department of Ulcers and Peripheral Vascular Surgery, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture Moxibustion, Tianjin 300381, China
| |
Collapse
|
6
|
Qian YY, Huang FF, Chen SY, Zhang WX, Wang Y, Du PF, Li G, Ding WB, Qian L, Zhan B, Chu L, Jiang DH, Yang XD, Zhou R. Therapeutic effect of recombinant Echinococcus granulosus antigen B subunit 2 protein on sepsis in a mouse model. Parasit Vectors 2024; 17:467. [PMID: 39548530 PMCID: PMC11566433 DOI: 10.1186/s13071-024-06540-x] [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: 06/08/2024] [Accepted: 10/17/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Sepsis is a potentially fatal systemic inflammatory response syndrome (SIRS) that threatens millions of lives worldwide. Echinococcus granulosus antigen B (EgAgB) is a protein released by the larvae of the tapeworm. This protein has been shown to play an important role in modulating host immune response. In this study we expressed EgAgB as soluble recombinant protein in E. coli (rEgAgB) and explored its protective effect on sepsis. METHODS The sepsis model was established by cecal ligation and puncture (CLP) procedure in BALB/c mice. The therapeutic effect of rEgAgB on sepsis was performed by interperitoneally injecting 5 µg rEgAgB in mice with CLP-induced sepsis and observing the 72 h survival rate after onset of sepsis. The proinflammatory cytokines [tumor necrosis factor (TNF)-α, interleukin (IL)-6] and regulatory cytokines [IL-10, transforming growth factor beta (TGF-β)] were measured in sera, and the histopathological change was observed in livers, kidneys, and lungs of septic mice treated with rEgAgB compared with untreated mice. The effect of rEgAgB on the macrophage polarization was performed in vitro by incubating rEgAgB with peritoneal macrophages. The levels of TLR2 and MyD88 were measured in these tissues to determine the involvement of TLR-2/MyD88 in the sepsis-induced inflammatory signaling pathway. RESULTS In vivo, we observed that treatment with rEgAgB significantly increased the survival rate of mice with CLP-induced sepsis up to 72 h while all mice without treatment died within the same period. The increased survival was associated with reduced pathological damage in key organs such as liver, lung, and kidneys. It was supported by the reduced proinflammatory cytokine levels and increased regulatory cytokine expression in peripheral blood and key organ tissues. Further study identified that treatment with rEgAgB promoted macrophage polarization from classically activated macrophage (M1) to regulatory M2-like macrophage via inhibiting TLR2/MyD88 signal pathway. CONCLUSIONS The therapeutic effects of rEgAgB on mice with sepsis was observed in a mice model that was associated with reduced inflammatory responses and increased regulatory responses, possibly through inducing polarization of macrophages from proinflammatory M1 to regulatory M2 phenotype through inhibiting TLR2/MyD88 inflammatory pathway.
Collapse
Affiliation(s)
- Ya-Yun Qian
- First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
- First People's Hospital of Changzhou, Changzhou, 213000, China
| | - Fei-Fei Huang
- First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
| | - Si-Yu Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214028, China
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Wei-Xiao Zhang
- First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
| | - Yin Wang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Peng-Fei Du
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Gen Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
| | - Wen-Bo Ding
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
| | - Lei Qian
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Liang Chu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
- Second Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
| | - Dong-Hui Jiang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China.
- Department of Critical Care Medicine, First People's Hospital of Haidong, Haidong, 810600, China.
| | - Xiao-Di Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China.
| | - Rui Zhou
- First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China.
| |
Collapse
|
7
|
Peng D, Li M, Yu Z, Yan T, Yao M, Li S, Liu Z, Li LF, Qiu HJ. Synergy between pluripotent stem cell-derived macrophages and self-renewing macrophages: Envisioning a promising avenue for the modelling and cell therapy of infectious diseases. Cell Prolif 2024:e13770. [PMID: 39537185 DOI: 10.1111/cpr.13770] [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: 06/28/2024] [Revised: 09/30/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
As crucial phagocytes of the innate immune system, macrophages (Mϕs) protect mammalian hosts, maintain tissue homeostasis and influence disease pathogenesis. Nonetheless, Mϕs are susceptible to various pathogens, including bacteria, viruses and parasites, which cause various infectious diseases, necessitating a deeper understanding of pathogen-Mϕ interactions and therapeutic insights. Pluripotent stem cells (PSCs) have been efficiently differentiated into PSC-derived Mϕs (PSCdMϕs) resembling primary Mϕs, advancing the modelling and cell therapy of infectious diseases. However, the mass production of PSCdMϕs, which lack proliferative capacity, relies on large-scale expansions of PSCs, thereby increasing both costs and culture cycles. Notably, Mϕs deficient in the MafB/c-Maf genes have been reported to re-enter the cell cycle with the stimulation of specific growth factor cocktails, turning into self-renewing Mϕs (SRMϕs). This review summarizes the applications of PSCdMϕs in the modelling and cell therapy of infectious diseases and strategies for establishing SRMϕs. Most importantly, we innovatively propose that PSCs can serve as a gene editing platform to creating PSC-derived SRMϕs (termed PSRMϕs), addressing the resistance of Mϕs against genetic manipulation. We discuss the challenges and possible solutions in creating PSRMϕs. In conclusion, this review provides novel insights into the development of physiologically relevant and expandable Mϕ models, highlighting the enormous potential of PSRMϕs as a promising avenue for the modelling and cell therapy of infectious diseases.
Collapse
Affiliation(s)
- Dingkun Peng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Meilin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhuoran Yu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Tingsheng Yan
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Meng Yao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Su Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhonghua Liu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, China
| | - Lian-Feng Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
8
|
Gomes-da-Silva NC, Silva França ÁR, Dos Santos CC, Alencar LMR, Rosas EC, Correa LB, Lorentino CMA, Santos ALS, Ricci-Junior E, Santos-Oliveira R. Nano-Enhanced Benzylpenicillin: Bridging Antibacterial Action with Anti-Inflammatory Potential against Antibiotic-Resistant Bacteria. Microbes Infect 2024:105436. [PMID: 39542238 DOI: 10.1016/j.micinf.2024.105436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
This study investigates the enhancement of benzylpenicillin's antibacterial properties using nanomedicine, specifically by developing benzylpenicillin nanoemulsions. To address the escalating issue of bacterial resistance, we employed the advanced techniques Raman spectroscopy and atomic force microscopy to analyze the nanoemulsions' molecular structure and characteristics. We then evaluated the impact of these nanoemulsions on nitric oxide production by macrophages to deternine their potential to modulate inflammatory responses. We further assessed the antibacterial effectiveness of the nanoparticles against the pathogens Streptococcus pyogenes (Group A Streptococcus) and Streptococcus agalactiae (Group B Streptococcus). The results of antibiograms showed significant efficacy against Gram-positive bacteria, with minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values, confirming their bactericidal potential. The investigation into the mechanism of action suggested substantial disruption to bacterial membrane integrity, underscoring a possible mode of antibacterial activity. Overall, the study provides valuable insights into the synergistic relationship between antibiotics and nanoparticles. In particular, it demonstrates the potential of benzylpenicillin nanoparticles to enhance the antimicrobial efficacy and influence inflammatory responses obtained by evaluating nitrite, IL-6 and TNF-α, offering promising avenues for future clinical applications and strategies to combat bacterial resistance.
Collapse
Affiliation(s)
- Natália Cristina Gomes-da-Silva
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Rio de Janeiro 21941906, RJ, Brazil
| | - Álefe Roger Silva França
- Biophysics and Nanosystems Laboratory, Federal University of Maranhão, Department of Physics, São Luis 65065690, MA, Brazil
| | - Clenilton Costa Dos Santos
- Biophysics and Nanosystems Laboratory, Federal University of Maranhão, Department of Physics, São Luis 65065690, MA, Brazil
| | | | - Elaine Cruz Rosas
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, 21040-360, Brazil
| | - Luana Barbosa Correa
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Rio de Janeiro 21941906, RJ, Brazil; Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, 21040-360, Brazil
| | - Carolline M A Lorentino
- Laboratory of Advanced Studies on Emerging and Resistant Microorganisms (LEAMER); Department of General Microbiology, Paulo de Góes Institute of Microbiology (IMPG), Federal University of Rio de Janeiro (UFRJ)
| | - André L S Santos
- Laboratory of Advanced Studies on Emerging and Resistant Microorganisms (LEAMER); Department of General Microbiology, Paulo de Góes Institute of Microbiology (IMPG), Federal University of Rio de Janeiro (UFRJ)
| | - Eduardo Ricci-Junior
- Federal University of Rio de Janeiro, School of Pharmacy, Rio de Janeiro, 21941900, RJ, Brazil
| | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Rio de Janeiro 21941906, RJ, Brazil; Rio de Janeiro State University, Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Rio de Janeiro 23070200, RJ, Brazil.
| |
Collapse
|
9
|
Chen S, Zhu H, Jounaidi Y. Comprehensive snapshots of natural killer cells functions, signaling, molecular mechanisms and clinical utilization. Signal Transduct Target Ther 2024; 9:302. [PMID: 39511139 PMCID: PMC11544004 DOI: 10.1038/s41392-024-02005-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/15/2024] Open
Abstract
Natural killer (NK) cells, initially identified for their rapid virus-infected and leukemia cell killing and tumor destruction, are pivotal in immunity. They exhibit multifaceted roles in cancer, viral infections, autoimmunity, pregnancy, wound healing, and more. Derived from a common lymphoid progenitor, they lack CD3, B-cell, or T-cell receptors but wield high cytotoxicity via perforin and granzymes. NK cells orchestrate immune responses, secreting inflammatory IFNγ or immunosuppressive TGFβ and IL-10. CD56dim and CD56bright NK cells execute cytotoxicity, while CD56bright cells also regulate immunity. However, beyond the CD56 dichotomy, detailed phenotypic diversity reveals many functional subsets that may not be optimal for cancer immunotherapy. In this review, we provide comprehensive and detailed snapshots of NK cells' functions and states of activation and inhibitions in cancer, autoimmunity, angiogenesis, wound healing, pregnancy and fertility, aging, and senescence mediated by complex signaling and ligand-receptor interactions, including the impact of the environment. As the use of engineered NK cells for cancer immunotherapy accelerates, often in the footsteps of T-cell-derived engineering, we examine the interactions of NK cells with other immune effectors and relevant signaling and the limitations in the tumor microenvironment, intending to understand how to enhance their cytolytic activities specifically for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Zhang Y, Song M, Fan J, Guo X, Tao S. Impact of probiotics-derived extracellular vesicles on livestock gut barrier function. J Anim Sci Biotechnol 2024; 15:149. [PMID: 39506860 PMCID: PMC11542448 DOI: 10.1186/s40104-024-01102-8] [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: 06/03/2024] [Accepted: 09/05/2024] [Indexed: 11/08/2024] Open
Abstract
Probiotic extracellular vesicles (pEVs) are biologically active nanoparticle structures that can regulate the intestinal tract through direct or indirect mechanisms. They enhance the intestinal barrier function in livestock and poultry and help alleviate intestinal diseases. The specific effects of pEVs depend on their internal functional components, including nucleic acids, proteins, lipids, and other substances. This paper presents a narrative review of the impact of pEVs on the intestinal barrier across various segments of the intestinal tract, exploring their mechanisms of action while highlighting the limitations of current research. Investigating the mechanisms through which probiotics operate via pEVs could deepen our understanding and provide a theoretical foundation for their application in livestock production.
Collapse
Affiliation(s)
- Yuhan Zhang
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Mengzhen Song
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Jinping Fan
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Xuming Guo
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China
| | - Shiyu Tao
- College of Animal Sciences and Technology, Huazhong Agricultural University, No. 1 Shizishan Street, Hongshan District, Wuhan, Hubei Province, 430070, China.
| |
Collapse
|
11
|
Yang H, Wang YY, Chang W, Zhai M, Du WJ, Jiang W, Xiang YW, Qin G, Yu J, Gong Y, Han Q. Primary sensory neuron-derived miR-let-7b underlies stress-elicited psoriasis. Brain Behav Immun 2024; 123:997-1010. [PMID: 39510418 DOI: 10.1016/j.bbi.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/09/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024] Open
Abstract
Psoriasis, a chronic autoimmune skin condition with significant global morbidity, badly impairs patients' quality of life. Stress has been identified as a prominent trigger for psoriasis, and effectively management of stress can ameliorate its pathological manifestations. However, the precise mechanisms by which stress influences psoriasis remain elusive. In this study, we found that mice subjected to chronic social defeat stress (CSDS) exhibit severer imiquimod (IMQ)-induced psoriasis with increased epidermal scaling, epidermal hyperplasia, number of epidermal ridges, itch, and skin inflammation than control mice. Mechanistic study reveals that CSDS leads to an elevated release of miR-let-7b, an endogenous ligand of Toll-like receptor 7 (TLR7), from the peripheral terminal of dorsal root ganglia (DRG) neurons into the skin. This process can stimulate skin-resident macrophages to release cytokines (such as IL-6 and TNF-a) and chemokines (such as MCP-1), subsequently promoting the recruitment of additional macrophages into the skin. Notably, the specific blockade of miR-let-7b in DRG neurons effectively relieve stress-induced exacerbations of psoriasis. Furthermore, intradermal injection of synthetic miR-let-7b can induce a psoriasis-like phenotype in wildtype mice, a phenomenon that can be countered by the application of a TLR7 antagonist. Additionally, microfluidic chamber coculture assays demonstrated that miR-let-7b released by DRG neurons activates macrophages via TLR7 expressed on these immune cells. Totally, this study found that stress-induced upregulation and release of miR-let-7b from DRG neurons stimulates macrophages to secrete more inflammatory cytokines and chemokines, thereby exacerbating skin inflammation and the psoriatic phenotype. These findings provide a potential therapeutic strategy targeting the miR-let-7b/TLR7 pathway to alleviate stress-induced exacerbation of psoriasis.
Collapse
Affiliation(s)
- Huan Yang
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yun-Yun Wang
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Weiqi Chang
- Department of Integrative Medicine and Neurobiology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mengying Zhai
- Department of Integrative Medicine and Neurobiology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wan-Jie Du
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Wencheng Jiang
- Traditional Chinese Medicine Dermatology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Yan-Wei Xiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, 201203 Shanghai, China
| | - Guoyou Qin
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai 200032, China
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai 200433, China.
| | - Ye Gong
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Qingjian Han
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
12
|
Carlà MM, Gambini G, Caporossi T, Giannuzzi F, Boselli F, Crincoli E, Ripa M, Rizzo S. Ocular Involvement in Systemic Sclerosis: Updated Review and New Insights on Microvascular Impairment. Ocul Immunol Inflamm 2024; 32:2209-2216. [PMID: 38466107 DOI: 10.1080/09273948.2024.2308030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/01/2024] [Accepted: 01/16/2024] [Indexed: 03/12/2024]
Abstract
Systemic sclerosis (SSc) is a chronic multisystemic disease characterized by immunological activation, diffuse vasculopathy, and generalized fibrosis exhibiting a variety of symptoms. A recognized precursor of SSc is Raynaud's phenomenon, which is part of the very early disease of systemic sclerosis (VEDOSS) in combination with nailfold videocapillaroscopy (NVC) impairment. The pathophysiology of ocular involvement, alterations in internal organs, and body integumentary system involvement in SSc patients are complicated and poorly understood, with multiple mechanisms presumptively working together. The most prevalent ocular symptoms of SSc are abnormalities of the eyelids and conjunctiva as well as dry eye syndrome, due to fibroblasts' dysfunction and inflammation of the ocular surface. In particular, lagophthalmos, blepharophimosis limitation of eyelid motion, eyelid telangiectasia, and rigidity or tightening of the lids may affect up to two-third of the patients. In addition, reduction in central corneal thickness, iris defects and higher rates of glaucoma were reported. In the first reports based on retinography or fluorescein angiography, about 50% of SSc patients showed signs of vascular disease: peripheral artery occlusion, thinning of retinal pigment epithelium and choroidal capillaries, ischemic areas surrounded by intraretinal extravasation and microaneurysms, and peripheral capillary non-perfusion. Successively, thanks to the advent of optical coherence tomography angiography (OCTA), several studies highlighted significant impairment of either the choriocapillaris and retinal vascular plexuses, also correlating with NVC involvement and skin disease, even in VEDOSS disease. Given the sensitivity of this technique, ocular micro-vasculopathy may act as a tool for early SSc identification and discriminate between disease stages.
Collapse
Affiliation(s)
- Matteo Mario Carlà
- Ophthalmology Department, "Fondazione Policlinico Universitario A. Gemelli, IRCCS", Rome, Italy
- Ophthalmology Department, Catholic University "Sacro Cuore", Rome, Italy
| | - Gloria Gambini
- Ophthalmology Department, "Fondazione Policlinico Universitario A. Gemelli, IRCCS", Rome, Italy
- Ophthalmology Department, Catholic University "Sacro Cuore", Rome, Italy
| | - Tomaso Caporossi
- Ophthalmology Department, Catholic University "Sacro Cuore", Rome, Italy
- Vitreoretinal Surgery Unit, Fatebenefratelli Isola Tiberina Gemelli Isola Hospital, Rome, Italy
| | - Federico Giannuzzi
- Ophthalmology Department, "Fondazione Policlinico Universitario A. Gemelli, IRCCS", Rome, Italy
- Ophthalmology Department, Catholic University "Sacro Cuore", Rome, Italy
| | - Francesco Boselli
- Ophthalmology Department, "Fondazione Policlinico Universitario A. Gemelli, IRCCS", Rome, Italy
- Ophthalmology Department, Catholic University "Sacro Cuore", Rome, Italy
| | - Emanuele Crincoli
- Ophthalmology Department, "Fondazione Policlinico Universitario A. Gemelli, IRCCS", Rome, Italy
- Ophthalmology Department, Catholic University "Sacro Cuore", Rome, Italy
| | - Matteo Ripa
- Department of Ophthalmology, William Harvey Hospital, East Kent Hospitals University NHS Foundation Trust, Willesborough, UK
| | - Stanislao Rizzo
- Ophthalmology Department, "Fondazione Policlinico Universitario A. Gemelli, IRCCS", Rome, Italy
- Ophthalmology Department, Catholic University "Sacro Cuore", Rome, Italy
| |
Collapse
|
13
|
Zhang T, Tai Z, Miao F, Zhao Y, Wang W, Zhu Q, Chen Z. Bioinspired nanovesicles derived from macrophage accelerate wound healing by promoting angiogenesis and collagen deposition. J Mater Chem B 2024. [PMID: 39480489 DOI: 10.1039/d3tb02158k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Macrophages play a crucial role in the process of wound healing. In order to effectively inhibit excessive inflammation and facilitate skin wound healing, it is necessary to transform overactive M1 macrophages in injured tissues into the M2 type. In this study, we have successfully generated bioinspired nanovesicles (referred to as M2BNVs) from M2 type macrophages. These nanovesicles not only possess physical and biological properties that closely resemble exosomes, but also offer a simpler preparation process and more abundant yield. Owing to their distinctive endogenous cargo, M2BNVs have the ability to re-educate M1 macrophages, shifting their phenotype towards the M2 type which is known to promote healing and possess anti-inflammatory properties. Consequently, M2BNVs effectively improve the prevailing pro-inflammatory microenvironment within the wound. Furthermore, M2BNVs also facilitate wound tissue regeneration and angiogenesis. Collectively, our findings demonstrate the potential of M2BNVs in promoting wound healing in mice.
Collapse
Affiliation(s)
- Tingrui Zhang
- School of Medicine, Shanghai University, Shanghai 200444, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, 1278 Baode Road, Shanghai 200443, P. R. China.
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, 1278 Baode Road, Shanghai 200443, P. R. China.
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Fengze Miao
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, 1278 Baode Road, Shanghai 200443, P. R. China.
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Yingchao Zhao
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, 1278 Baode Road, Shanghai 200443, P. R. China.
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Weifan Wang
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, 1278 Baode Road, Shanghai 200443, P. R. China.
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, 1278 Baode Road, Shanghai 200443, P. R. China.
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| | - Zhongjian Chen
- School of Medicine, Shanghai University, Shanghai 200444, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, 1278 Baode Road, Shanghai 200443, P. R. China.
- Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Shan X, Ji Z, Wang B, Zhang Y, Dong H, Jing W, Zhou Y, Hu P, Cui Y, Li Z, Yu S, Zhou J, Wang T, Shen L, Liu Y, Yu Q. Riboflavin kinase binds and activates inducible nitric oxide synthase to reprogram macrophage polarization. Redox Biol 2024; 78:103413. [PMID: 39536592 DOI: 10.1016/j.redox.2024.103413] [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: 09/20/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Riboflavin kinase (RFK) is essential in riboflavin metabolism, converting riboflavin to flavin mononucleotide (FMN), which is further processed to flavin adenine dinucleotide (FAD). While RFK enhances macrophage phagocytosis of Listeria monocytogenes, its role in macrophage polarization is not well understood. Our study reveals that RFK deficiency impairs M(IFN-γ) and promotes M(IL-4) polarization, both in vitro and in vivo. Mechanistically, RFK interacts with inducible nitric oxide (NO) synthase (iNOS), which requires FMN and FAD as cofactors for activation, leading to increased NO production that alters energy metabolism by inhibiting the tricarboxylic acid cycle and mitochondrial electron transport chain. Exogenous FAD reverses the metabolic and polarization changes caused by RFK deficiency. Furthermore, bone marrow adoptive transfer from high-riboflavin-fed mice into wild-type tumor-bearing mice reprograms tumor-associated macrophage polarization and inhibits tumor growth. These results suggest that targeting RFK-iNOS or modulating riboflavin metabolism could be potential therapies for macrophage-related immune diseases.
Collapse
Affiliation(s)
- Xiao Shan
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000, China; Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Zemin Ji
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Baochen Wang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yanan Zhang
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hongyuan Dong
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Weijia Jing
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yanzhao Zhou
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China; University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Penghui Hu
- Department of Critical Care Medicine, Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yan Cui
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zihan Li
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Sujun Yu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Ting Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin 300070, China
| | - Long Shen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China; Tianjin Institutes of Health Science, Tianjin 301600, China.
| | - Yuping Liu
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| | - Qiujing Yu
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| |
Collapse
|
15
|
Xiong B, Li C, Hong G, Li J, Luo Q, Gong J, Lai X. HMGB1/TREM1 crosstalk between heat-injured hepatocytes and macrophages promotes HCC progression after RFA. J Cancer Res Clin Oncol 2024; 150:480. [PMID: 39465435 PMCID: PMC11513699 DOI: 10.1007/s00432-024-05996-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
PURPOSE Tumor recurrence after radiofrequency ablation (RFA) affects the survival rate of patients and limits its clinical application. Tumor recurrence around the ablation area may be related to the thermal injury of hepatocytes (HCs) around the tumor, but the specific mechanism is still unclear. METHODS A liver cancer thermal injury mouse model was established via RFA in the C57BL/6 mice. Primary HCs and Kupffer cells (KCs) were isolated and cultured to assess their sensitivity to thermal injury via the MTT assay. Flow cytometry was used to assess macrophage polarization. Furthermore, Western blotting and co-immunoprecipitation (co-IP) were utilized to evaluate the protein expression of intracellular signaling pathway. Finally, Transwell and wound healing assays was conducted to evaluate the invasion potential of liver cancer cells. RESULTS Our findings revealed that RFA-induced liver thermal injury promoted the upregulation and secretion of HMGB1 in HCs. HMGB1 had a protective effect on HCs thermal injury, potentially mediated through autophagy regulation. Heat-injured HCs release HMGB1, which activates the TREM1/JAK2/STAT3 signaling pathway in KCs, thus fostering an immunosuppressive tumor microenvironment (TME). Moreover, HMGB1 secretion by heat-injured HCs exacerbates the migration and invasion of HCC cells by influencing macrophage polarization. CONCLUSION RFA-induced thermal injury triggers HMGB1 release from HCs, driving macrophage M2 polarization and increasing the invasion ability of liver cancer cells. These findings reveal a potential therapeutic target for combating liver cancer recurrence following thermal ablation.
Collapse
Affiliation(s)
- Bin Xiong
- Hepatobiliary Surgery, The People's Hospital of Tongnan District Chongqing City, Chongging, China
- Chongqing Hospital of Traditional Chinese Medicine, Chongging, China
| | - Chunming Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Guoqing Hong
- Hepatobiliary Surgery, The People's Hospital of Tongnan District Chongqing City, Chongging, China
| | - Junke Li
- Hepatobiliary Surgery, The People's Hospital of Tongnan District Chongqing City, Chongging, China
| | - Qing Luo
- Hepatobiliary Surgery, The People's Hospital of Tongnan District Chongqing City, Chongging, China
| | - Jianping Gong
- Hepatobiliary Surgery, The People's Hospital of Tongnan District Chongqing City, Chongging, China
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xing Lai
- Hepatobiliary Surgery, The People's Hospital of Tongnan District Chongqing City, Chongging, China.
- Chongqing Hospital of Traditional Chinese Medicine, Chongging, China.
| |
Collapse
|
16
|
Yoon C, Kim HK, Ham YS, Gil WJ, Mun SJ, Cho E, Yuk JM, Yang CS. Toxoplasma gondii macrophage migration inhibitory factor shows anti- Mycobacterium tuberculosis potential via AZIN1/STAT1 interaction. SCIENCE ADVANCES 2024; 10:eadq0101. [PMID: 39453997 PMCID: PMC11506136 DOI: 10.1126/sciadv.adq0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/20/2024] [Indexed: 10/27/2024]
Abstract
Mycobacterium tuberculosis (MTB) is a pathogenic bacterium, belonging to the family Mycobacteriaceae, that causes tuberculosis (TB). Toxoplasma gondii macrophage migration inhibitory factor (TgMIF), a protein homolog of macrophage migration inhibitory factor, has been explored for its potential to modulate immune responses during MTB infections. We observed that TgMIF that interacts with CD74, antizyme inhibitor 1 (AZIN1), and signal transducer and activator of transcription 1 (STAT1) modulates endocytosis, restoration of mitochondrial function, and macrophage polarization, respectively. These interactions promote therapeutic efficacy in mice infected with MTB, thereby presenting a potential route to host-directed therapy development. Furthermore, TgMIF, in combination with first-line TB drugs, significantly inhibited drug-resistant MTB strains, including multidrug-resistant TB. These results demonstrate that TgMIF is potentially a multifaceted therapeutic agent against TB, acting through immune modulation, enhancement of mitochondrial function, and dependent on STAT1 and AZIN1 pathways.
Collapse
Affiliation(s)
- Chanjin Yoon
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
- Institute of Natural Science & Technology, Hanyang University, Ansan 15588, South Korea
| | - Hyo Keun Kim
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Yu Seong Ham
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Woo Jin Gil
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
| | - Seok-Jun Mun
- Department of Bionano Engineering, Hanyang University, Seoul 04673, South Korea
| | - Euni Cho
- Department of Bionano Engineering, Hanyang University, Seoul 04673, South Korea
| | - Jae-Min Yuk
- Department of Infection Biology and Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Chul-Su Yang
- Department of Molecular and Life Science, Hanyang University, Ansan 15588, South Korea
- Department of Medicinal and Life Science, Hanyang University, Ansan 15588, South Korea
| |
Collapse
|
17
|
Li Y, Li M, Zheng J, Ma Z, Yu T, Zhu Y, Li P, Nie F. Ultrasound-Responsive Nanocarriers Delivering siRNA and Fe 3O 4 Nanoparticles Reprogram Macrophages and Inhibit M2 Polarization for Enhanced NSCLC Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56634-56652. [PMID: 39378273 DOI: 10.1021/acsami.4c10036] [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: 10/10/2024]
Abstract
Lung cancer has emerged as the second most common type of malignant tumor worldwide, and it has the highest mortality rate. The overall 5-year survival rate stands at less than 20%, which is primarily related to the limited therapeutic options and the complexity of the tumor immune microenvironment. In the tumor microenvironment, M1 macrophages are known for their tumor-killing capabilities. Although they are less numerous, they play an important role in tumor immunity. Therefore, increasing M1 macrophages' presence is considered a strategy to enhance targeted phagocytosis and antitumor efficacy in nonsmall cell lung cancer (NSCLC). This study introduces the development of folic acid (FA)-conjugated liposomal nanobubbles for precise delivery of PFH, STAT3 siRNA, and Fe3O4 to the tumor microenvironment. These encapsulated PFH liposomal nanobubbles exhibit significant visualization potential and underwent phase transition when exposed to low-intensity focused ultrasound (LIFU). The release of Fe3O4 activates the IRF5 signaling pathway, converting M2-like macrophages to M1. In addition, STAT3 siRNA effectively interrupts the JAK-STAT3 pathway, inhibiting the polarization of M2-like macrophages in tumor-associated macrophages (TAMs). This dual-action therapy facilitates T-cell activation and proliferation, thereby enhancing the immune response against NSCLC.
Collapse
Affiliation(s)
- Yuanyuan Li
- Ultrasound Medical Center, Gansu Province Clinical Research Center for Ultrasonography, Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Ming Li
- Ultrasound Medical Center, Gansu Province Clinical Research Center for Ultrasonography, Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Jun Zheng
- The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Zhen Ma
- Peking University Third Hospital, Beijing 100191, China
| | - Tingting Yu
- Ultrasound Medical Center, Gansu Province Clinical Research Center for Ultrasonography, Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Yangyang Zhu
- Ultrasound Medical Center, Gansu Province Clinical Research Center for Ultrasonography, Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Pan Li
- The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, People's Republic of China
| | - Fang Nie
- Ultrasound Medical Center, Gansu Province Clinical Research Center for Ultrasonography, Gansu Province Medical Engineering Research Center for Intelligence Ultrasound, Lanzhou University Second Hospital, Lanzhou 730000, China
| |
Collapse
|
18
|
Ehrenzeller SA, Lukesh NR, Stiepel RT, Middleton DD, Nuzzolo SM, Tate AJ, Batty CJ, Bachelder EM, Ainslie KM. Comparison of emulsion and spray methods for fabrication of rapamycin-loaded acetalated dextran microparticles. RSC PHARMACEUTICS 2024; 1:727-741. [PMID: 39415944 PMCID: PMC11474811 DOI: 10.1039/d4pm00054d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/04/2024] [Indexed: 10/19/2024]
Abstract
Rapamycin (rapa), an immunosuppressive medication, has demonstrated considerable effectiveness in reducing organ transplant rejection and treating select autoimmune diseases. However, the standard oral administration of rapa results in poor bioavailability, broad biodistribution, and harmful off-target effects, necessitating improved drug delivery formulations. Polymeric microparticles (MPs) are one such solution and have demonstrated promise in pre-clinical studies to improve the therapeutic efficacy of rapa. Nevertheless, MP formulations are highly diverse, and fabrication method selection is a critical consideration in formulation design. Herein, we compared common fabrication processes for the development of rapa-loaded MPs. Using the biopolymer acetalated dextran (Ace-DEX), rapa-loaded MPs were fabricated by both emulsion (homogenization and sonication) and spray (electrospray and spray drying) methods, and resultant MPs were characterized for size, morphology, surface charge, and drug release kinetics. MPs were then screened in LPS-stimulated macrophages to gauge immunosuppressive efficacy relative to soluble drug. We determined that homogenized MPs possessed the most optimal combination of sizing, tunable drug release kinetics, and immunosuppressive efficacy, and we subsequently demonstrated that these characteristics were maintained across a range of potential rapa loadings. Further, we performed in vivo trafficking studies to evaluate depot kinetics and cellular uptake at the injection site after subcutaneous injection of homogenized MPs. We observed preferential MP uptake by dendritic cells at the depot, highlighting the potential for MPs to direct more targeted drug delivery. Our results emphasize the significance of fabrication method in modulating the efficacy of MP systems and inform improved formulation design for the delivery of rapa.
Collapse
Affiliation(s)
- Stephen A Ehrenzeller
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Nicole Rose Lukesh
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Rebeca T Stiepel
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Denzel D Middleton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Steven M Nuzzolo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Aliyah J Tate
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Cole J Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University Chapel Hill North Carolina USA
- Department of Microbiology & Immunology, UNC School of Medicine, University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| |
Collapse
|
19
|
Su L, Rao K, Wang L, Pu L, Zhang Z, Li H, Li R, Liu D. Prenylated Dihydroflavonol from Sophora flavescens Regulate the Polarization and Phagocytosis of Macrophages In Vitro. Molecules 2024; 29:4741. [PMID: 39407669 PMCID: PMC11477850 DOI: 10.3390/molecules29194741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
As an important member of innate immunity, macrophages show remarkable plasticity and heterogeneity, and play an important role in immune regulation, tissue development, homeostasis of the internal environment and injury repair. However, the excessive activation of macrophages is closely related to the occurrence and development of many diseases. The prenylated flavonoid structure is one of the characteristic structures isolated from Sophora flavescens, with anti-inflammatory, anti-tumor, anti-allergy and other effects. In this study, the effects of (2R)-3β,7,4'-trihydroxy-5-methoxy-8-prenylflavanone (TMP), a prenylated dihydroflavonol, on the polarization and phagocytosis of macrophages were systematically studied. In LPS-induced M1-type macrophages, TMP dose-dependently inhibited the expression of COX-2, iNOS and the secretion of NO, IL-1β, IL-6 and IL-18, showing an inhibitory effect on M1 polarization. Further experiments revealed that it was related to the inhibition of TLR4-related AKT/mTOR, MAPK and NF-κB signaling pathways; in IL-4-induced M2-type macrophages, TMP down-regulated the expression of M2-related Arg1, IL-10, TGF-β, CD206 and CD163, as well as the phosphorylation levels of AKT1 and STAT6. For macrophages in a physiological state, it was very important for cells to return from a stress state to a phenotypic stability in the M0 state. These results indicated that TMP negatively regulated the M1/M2 polarization of macrophages, and made them tend to M0 homeostasis, which might provide new theoretical and data support for explaining the anti-inflammatory immunoregulatory activity of Sophora flavescens.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rongtao Li
- Center for Pharmaceutical Sciences and Engineering, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Dan Liu
- Center for Pharmaceutical Sciences and Engineering, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
20
|
Shi Z, Zhou M, Zhai J, Sun J, Wang X. Novel therapeutic strategies and drugs for idiopathic pulmonary fibrosis. Arch Pharm (Weinheim) 2024; 357:e2400192. [PMID: 38961537 DOI: 10.1002/ardp.202400192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease of unknown etiology. Currently, drugs used to treat IPF in clinical practice exhibit severe side effects and limitations. To address these issues, this paper discusses the therapeutic effects of preclinical targeted drugs (such as STAT3 and TGF-β/Smad pathway inhibitors, chitinase inhibitors, PI3K and phosphodiesterase inhibitors, etc.) and natural products on IPF. Through a summary of current research progress, it is found that natural products possess multitarget effects, stable therapeutic efficacy, low side effects, and nondrug dependence. Furthermore, we discuss the significant prospects of natural product molecules in combating fibrosis by influencing the immune system, expecting that current analytical data will aid in the development of new drugs or the investigation of active ingredients in natural products for potential IPF treatments in the future.
Collapse
Affiliation(s)
- Zezhou Shi
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Min Zhou
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Jingfang Zhai
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Jie Sun
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| | - Xiaojing Wang
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, China
| |
Collapse
|
21
|
Li H, Hu YF, Wang XR, Ouyang KW, Wang H, Wang KW, Chang W, Zhang J, Yuan Z, Xiong YW, Zhu HL, Yang L, Wang H. Suppressed testicular macrophage M1 polarization by HDAC5 enforces insensitivity to LPS-elicited blood-testis barrier damage. Food Chem Toxicol 2024; 192:114940. [PMID: 39151879 DOI: 10.1016/j.fct.2024.114940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Infertility caused by lipopolysaccharide (LPS) exposure due to infection is endangering male fertility worldwide, but the mechanism remains unclear. The blood-testis barrier (BTB) is essential for maintaining spermatogenesis and male fertility. In the present study, we showed that LPS (5.0 mg/kg) treatment markedly down-regulated the expression of BTB-related proteins, expanded the biotin penetration distance and caused histopathological injury in seminiferous tubules in mouse testes. Notably, testicular macrophage M1 polarization induced by LPS seems to be related to BTB damage, which was well confirmed by co-culture of RAW264.7 and TM4 cells in vitro. Interestingly, a low-dose LPS (0.1 mg/kg) pretreatment attenuated down-regulation of BTB-related proteins expression and histopathological injury and shorten biotin penetration distance in seminiferous tubules caused by LPS. Correspondingly, a low-dose LPS pretreatment suppresses testicular macrophage M1 polarization induced by LPS in mouse testes. Further experiments revealed that histone deacetylase 5 (HDAC5) was markedly down-regulated at 2 h and slightly down-regulated at 8 h, but up-regulated at 24 h in mouse testes after LPS treatment. Additionally, low-dose LPS pretreatment against the down-regulation of HDAC5 protein caused by LPS treatment. Notably, the suppressed testicular macrophage M1 polarization by low-dose LPS pretreatment was broken by BRD4354, a specific inhibitor of HDAC5 in vitro. These results suggest suppressed testicular macrophage M1 polarization by HDAC5 enforces insensitivity to LPS-elicited BTB damage.
Collapse
Affiliation(s)
- Hao Li
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China
| | - Yi-Fan Hu
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China; Wuxi Maternity and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214000, China
| | - Xin-Run Wang
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China
| | - Kong-Wen Ouyang
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China; Wuxi Maternity and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214000, China
| | - Hua Wang
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China; Department of Respiratory Medicine, Anhui Provincial Children's Hospital, Hefei, 230000, China
| | - Kai-Wen Wang
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China
| | - Wei Chang
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China
| | - Jin Zhang
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China
| | - Zhi Yuan
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China
| | - Yong-Wei Xiong
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China
| | - Hua-Long Zhu
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China
| | - Lan Yang
- Wuxi Maternity and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214000, China.
| | - Hua Wang
- Department of Toxicology, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, 230000, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, 230000, China.
| |
Collapse
|
22
|
Shimojima Y, Ichikawa T, Kishida D, Takamatsu R, Sekijima Y. HLA-E-expressing macrophage polarization and increased NKG2A/CD94 expression in adult-onset Still's disease. Immunol Res 2024; 72:1108-1119. [PMID: 38958876 DOI: 10.1007/s12026-024-09512-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024]
Abstract
We investigated the phenotypic characteristics of human leukocyte antigen (HLA)-E-expressing macrophages, NKG2A/CD94 expression in T and natural killer (NK) cells, and their interactions in patients with adult-onset Still's disease (AOSD). Peripheral blood mononuclear cells from 22 patients with AOSD and 22 healthy controls (HC) were used. Isolated monocytes were cultured first with macrophage colony-stimulating factor to differentiate into M0 macrophages and subsequently with lipopolysaccharide/interferon-γ or interleukin-4 to differentiate into M1 or M2 macrophages, respectively. HLA-E and NKG2A/CD94 expression levels were evaluated using quantitative RT-PCR and flow cytometry. HLA-E expression in M0 and M2 macrophages was significantly higher in patients with AOSD than in HC, and was positively correlated with serum C-reactive protein levels and erythrocyte sedimentation rate. NKG2A/CD94 expression in CD4 + and CD8 + T cells was significantly higher in patients with AOSD than in HC, but that in NK cells was not significantly different. In patients with AOSD, NKG2A expression in CD4 + T cells positively correlated with HLA-E expression in M0, M1, and M2 macrophages. CD94 expression in CD8 + T cells inversely correlated with HLA-E expression in M1 and M2 macrophages. NKG2A and CD94 expression in NK cells inversely correlated with HLA-E expression in M0, M1, and M2 macrophages. No significant correlation was observed between HLA-E and NKG2A/CD94 expression in HC. Increased expression of HLA-E in macrophages and NKG2A/CD94 in T cells can be observed in the inflammatory condition of AOSD. HLA-E-expressing macrophages may be associated with NKG2A/CD94 expression in T and NK cells with different correlations.
Collapse
Affiliation(s)
- Yasuhiro Shimojima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
| | - Takanori Ichikawa
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Dai Kishida
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Ryota Takamatsu
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Yoshiki Sekijima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| |
Collapse
|
23
|
Campitiello R, Soldano S, Gotelli E, Hysa E, Montagna P, Casabella A, Paolino S, Pizzorni C, Sulli A, Smith V, Cutolo M. The intervention of macrophages in progressive fibrosis characterizing systemic sclerosis: A systematic review. Autoimmun Rev 2024; 23:103637. [PMID: 39255852 DOI: 10.1016/j.autrev.2024.103637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND AND AIM Systemic sclerosis (SSc) is an immune mediated connective tissue disease characterized by microvascular dysfunction, aberrant immune response, and progressive fibrosis. Although the immuno-pathophysiological mechanisms underlying SSc are not fully clarified, they are often associated with a dysfunctional macrophage activation toward an alternative (M2) phenotype induced by cytokines [i.e., IL-4, IL-10, IL-13, and transforming growth factor (TGF-β)] involved in the fibrotic and anti-inflammatory process. A spectrum of macrophage activation state has been identified ranging from M1 to M2 phenotype, gene expression of phenotype markers, and functional aspects. This systematic review aims to analyze the importance of M2 macrophage polatization during the immune mediated process and the identification of specific pathways, cytokines, and chemokines involved in SSc pathogenesis. Moreover, this review provides an overview on the in vitro and in vivo studies aiming to test therapeutic strategies targeting M2 macrophages. METHODS A systematic literature review was performed according to the preferred Reported Items for Systematic Reviews and Meta-Analyses (PRISMA). The search encompassed the online medical databases PubMed and Embase up to the 30th of June 2024. Original research manuscripts (in vitro study, in vivo study), animal model and human cohort, were considered for the review. Exclusion criteria encompassed reviews, case reports, correspondences, and conference abstracts/posters. The eligible manuscripts main findings were critically analyzed, discussed, and summarized in the correspondent tables. RESULTS Out of the 77 screened abstracts, 49 papers were deemed eligible. Following a critical analysis, they were categorized according to the primary (29 original articles) and secondary (20 original articles) research objectives of this systematic review. The data from the present systematic review suggest the pivotal role of M2 macrophages differentiation and activation together with the dysregulation of the immune system in the SSc pathogenesis. Strong correlations have been found between M2 macrophage presence and clinical manifestations in both murine and human tissue samples. Interestingly, the presence of M2 cell surface markers on peripheral blood monocytes has been highlighted, suggesting a potential biomarker role for this finding. Therapeutic effects reducing M2 macrophage activities have been observed and/or tested for existing and for new drugs, demonstrating potential efficacy in modulating the pro-fibrotic immune response for treatment of SSc. CONCLUSIONS The increased M2 macrophage activation in course of SSc seems to offer new insights on the self-amplifying inflammatory and fibrotic response by the immune system on such disease. Therefore, the revaluation of immunomodulatory and ongoing antifibrotic therapies, as well as novel therapeutical approaches in SSc that contribute to limit the M2 macrophage activation are matter of intense investigations.
Collapse
Affiliation(s)
- Rosanna Campitiello
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Stefano Soldano
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy.
| | - Emanuele Gotelli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy
| | - Elvis Hysa
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Paola Montagna
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy.
| | - Andrea Casabella
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Sabrina Paolino
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Carmen Pizzorni
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Alberto Sulli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Vanessa Smith
- Department of Rheumatology, Ghent University Hospital, University of Ghent, Ghent, Belgium; Department of Internal Medicine, Ghent University Hospital, University of Ghent, Ghent, Belgium; Unit for Molecular Immunology and Inflammation, Flemish Institute for Biotechnology, Inflammation Research Center, Ghent, Belgium.
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
24
|
Zhao X, Wang M, Zhang Y, Zhang Y, Tang H, Yue H, Zhang L, Song D. Macrophages in the inflammatory response to endotoxic shock. Immun Inflamm Dis 2024; 12:e70027. [PMID: 39387442 PMCID: PMC11465138 DOI: 10.1002/iid3.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/24/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Endotoxic shock, particularly prevalent in intensive care units, represents a significant medical challenge. Endotoxin, upon invading the host, triggers intricate interactions with the innate immune system, particularly macrophages. This activation leads to the production of inflammatory mediators such as tumor necrosis factor-alpha, interleukin-6, and interleukin-1-beta, as well as aberrant activation of the nuclear factor-kappa-B and mitogen-activated protein kinase signaling pathways. OBJECTIVE This review delves into the intricate inflammatory cascades underpinning endotoxic shock, with a particular focus on the pivotal role of macrophages. It aims to elucidate the clinical implications of these processes and offer insights into potential therapeutic strategies. RESULTS Macrophages, central to immune regulation, manifest in two distinct subsets: M1 (classically activated subtype) macrophages and M2 (alternatively activated subtype) macrophages. The former exhibit an inflammatory phenotype, while the latter adopt an anti-inflammatory role. By modulating the inflammatory response in patients with endotoxic shock, these macrophages play a crucial role in restoring immune balance and facilitating recovery. CONCLUSION Macrophages undergo dynamic changes within the immune system, orchestrating essential processes for maintaining tissue homeostasis. A deeper comprehension of the mechanisms governing macrophage-mediated inflammation lays the groundwork for an anti-inflammatory, targeted approach to treating endotoxic shock. This understanding can significantly contribute to the development of more effective therapeutic interventions.
Collapse
Affiliation(s)
- Xinjie Zhao
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
- School of MedicineXizang Minzu UniversityXianyangChina
| | - Mengjie Wang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| | - Yanru Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| | - Yiyi Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| | - Haojie Tang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| | - Hongyi Yue
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| | - Li Zhang
- Affiliated Hospital of Xizang Minzu UniversityXianyangShaanxiChina
| | - Dan Song
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of MedicineXizang Minzu UniversityXianyangShaanxiChina
| |
Collapse
|
25
|
Du Y, Zhang Y, Jiang Z, Xu L, Ru J, Wei S, Chen W, Dong R, Zhang S, Jia T. Triptolide alleviates acute gouty arthritis caused by monosodium urate crystals by modulating macrophage polarization and neutrophil activity. Immunol Lett 2024; 269:106907. [PMID: 39122094 DOI: 10.1016/j.imlet.2024.106907] [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: 01/30/2024] [Revised: 07/16/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
The present study focused on the efficacy and role of triptolide (TPL) in relieving symptoms of acute gouty arthritis (AGA) in vivo and in vitro. The effects of TPL in AGA were investigated in monosodium urate (MSU)-treated rat ankles, RAW264.7 macrophages, and neutrophils isolated from mouse peritoneal cavity. Observation of pathological changes in the ankle joint of rats. Enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction (RT-qPCR) were performed to detect the expression levels of inflammatory factors and chemokines. The levels of the indicators of macrophage M1/M2 polarization, and the mechanistic targets of Akt and rapamycin complex 2, were determined via western blotting and RT-qPCR. The expression levels of CD86 and CD206 were detected using immunohistochemistry. Neutrophil migration was observed via air pouch experiments in vivo and Transwell cell migration assay in vitro. Myeloperoxidase (MPO) and Neutrophil elastase (NE) release was analyzed by via immunohistochemistry and immunofluorescence. The expression levels of beclin-1, LC3B, Bax, Bcl-2, and cleaved caspase-3 in neutrophils were determined via western blotting and immunofluorescence. Neutrophil apoptosis was detected using the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Our results suggest that TPL inhibited inflammatory cell infiltration in rat ankle joints and inflammatory factor and chemokine secretion in rat serum, regulated macrophage polarization through the PI3K/AKT signaling pathway, suppressed inflammatory factor and chemokine expression in neutrophils, and inhibited neutrophil migration, neutrophil extracellular trap formation, transitional autophagy, and apoptosis. This suggests that TPL can prevent and treat MSU-induced AGA by regulating macrophage polarization through the PI3K/Akt pathway and modulating neutrophil activity.
Collapse
Affiliation(s)
- Yan Du
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Yurong Zhang
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Zhuxin Jiang
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Lianjie Xu
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Jing Ru
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Shanshan Wei
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Wenhui Chen
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China; Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, Yunnan 450500, , China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 450500, , China
| | - Renjie Dong
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Shan Zhang
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China; Yunnan Provincial Key Laboratory of Molecular Biology for Sinomedicine, Kunming, Yunnan 450500, , China; Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 450500, , China.
| | - Tao Jia
- Department of Orthopedics, First Clinical Medical College of Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650021, , China.
| |
Collapse
|
26
|
Bakery HH, Hussein HAA, Ahmed OM, Abuelsaad ASA, Khalil RG. The potential therapeutic role of IL-35 in pathophysiological processes in type 1 diabetes mellitus. Cytokine 2024; 182:156732. [PMID: 39126765 DOI: 10.1016/j.cyto.2024.156732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/01/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
A chronic autoimmune condition known as type 1 diabetes mellitus (T1DM) has characteristics marked by a gradual immune-mediated deterioration of the β-cells that produce insulin and causes overt hyperglycemia. it affects more than 1.2 million kids and teenagers (0-19 years old). In both, the initiation and elimination phases of T1DM, cytokine-mediated immunity is crucial in controlling inflammation. T regulatory (Treg) cells, a crucial anti-inflammatory CD4+ T cell subset, secretes interleukin-35 (IL-35). The IL-35 has immunomodulatory properties by inhibiting pro-inflammatory cells and cytokines, increasing the secretion of interleukin-10 (IL-10) as well as transforming Growth Factor- β (TGF-β), along with stimulating the Treg and B regulatory (Breg) cells. IL-35, it is a possible target for cutting-edge therapies for cancers, inflammatory, infectious, and autoimmune diseases, including TIDM. Unanswered questions surround IL-35's function in T1DM. Increasing data suggests Treg cells play a crucial role in avoiding autoimmune T1DM. Throughout this review, we will explain the biological impacts of IL-35 and highlight the most recently progresses in the roles of IL-35 in treatment of T1DM; the knowledge gathered from these findings might lead to the development of new T1DM treatments. This review demonstrates the potential of IL-35 as an effective autoimmune diabetes inhibitor and points to its potential therapeutic value in T1DM clinical trials.
Collapse
Affiliation(s)
- Heba H Bakery
- Immunology Division, Faculty of Science, Beni-Suef University, Egypt
| | - Heba A A Hussein
- Faculty of Medicine, Egyptian Fellowship of Radiology, Beni-Suef University, Egypt
| | - Osama M Ahmed
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Egypt
| | | | - Rehab G Khalil
- Immunology Division, Faculty of Science, Beni-Suef University, Egypt.
| |
Collapse
|
27
|
Zhu W, Qiong D, Changzhi X, Meiyu J, Hui L. Macrophage polarization regulation shed lights on immunotherapy for CaOx kidney stone disease. Biomed Pharmacother 2024; 179:117336. [PMID: 39180792 DOI: 10.1016/j.biopha.2024.117336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024] Open
Abstract
Kidney stone disease (KSD) is a major public health concern associated with high morbidity and recurrence, places a significant burden on the health care system worldwide. Calcium oxalate (CaOx) alone or a mixture of CaOx and calcium phosphate stones accounting for more than 80 % of cases. However, beyond surgical removal, the prevention and reduction of recurrence of CaOx kidney stones have always been a challenge. Given that macrophages are traditional innate immune cells that play critical roles in the clearance of pathogens and the maintenance of tissue homeostasis, which have gained more and more interests in nephrolithiasis. Several studies recently clearly demonstrated that M2-macrophage could reduce the renal calcium oxalate (CaOx) crystal acumination, and provide premise insights and therapeutic options for KSD by modulating the macrophage phenotypes. However, the mechanism of macrophage-polarization regulation and that effects on kidney stone prevention and treatments are far from clear. Here, we comprehensively reviewed the literatures related to cytokines, epigenetic modifications and metabolic reprograming of macrophage in CaOx kidney stone disease, aimed to provide better understandings on macrophage polarization regulation as well as its potential clinical applications in CaOx kidney stone disease treatments and prevention.
Collapse
Affiliation(s)
- Wang Zhu
- Department of Urology, The People's Hospital of Longhua, Shenzhen 518109, Guangdong, China.
| | - Deng Qiong
- Department of Urology, The People's Hospital of Longhua, Shenzhen 518109, Guangdong, China
| | - Xu Changzhi
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jin Meiyu
- Department of Urology, The People's Hospital of Longhua, Shenzhen 518109, Guangdong, China
| | - Liang Hui
- Department of Urology, The People's Hospital of Longhua, Shenzhen 518109, Guangdong, China.
| |
Collapse
|
28
|
Song J, Peng D, Peng Y, Zhao G, Ren Y, Guo L, Ren L, Zhang X, Xie X, Zhang Y, Cao L, Li Y. The new pattern for dual NOTCH pathway involving nuclear transcription and mitochondrial regulation supports therapeutic mechanism of 4-butyl benzophenone derivatives against SIRS. Free Radic Biol Med 2024; 223:306-324. [PMID: 39134162 DOI: 10.1016/j.freeradbiomed.2024.07.036] [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: 04/12/2024] [Revised: 07/01/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024]
Abstract
The systemic inflammatory response syndrome (SIRS) represents a self-amplifying cascade of inflammatory reactions and pathophysiological states triggered by infectious or non-infectious factors. The identification of disease targets and differential proteins in the liver (the unique and important immune organ) of SIRS mice treated with the lead compound D1 was conducted using the Genecards database and proteomic analysis, respectively. Subsequently, NOTCH1 was identified as the potential hub target via an intersection analysis between the aforementioned differentially expressed proteins and disease targets. Based on our previous research on the structure-activity relationship, we designed and synthesized a series of SIRS-related derivatives, wherein butyl, halogen, and ester groups were incorporated into benzophenone, aiming at exploring the anti-inflammatory protective action from the perspective of macrophage polarization. Notably, these derivatives exhibited a direct binding capability to the O-glucosylation site (SER496) or its vicinities (such as SER492, VAL485) of NOTCH1 using docking, SPR, DARTS, and CETSA techniques. Mechanistically, derivative D6 exerted anti-inflammatory effects via the dual NOTCH pathway. Firstly, it could inhibit NOTCH1 nuclear transcriptional activity, attenuate the interaction between NICD and RBPJK, concurrently suppress NF-κB and NLRP3 inflammasome (NLRP3, ASC, and cleaved CASP1) activation, and promote NICD (NOTCH1 active fragments) ubiquitination metabolism (the nuclear transcriptional pathway). Secondly, it might possess the ability to increase PGC1α level, subsequently, enhance ATP and MMP levels, mitigate ROS production, increase mitochondrial numbers, and ameliorate mitochondrial inflammatory damage (the mitochondrial pathway). Importantly, the activator Jagged1 could effectively reverse the aforementioned effects, while the inhibitor DAPT exhibited a synergistic effect, suggesting that the nuclear transcriptional regulation and mitochondrial regulation were both in a NOTCH1-dependent manner. Subsequently, it effectively alleviated the inflammatory response and preserved organ function as evidenced by up-regulating M2-type macrophage-related anti-inflammatory cytokines (IL10, TGFβ, CD206, and ARG1) and down-regulating M1-type macrophage-related pro-inflammatory cytokines (NO, IL6, IL18, iNOS, TNFα, CD86, and IL1β). In a word, derivative D6 modulated macrophage polarization and effectively mitigated SIRS by targeting inhibition of the dual NOTCH pathway.
Collapse
Affiliation(s)
- Jiayu Song
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Dan Peng
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yu Peng
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Guang Zhao
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yuan Ren
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lina Guo
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Luyao Ren
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Xiaohui Zhang
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Xiaoxia Xie
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yajie Zhang
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lingya Cao
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yunlan Li
- School of Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, PR China; School of Public Health, Shaanxi University of Chinese Medicine, Xi'an 712046, PR China.
| |
Collapse
|
29
|
Cerdeira CD, Brigagão MRPL. Targeting Macrophage Polarization in Infectious Diseases: M1/M2 Functional Profiles, Immune Signaling and Microbial Virulence Factors. Immunol Invest 2024; 53:1030-1091. [PMID: 38913937 DOI: 10.1080/08820139.2024.2367682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
INTRODUCTION An event of increasing interest during host-pathogen interactions is the polarization of patrolling/naive monocytes (MOs) into macrophage subsets (MФs). Therapeutic strategies aimed at modulating this event are under investigation. METHODS This review focuses on the mechanisms of induction/development and profile of MФs polarized toward classically proinflammatory (M1) or alternatively anti-inflammatory (M2) phenotypes in response to bacteria, fungi, parasites, and viruses. RESULTS AND DISCUSSION It highlights nuclear, cytoplasmic, and cell surface receptors (pattern recognition receptors/PPRs), microenvironmental mediators, and immune signaling. MФs polarize into phenotypes: M1 MФs, activated by IFN-γ, pathogen-associated molecular patterns (PAMPs, e.g. lipopolysaccharide) and membrane-bound PPRs ligands (TLRs/CLRs ligands); or M2 MФs, induced by interleukins (ILs-4, -10 and -13), antigen-antibody complexes, and helminth PAMPs. Polarization toward M1 and M2 profiles evolve in a pathogen-specific manner, with or without canonicity, and can vary widely. Ultimately, this can result in varying degrees of host protection or more severe disease outcome. On the one hand, the host is driving effective MФs polarization (M1 or M2); but on the other hand, microorganisms may skew the polarization through virulence factors to increase pathogenicity. Cellular/genomic reprogramming also ensures plasticity of M1/M2 phenotypes. Because modulation of polarization can occur at multiple points, new insights and emerging perspectives may have clinical implications during the inflammation-to-resolution transition; translated into practical applications as for therapeutic/vaccine design target to boost microbicidal response (M1, e.g. triggering oxidative burst) with specifics PAMPs/IFN-γ or promote tissue repair (M2, increasing arginase activity) via immunotherapy.
Collapse
|
30
|
Cai T, Dai J, Lin Y, Bai Z, Li J, Meng W. N-acetyltransferase 10 affects the proliferation of intrahepatic cholangiocarcinoma and M2-type polarization of macrophages by regulating C-C motif chemokine ligand 2. J Transl Med 2024; 22:875. [PMID: 39350174 PMCID: PMC11440763 DOI: 10.1186/s12967-024-05664-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND N-acetyltransferase 10 (NAT10) plays a crucial role in the occurrence and development of various tumors. However, the current regulatory mechanism of NAT10 in tumors is limited to its presence in tumor cells. Here, we aimed to reveal the role of NAT10 in intrahepatic cholangiocarcinoma (ICC) and investigate its effect on macrophage polarization in the tumor microenvironment (TME). METHODS The correlation between NAT10 and ICC clinicopathology was analyzed using tissue microarray (TMA), while the effect of NAT10 on ICC proliferation was verified in vitro and in vivo. Additionally, the downstream target of NAT10, C-C motif chemokine ligand 2 (CCL2), was identified by Oxford Nanopore Technologies full-length transcriptome sequencing, RNA immunoprecipitation-quantitative polymerase chain reaction, and coimmunoprecipitation experiments. It was confirmed by co-culture that ICC cells could polarize macrophages towards M2 type through the influence of NAT10 on CCL2 protein expression level. Through RNA-sequencing, molecular docking, and surface plasmon resonance (SPR) assays, it was confirmed that berberine (BBR) can specifically bind CCL2 to inhibit ICC development. RESULTS High expression level of NAT10 was associated with poor clinicopathological manifestations of ICC. In vitro, the knockdown of NAT10 inhibited the proliferative activity of ICC cells and tumor growth in vivo, while its overexpression promoted ICC proliferation. Mechanically, by binding to CCL2 messenger RNA, NAT10 increased CCL2 protein expression level in ICC and their extracellular matrix, thereby promoting the proliferation of ICC cells and M2-type polarization of macrophages. BBR can target CCL2, inhibit ICC proliferation, and reduce M2-type polarization of macrophages. CONCLUSIONS NAT10 promotes ICC proliferation and M2-type polarization of macrophages by up-regulating CCL2, whereas BBR inhibits ICC proliferation and M2-type polarization of macrophages by inhibiting CCL2.
Collapse
Affiliation(s)
- Teng Cai
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637600, China
| | - Jianye Dai
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Yanyan Lin
- The Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Zhongtian Bai
- The Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
- Gansu Province Key Laboratory Biotherapy and Regenerative Medicine, Lanzhou, 730000, China.
| | - Jingdong Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637600, China.
| | - Wenbo Meng
- The Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
- Gansu Province Key Laboratory Biotherapy and Regenerative Medicine, Lanzhou, 730000, China.
| |
Collapse
|
31
|
Yan Q, Liu H, Zhu R, Zhang Z. Contribution of macrophage polarization in bone metabolism: A literature review. Cytokine 2024; 184:156768. [PMID: 39340960 DOI: 10.1016/j.cyto.2024.156768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Macrophage polarization divides macrophages into two main cell subpopulations, classically and alternatively activated macrophages (M1 and M2, respectively). M1 polarization promotes osteoclastogenesis, while M2 polarization promotes osteogenesis. The physiological homeostasis of bone metabolism involves a high dynamic balance between osteoclastic-mediated bone resorption and formation. Reportedly, M1/M2 imbalance causes the onset and persistence of inflammation-related bone diseases. Therefore, understanding the research advances in functions and roles of macrophages in such diseases will provide substantial guidance for improved treatment of bone diseases. In this review, we underscore and summarize the research advances in macrophage polarization, and bone-related diseases, such as rheumatoid arthritis, osteoarthritis, and osteoporosis, over the last 5 years. Our findings showed that targeting macrophages and balancing macrophage polarization can effectively reduce inflammation and decrease bone destruction while promoting bone formation and vascular repair. These results indicate that regulating macrophage and macrophage polarization to restore homeostasis is a prospective approach for curing bone-related diseases.
Collapse
Affiliation(s)
- Qiqi Yan
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Haixia Liu
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ruyuan Zhu
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Zhiguo Zhang
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
32
|
Kang M, Jia H, Feng M, Ren H, Gao J, Liu Y, Zhang L, Zhou MS. Cardiac macrophages in maintaining heart homeostasis and regulating ventricular remodeling of heart diseases. Front Immunol 2024; 15:1467089. [PMID: 39372400 PMCID: PMC11449765 DOI: 10.3389/fimmu.2024.1467089] [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: 07/19/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Macrophages are most important immune cell population in the heart. Cardiac macrophages have broad-spectrum and heterogeneity, with two extreme polarization phenotypes: M1 pro-inflammatory macrophages (CCR2-ly6Chi) and M2 anti-inflammatory macrophages (CCR2-ly6Clo). Cardiac macrophages can reshape their polarization states or phenotypes to adapt to their surrounding microenvironment by altering metabolic reprogramming. The phenotypes and polarization states of cardiac macrophages can be defined by specific signature markers on the cell surface, including tumor necrosis factor α, interleukin (IL)-1β, inducible nitric oxide synthase (iNOS), C-C chemokine receptor type (CCR)2, IL-4 and arginase (Arg)1, among them, CCR2+/- is one of most important markers which is used to distinguish between resident and non-resident cardiac macrophage as well as macrophage polarization states. Dedicated balance between M1 and M2 cardiac macrophages are crucial for maintaining heart development and cardiac functional and electric homeostasis, and imbalance between macrophage phenotypes may result in heart ventricular remodeling and various heart diseases. The therapy aiming at specific target on macrophage phenotype is a promising strategy for treatment of heart diseases. In this article, we comprehensively review cardiac macrophage phenotype, metabolic reprogramming, and their role in maintaining heart health and mediating ventricular remodeling and potential therapeutic strategy in heart diseases.
Collapse
Affiliation(s)
- Mengjie Kang
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Hui Jia
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
- School of Traditional Chinese Medicine, Shenyang Medical College, Shenyang, China
| | - Mei Feng
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Haolin Ren
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Junjia Gao
- Department of Cardiology, Second Affiliated Hospital, Shenyang Medical College, Shenyang, China
| | - Yueyang Liu
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
- School of Pharmacy, Shenyang Medical College, Shenyang, China
| | - Lu Zhang
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| | - Ming-Sheng Zhou
- Science and Experiment Research Center, Shenyang Medical College & Shenyang Key Laboratory of Vascular Biology, Science and Experimental Research Center, Shenyang Medical College, Shenyang, China
| |
Collapse
|
33
|
Li C, Yu X, Han X, Lian C, Wang Z, Shao S, Shao F, Wang H, Ma S, Liu J. Innate immune cells in tumor microenvironment: A new frontier in cancer immunotherapy. iScience 2024; 27:110750. [PMID: 39280627 PMCID: PMC11399700 DOI: 10.1016/j.isci.2024.110750] [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] [Indexed: 09/18/2024] Open
Abstract
Innate immune cells, crucial in resisting infections and initiating adaptive immunity, play diverse and significant roles in tumor development. These cells, including macrophages, granulocytes, dendritic cells (DCs), innate lymphoid cells, and innate-like T cells, are pivotal in the tumor microenvironment (TME). Innate immune cells are crucial components of the TME, based on which various immunotherapy strategies have been explored. Immunotherapy strategies, such as novel immune checkpoint inhibitors, STING/CD40 agonists, macrophage-based surface backpack anchoring, ex vivo polarization approaches, DC-based tumor vaccines, and CAR-engineered innate immune cells, aim to enhance their anti-tumor potential and counteract cancer-induced immunosuppression. The proximity of innate immune cells to tumor cells in the TME also makes them excellent drug carriers. In this review, we will first provide a systematic overview of innate immune cells within the TME and then discuss innate cell-based therapeutic strategies. Furthermore, the research obstacles and perspectives within the field will also be addressed.
Collapse
Affiliation(s)
- Changhui Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyu Yu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyan Han
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Chen Lian
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Zijin Wang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Fangwei Shao
- National Key Laboratory of Biobased Transportation Fuel Technology, ZJU-UIUC Institute, Zhejiang University, Hangzhou 310027, China
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Shenglin Ma
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou 310002, China
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
34
|
Kerns S, Owen KA, Daamen A, Kain J, Grammer AC, Lipsky PE. Genetic association with autoimmune diseases identifies molecular mechanisms of coronary artery disease. iScience 2024; 27:110715. [PMID: 39262791 PMCID: PMC11387803 DOI: 10.1016/j.isci.2024.110715] [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: 06/04/2024] [Revised: 06/28/2024] [Accepted: 08/08/2024] [Indexed: 09/13/2024] Open
Abstract
Autoimmune patients have a significantly increased risk of developing coronary artery disease (CAD) compared to the general population. However, autoimmune patients often lack traditional risk factors for CAD and there is increasing recognition of inflammation in CAD development. In this study, we leveraged genome-wide association study (GWAS) data to understand whether there is a genetic relationship between CAD and autoimmunity. Statistical genetic comparison methods were used to identify correlated and causal SNPs between various autoimmune diseases and CAD. Pleiotropic SNPs were identified by cross-phenotype association analysis (CPASSOC) and overlap between GWAS. Causal SNPs were identified using Mendelian Randomization (MR) and Colocalization (COLOC). Using SNP-to-gene mapping, we additionally identified pleiotropic and causal genes and pathways associated between autoimmunity and CAD, which were contextualized by documentation of enrichment in individual cell types identified from coronary atherosclerotic plaques by single-cell RNA sequencing. These results provide insight into potential inflammatory therapeutic targets for CAD.
Collapse
Affiliation(s)
- Sophia Kerns
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Katherine A Owen
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Andrea Daamen
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Jessica Kain
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
- Stanford University Department of Genetics, Stanford, CA 94305, USA
| | - Amrie C Grammer
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Peter E Lipsky
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| |
Collapse
|
35
|
Dong T, Barasa L, Yu X, Ouyang W, Shao L, Quan C, Wang SH, Zhang J, Salmon M, Tsung A, Alam HB, Ma J, Thompson PR, Li Y. AFM41a: A Novel PAD2 Inhibitor for Sepsis Treatment-Efficacy and Mechanism. Int J Biol Sci 2024; 20:5043-5055. [PMID: 39430255 PMCID: PMC11488583 DOI: 10.7150/ijbs.97166] [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: 04/10/2024] [Accepted: 09/05/2024] [Indexed: 10/22/2024] Open
Abstract
Pseudomonas aeruginosa (PA) infection can cause pneumonia and sepsis by activating peptidyl-arginine deiminase (PAD) and triggering the formation of neutrophil extracellular traps (NETs). Our previous research has elucidated the crucial role of PAD2 in regulating CitH3 production and NETosis signaling following bacterial infection. Therefore, targeting PAD2 with selective inhibitors holds promise for treating PA-induced sepsis. Here, we compare the structure and function of two PAD2 inhibitors, AFM32a and AFM41a, and investigate their biological effects in mice subjected with PA. We analyze their impact on PAD2 inhibition, macrophage polarization, and other host defense mechanisms against PA-induced sepsis utilizing both in vivo and in vitro approaches. Our findings demonstrate that both PAD2 inhibitors (AFM32a and AFM41a) and Pad2 deficiency substantially enhance protection against PA-induced sepsis, with AFM41a showing superior efficacy over AFM32a. This protective effect is marked by improved survival rates, reduced bacterial growth in mice subjected to PA infection, and the promotion of M2 macrophage polarization coupled with enhanced autophagic activity. Our results advocate for targeting PAD2 as an effective strategy to bolster host defenses against PA infection. Utilizing AFM41a to promote M2 macrophage polarization and autophagy offers promising avenues for the treatment of PA infection and the improvement of sepsis outcomes.
Collapse
Affiliation(s)
- Tao Dong
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Leonard Barasa
- Program in Chemical Biology, Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Xin Yu
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, Zhejiang Province, China
| | - Wenlu Ouyang
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Changsha, China
| | - Liujiazi Shao
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chao Quan
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
- Department of Urology, The Xiangya Hospital, Changsha, China
| | - Su He Wang
- Internal Medicine, Division of Allergy, University of Michigan Health System, Ann Arbor, MI, USA
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Morgan Salmon
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Allan Tsung
- Department of Surgery, Division of Surgical Science, University of Virginia, Charlottesville, VA 22903, USA
| | - Hasan B. Alam
- Department of Surgery, Northwestern University, Arkes Pavilion, 676 N St Clair St Ste 2320, Chicago, IL 60611, USA
| | - Jianjie Ma
- Department of Surgery, Division of Surgical Science, University of Virginia, Charlottesville, VA 22903, USA
| | - Paul R. Thompson
- Program in Chemical Biology, Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Yongqing Li
- Department of Surgery, University of Michigan Health System, Ann Arbor, MI, USA
| |
Collapse
|
36
|
Shao Y, Han S, Hou Z, Yang C, Zhao Y. Tumor-associated macrophages within the immunological milieu: An emerging focal point for therapeutic intervention. Heliyon 2024; 10:e36839. [PMID: 39281573 PMCID: PMC11401039 DOI: 10.1016/j.heliyon.2024.e36839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
Tumor-associated macrophages play an important role in the tumor immune microenvironment, and regulating the function of tumor-associated macrophages has important therapeutic potential in tumor therapy. Mature macrophages could migrate to the tumor microenvironment, influencing multiple factors such as tumor cell proliferation, invasion, metastasis, extracellular matrix remodeling, immune suppression, and drug resistance. As a major component of the tumor microenvironment, tumor-associated macrophages crosstalk with other immune cells. Currently, tumor-associated macrophages have garnered considerable attention in tumor therapy, broadening the spectrum of drug selection to some extent, thereby aiding in mitigating the prevailing clinical drug resistance dilemma. This article summarizes the recent advances in tumor-associated macrophages concerning immunology, drug targeting mechanisms for tumor-associated macrophages treatment, new developments, and existing challenges, offering insights for future therapeutic approaches. In addition, this paper summarized the impact of tumor-associated macrophages on current clinical therapies, discussed the advantages and disadvantages of targeted tumor-associated macrophages therapy compared with existing tumor therapies, and predicted and discussed the future role of targeted tumor-associated macrophages therapy and the issues that need to be focused on.
Collapse
Affiliation(s)
- Yanchi Shao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Song Han
- The First Hospital of Jilin University, Changchun, China
| | - Zhenxin Hou
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Chen Yang
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yanbin Zhao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
37
|
Ma R, Wang A, Yang M, Huang Z, Liu G, Wei Q, Lu Y, Wei H, Wang J, Tang Q, Pu J. Hsa_circ_0000092 up-regulates IL24 by SMC1A to induce macrophages M2 polarization. Heliyon 2024; 10:e36517. [PMID: 39296099 PMCID: PMC11408814 DOI: 10.1016/j.heliyon.2024.e36517] [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: 10/11/2023] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/21/2024] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) as the malignant cancers with high morbidity. The EMT of HCC has closely linked to the metastasis and recurrence. Moreover, tumor-associated macrophages (TAMs) can interact with HCC cells in the immune microenvironment; the M2 polarization of TAMs enhance the HCC cells EMT. The mechanism between HCC cells and TAMs is still unclear and our study was aimed to uncover it. Methods We performed RT-qPCR and western to detach the RNA and protein expression. The relationship among has_circ_0000092, U2AF2, SMC1A and IL24 were revealed through mechanism experiments. Rescue assays were implemented to determine how circ_0000092 modulates M2 polarization of TAMs. Results As detected by RT-qPCR, has_circ_0000092 was with high expression in HCC cells and could recruit U2AF2 to promote transcription of SMC1A. Moreover, circ_0000092 could control macrophage M2 polarization via promoting IL24 expression in HCC cells. Conclusion To conclude, hsa_circ_0000092 can up-regulates IL24 by SMC1A to induce macrophages M2 polarization.
Collapse
Affiliation(s)
- Rihai Ma
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| | - Anmin Wang
- Graduate College of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| | - Meng Yang
- Graduate College of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| | - Zihua Huang
- Graduate College of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| | - Guoman Liu
- Graduate College of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| | - Qing Wei
- Graduate College of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| | - Yuan Lu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| | - Huamei Wei
- Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| | - Jianchu Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| | - Qianli Tang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| | - Jian Pu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Zhuang Autonomous Region, 533000, China
| |
Collapse
|
38
|
Lu J, Zhang J, Jiang H, Hu Z, Zhang Y, He L, Yang J, Xie Y, Wu D, Li H, Zeng K, Tan P, Xiao Q, Song Z, Pan C, Bai X, Yu X. Vangl2 suppresses NF-κB signaling and ameliorates sepsis by targeting p65 for NDP52-mediated autophagic degradation. eLife 2024; 12:RP87935. [PMID: 39269442 PMCID: PMC11398866 DOI: 10.7554/elife.87935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024] Open
Abstract
Van Gogh-like 2 (Vangl2), a core planar cell polarity component, plays an important role in polarized cellular and tissue morphology induction, growth development, and cancer. However, its role in regulating inflammatory responses remains elusive. Here, we report that Vangl2 is upregulated in patients with sepsis and identify Vangl2 as a negative regulator of The nuclear factor-kappaB (NF-κB) signaling by regulating the protein stability and activation of the core transcription component p65. Mice with myeloid-specific deletion of Vangl2 (Vangl2ΔM) are hypersusceptible to lipopolysaccharide (LPS)-induced septic shock. Vangl2-deficient myeloid cells exhibit enhanced phosphorylation and expression of p65, therefore, promoting the secretion of proinflammatory cytokines after LPS stimulation. Mechanistically, NF-κB signaling-induced-Vangl2 recruits E3 ubiquitin ligase PDLIM2 to catalyze K63-linked ubiquitination on p65, which serves as a recognition signal for cargo receptor NDP52-mediated selective autophagic degradation. Taken together, these findings demonstrate Vangl2 as a suppressor of NF-κB-mediated inflammation and provide insights into the crosstalk between autophagy and inflammatory diseases.
Collapse
Affiliation(s)
- Jiansen Lu
- Department of Joint Surgery, the Fifth Affiliated Hospital, Southern Medical UniversityGuangzhouChina
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Jiahuan Zhang
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Huaji Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Orthopaedics, Yuebei People's Hospital Affiliated to Medical College of Shantou UniversityShaoguanChina
| | - Zhiqiang Hu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yufen Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Lian He
- Department of Pharmacology, School of Medicine, Southern University of Science and TechnologyShenzhenChina
- Institute of Biosciences and Technology, College of Medicine, Texas A&M UniversityHoustonUnited States
| | - Jianwu Yang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yingchao Xie
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Dan Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Hongyu Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Ke Zeng
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Peng Tan
- Department of Pharmacology, School of Medicine, Southern University of Science and TechnologyShenzhenChina
- Klarman Cell Observatory, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Qingyue Xiao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Zijing Song
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Chenglong Pan
- Department of Joint Surgery, the Fifth Affiliated Hospital, Southern Medical UniversityGuangzhouChina
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
39
|
Antony F, Kinha D, Nowińska A, Rouse BT, Suryawanshi A. The immunobiology of corneal HSV-1 infection and herpetic stromal keratitis. Clin Microbiol Rev 2024; 37:e0000624. [PMID: 39078136 PMCID: PMC11391706 DOI: 10.1128/cmr.00006-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024] Open
Abstract
SUMMARYHuman alphaherpesvirus 1 (HSV-1) is a highly successful neurotropic pathogen that primarily infects the epithelial cells lining the orofacial mucosa. After primary lytic replication in the oral, ocular, and nasal mucosal epithelial cells, HSV-1 establishes life-long latency in neurons within the trigeminal ganglion. Patients with compromised immune systems experience frequent reactivation of HSV-1 from latency, leading to virus entry in the sensory neurons, followed by anterograde transport and lytic replication at the innervated mucosal epithelial surface. Although recurrent infection of the corneal mucosal surface is rare, it can result in a chronic immuno-inflammatory condition called herpetic stromal keratitis (HSK). HSK leads to gradual vision loss and can cause permanent blindness in severe untreated cases. Currently, there is no cure or successful vaccine to prevent latent or recurrent HSV-1 infections, posing a significant clinical challenge to managing HSK and preventing vision loss. The conventional clinical management of HSK primarily relies on anti-virals to suppress HSV-1 replication, anti-inflammatory drugs (such as corticosteroids) to provide symptomatic relief from pain and inflammation, and surgical interventions in more severe cases to replace damaged cornea. However, each clinical treatment strategy has limitations, such as local and systemic drug toxicities and the emergence of anti-viral-resistant HSV-1 strains. In this review, we summarize the factors and immune cells involved in HSK pathogenesis and highlight alternate therapeutic strategies for successful clinical management of HSK. We also discuss the therapeutic potential of immunoregulatory cytokines and immunometabolism modulators as promising HSK therapies against emerging anti-viral-resistant HSV-1 strains.
Collapse
Affiliation(s)
- Ferrin Antony
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - Divya Kinha
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Anna Nowińska
- Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
- Ophthalmology Department, Railway Hospital in Katowice, Katowice, Poland
| | - Barry T Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Amol Suryawanshi
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
40
|
Xu X, Peng J, Wang N, Ocansey DKW, Zhang X, Mao F. hucMSC-Ex alleviates inflammatory bowel disease in mice by enhancing M2-type macrophage polarization via the METTL3-Slc37a2-YTHDF1 axis. Cell Biol Toxicol 2024; 40:74. [PMID: 39259386 PMCID: PMC11390928 DOI: 10.1007/s10565-024-09921-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
Human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Ex) have emerged as a new treatment strategy for inflammatory bowel disease (IBD) due to their immunoregulatory function. N6-methyladenosine (m6A) plays a crucial role in regulating intestinal immunity, especially in IBD where macrophages play an important role, although its mechanism is not yet fully understood. From this perspective, this research aimed to evaluate the effect of hucMSC-Ex on m6A modification of macrophages in IBD. In the process of alleviating inflammation, hucMSC-Ex promotes macrophage polarization toward the M2 type and regulates intracellular m6A levels by upregulating the expression of m6A "Writer" METTL3 and "Reader" YTHDF1. Solute Carrier Family 37 Member 2 (Slc37a2) was identified by Methylation RNA immunoprecipitation sequencing as the target molecule of the hucMSC-Ex. Mechanically, hucMSC-Ex promoted the binding of METTL3 to the Slc37a2 mRNA complex, and enhanced the binding of Slc37a2 to YTHDF1 to upregulate the intracellular expression of Slc37a2, thereby attenuating the pro-inflammatory function of macrophage. This study confirms the modulatory role of hucMSC-Ex on the m6A modification of macrophages in IBD, providing a new scientific basis for the treatment of IBD with hucMSC-Ex.
Collapse
Affiliation(s)
- Xinwei Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, 222006, Jiangsu, P.R. China
- Clinical Laboratory, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221000, Jiangsu, China
| | - Jianhua Peng
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, 212300, Jiangsu, P.R. China
| | - Naijian Wang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, 222006, Jiangsu, P.R. China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, CC0959347, Cape Coast, Ghana
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China.
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, 222006, Jiangsu, P.R. China.
| |
Collapse
|
41
|
Zhang WB, Chen ZX, Liu Z, Qian XY, Ge YZ, Zhang HY, Xu WT, Shan LT, Zhao DB. PBMC-mediated modulation of macrophage polarization in RAW264.7 cells through STAT1/STAT6 signaling cascades. Int Immunopharmacol 2024; 138:112651. [PMID: 38986303 DOI: 10.1016/j.intimp.2024.112651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
Peripheral blood mononuclear cells (PBMC), sourced autologously, offer numerous advantages when procured: easier acquisition process, no in vitro amplification needed, decreased intervention and overall increased acceptability make PBMC an attractive candidate for cell therapy treatment. However, the exact mechanism by which PBMC treat diseases remains poorly understood. Immune imbalance is the pathological basis of many diseases, with macrophages playing a crucial role in this process. However, research on the role and mechanisms of PBMC in regulating macrophages remains scarce. This study employed an in vitro co-culture model of PBMC and RAW264.7 macrophages to explore the role and mechanisms of PBMC in regulating macrophages. The results showed that the co-culturing led to decreased expression of inflammatory cytokines and increased expression of anti-inflammatory cytokines in RAW264.7 or in the culture supernatant. Additionally, the pro-inflammatory, tissue matrix-degrading M1 macrophages decreased, while the anti-inflammatory, matrix-synthesizing, regenerative M2 macrophages increased in both RAW264.7 and monocytes within PBMC. Moreover, co-cultured macrophages exhibited a significantly decreased p-STAT1/STAT1 ratio, while the p-STAT6/STAT6 ratio significantly increased. This suggests that PBMC may inhibit M1 macrophage polarization by blocking STAT1 signaling cascades and may promote M2 macrophage polarization through the activation of STAT6 signaling cascades. Overall, this study sheds light on the role and mechanism of PBMC in regulating macrophages. Moreover, it was found that monocytes within co-cultured PBMC differentiated into M2 macrophages in the presence of macrophages. This finding provides experimental evidence for the use of PBMC in treating inflammatory diseases, especially macrophage-depleting inflammatory diseases such as osteoarthritis.
Collapse
Affiliation(s)
- Wen-Bo Zhang
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zu-Xiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Zhen Liu
- Department of Anatomy, Naval Medical University, Shanghai 200433, China
| | - Xin-Yu Qian
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan-Zhi Ge
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Hai-Yan Zhang
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Wen-Ting Xu
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Le-Tian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| | - Dong-Bao Zhao
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
42
|
Tail M, Zhang H, Zheng G, Harms AK, Hatami M, Skutella T, Kiening K, Unterberg A, Zweckberger K, Younsi A. Sonic Hedgehog reduces inflammatory response, decreases blood-spinal cord barrier permeability, and improves locomotor function recovery in an acute spinal cord injury rat model. J Inflamm (Lond) 2024; 21:34. [PMID: 39227870 PMCID: PMC11373473 DOI: 10.1186/s12950-024-00404-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Sonic Hedgehog (Shh), extensively researched for its role in early neurogenesis and brain development, has recently been recognized for its neuroprotective potential following neuronal injuries. This study examines the immediate impact of early administered Shh on the local inflammatory response post-acute spinal cord injury in rats. METHODS Thirty-four female Wistar rats underwent either sham surgery (laminectomy; n = 10) or clip compression/contusion spinal cord injury (SCI) at the T9 level. This was followed by implantation of an osmotic pump and a subdural catheter for continuous intrathecal delivery of Shh (n = 12) or placebo (NaCl; n = 12). Locomotor function was assessed at 3- and 7-days post-injury (dpi) using the Basso, Beattie, and Bresnahan (BBB) score and the Gridwalk test. Animals were euthanized after 3 or 7 days for immunohistochemical analysis of the local inflammatory reaction and immune cell migration. RESULTS Shh-treated rats demonstrated significant hindlimb movement and coordination improvements at 7 days post-injury, compared to controls. This enhancement was accompanied by a significant reduction in both immune cell presence and blood plasma products within spinal cord lesions, suggesting Shh's dual role in modulating immune cell migration and maintaining the integrity of the blood-spinal cord barrier. Separately, these Shh-treated rats also showed an increase in M(IL-4) polarization of macrophages, further underlining the potential therapeutic impact of Shh in post-injury recovery. Notably, these effects were not evident at three days post-injury. CONCLUSION Shh application at 7 days post-injury showed immunomodulatory effects, possibly via enhanced blood-spinal cord barrier integrity, reduced immune cell migration, and increased anti-inflammatory immune cell differentiation. These mechanisms collectively contribute to enhanced locomotor recovery.
Collapse
Affiliation(s)
- Mohamed Tail
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
- Department of Neurology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Hao Zhang
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Guoli Zheng
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Anna-Kathrin Harms
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Maryam Hatami
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120, Heidelberg, Germany
| | - Thomas Skutella
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, 69120, Heidelberg, Germany
| | - Karl Kiening
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Klaus Zweckberger
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, Heidelberg University Hospital, 69120, Heidelberg, Germany.
| |
Collapse
|
43
|
Wei Z, Liu C, Liang J, Zhou X, Xue K, Wang K, Zhang X. Characterization of Mitoribosomal Small Subunit unit genes related immune and pharmacogenomic landscapes in renal cell carcinoma. IUBMB Life 2024; 76:647-665. [PMID: 38551358 DOI: 10.1002/iub.2818] [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: 01/16/2024] [Accepted: 02/23/2024] [Indexed: 08/31/2024]
Abstract
Mitoribosomes are essential for the production of biological energy. The Human Mitoribosomal Small Subunit unit (MRPS) family, responsible for encoding mitochondrial ribosomal small subunits, is actively engaged in protein synthesis within the mitochondria. Intriguingly, MRPS family genes appear to play a role in cancer. A multistep process was employed to establish a risk model associated with MRPS genes, aiming to delineate the immune and pharmacogenomic landscapes in clear cell renal cell carcinoma (ccRCC). MRPScores were computed for individual patients to assess their responsiveness to various treatment modalities and their susceptibility to different therapeutic targets and drugs. While MRPS family genes have been implicated in various cancers as oncogenes, our findings reveal a contrasting tumor suppressor role for MRPS genes in ccRCC. Utilizing an MRPS-related risk model, we observed its excellent prognostic capability in predicting survival outcomes for ccRCC patients. Remarkably, the subgroup with high MRPS-related scores (MRPScore) displayed poorer prognosis but exhibited a more robust response to immunotherapy. Through in silico screening of 2183 drug targets and 1646 compounds, we identified two targets (RRM2 and OPRD1) and eight agents (AZ960, carmustine, lasalocid, SGI-1776, AZD8055_1059, BPD.00008900_1998, MK.8776_2046, and XAV939_1268) with potential therapeutic implications for high-MRPScore patients. Our study represents the pioneering effort in proposing that molecular classification, diagnosis, and treatment strategies can be formulated based on MRPScores. Indeed, a high MRPScore profile appears to elevate the risk of tumor progression and mortality, potentially through its influence on immune regulation. This suggests that the MRPS-related risk model holds promise as a prognostic predictor and may offer novel insights into personalized therapeutic strategies.
Collapse
Affiliation(s)
- Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenchen Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaqian Liang
- Department of Urology, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Zhou
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kaming Xue
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Xin M, Xu A, Tian J, Wang L, He Y, Jiang H, Yang B, Li B, Sun Y. Anthocyanins as natural bioactives with anti-hypertensive and atherosclerotic potential: Health benefits and recent advances. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155889. [PMID: 39047414 DOI: 10.1016/j.phymed.2024.155889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/04/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Hypertension is a highly prevalent chronic metabolic illness affecting individuals of all age groups. Furthermore, it is a significant risk factor for the development of atherosclerosis (AS), as a correlation between hypertension and AS has been observed. However, the effective treatments for either of these disorders appear to be uncommon. METHODS A systematic search of articles published in PubMed, Web of Science, ScienceDirect, Scopus, and Google Scholar databases over the last decade was performed using the following keywords: hypertension, AS, anthocyanins, antioxidants, gut microbes, health benefits, and bioactivity. RESULTS The available research indicates that anthocyanin consumption can achieve antioxidant effects by inducing the activation of intracellular nuclear factor erythroid 2-related factor (Nrf2) and the expression of antioxidant genes. Moreover, previous reports showed that anthocyanins can enhance the human body's ability to fight against inflammation and cancer through the inhibition of inflammatory factors and the regulation of related signaling pathways. They can also protect the blood vessels and nervous system by regulating the production and function of endothelial nitric oxide synthase (eNOS). Gut microorganisms play an important role in various chronic diseases. Our research has also investigated the role of anthocyanins in the metabolism of the gut microbiota, leading to significant breakthroughs. This study not only presents a unique strategy for reducing the risk of cardiovascular diseases (CVDs) without the need for medicine but also provides insights into the development and utilization of intestinal probiotic dietary supplements. CONCLUSION In this review, different in vitro and in vivo studies have shown that anthocyanins slow down the onset and progression of hypertension and AS through different mechanisms. In addition, gut microbial metabolites also play a crucial role in diseases through the gut-liver axis.
Collapse
Affiliation(s)
- Meili Xin
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, 110866, China
| | - Aihua Xu
- Department of Rehabilitation Medicine, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, 110866, China
| | - Liang Wang
- Zhejiang Lanmei Technology Co., Ltd., Zhuji, Zhejiang 311800, China
| | - Ying He
- Zhejiang Lanmei Technology Co., Ltd., Zhuji, Zhejiang 311800, China
| | - Hongzhou Jiang
- Anhui Ziyue Biotechnology Co., Ltd, Wuhu, Anhui,241000, China
| | - Baoru Yang
- Food Chemistry and Food Development, Department of Life Technologies, University of Turku, FI-20014 Turun yliopisto, Finland
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, 110866, China.
| | - Yongxin Sun
- Department of Rehabilitation Medicine, The First Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
45
|
Zong YH, Cao JF, Zhao Y, Gao M, Chen WL, Wu M, Xu X, Xu ZY, Zhang XQ, Tang JZ, Liu Y, Hu XS, Wang SQ, Zhang X. Mechanism of Lian Hua Qing Wen capsules regulates the inflammatory response caused by M 1 macrophage based on cellular experiments and computer simulations. Acta Trop 2024; 257:107320. [PMID: 39002739 DOI: 10.1016/j.actatropica.2024.107320] [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: 04/27/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE The polarization of macrophages with the resulting inflammatory response play a crucial part in tissue and organ damage due to inflammatory. Study has proved Lian Hua Qing Wen capsules (LHQW) can reduce activation of inflammatory response and damage of tissue derived from the inflammatory reactions. However, the mechanism of LHQW regulates the macrophage-induced inflammatory response is unclear. Therefore, we investigated the mechanism of LHQW regulated the inflammatory response of M1 macrophages by cellular experiments and computer simulations. METHODS This study has analysed the targets and mechanisms of macrophage regulating inflammatory response at gene and protein levels through bioinformatics. The monomeric components of LHQW were analyzed by High Performance Liquid Chromatography (HPLC). We established the in vitro cell model by M1 macrophages (Induction of THP-1 cells into M1 macrophages). RT-qPCR and immunofluorescence were used to detect changes in gene and protein levels of key targets after LHQW treatment. Computer simulations were utilized to verify the binding stability of monomeric components and protein targets. RESULTS Macrophages had 140,690 gene targets, inflammatory response had 12,192 gene targets, intersection gene targets were 11,772. Key monomeric components (including: Pinocembrin, Fargesone-A, Nodakenin and Bowdichione) of LHQW were screened by HPLC. The results of cellular experiments indicated that LHQW could significantly reduce the mRNA expression of CCR5, CSF2, IFNG and TNF, thereby alleviating the inflammatory response caused by M1 macrophage. The computer simulations further validated the binding stability and conformation of key monomeric components and key protein targets, and IFNG/Nodakenin was able to form the most stable binding conformation for its action. CONCLUSION In this study, the mechanism of LHQW inhibits the polarization of macrophages and the resulting inflammatory response was investigated by computer simulations and cellular experiments. We found that LHQW may not only reduce cell damage and death by acting on TNF and CCR5, but also inhibit the immune recognition process and inflammatory response by regulating CSF2 and IFNG to prevent polarization of macrophages. Therefore, these results suggested that LHQW may act through multiple targets to inhibit the polarization of macrophages and the resulting inflammatory response.
Collapse
Affiliation(s)
| | - Jun-Feng Cao
- College of Medicine, Southwest Jiaotong University, Chengdu, PR China
| | | | - Miao Gao
- Chengdu Medical College, Chengdu, PR China
| | | | - Mei Wu
- Chengdu Medical College, Chengdu, PR China
| | - Xiang Xu
- Chengdu Medical College, Chengdu, PR China
| | | | | | | | - Yulin Liu
- Chengdu Medical College, Chengdu, PR China
| | | | | | - Xiao Zhang
- Chengdu Medical College, Chengdu, PR China.
| |
Collapse
|
46
|
Wang J, Liu JJ, Tang ZY, Liang QQ, Cui JW. Acupuncture promotes neurological recovery and regulates lymphatic function after acute inflammatory nerve root injury. Heliyon 2024; 10:e35702. [PMID: 39229545 PMCID: PMC11369430 DOI: 10.1016/j.heliyon.2024.e35702] [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: 07/19/2023] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 09/05/2024] Open
Abstract
Aims To investigate the therapeutic effect of acupuncture on acute inflammatory nerve root injury by regulating lymphatic function. Main methods A mouse model of L5 nerve root compression was used to simulate acute nerve root injury. After modeling, acupuncture treatment was given each day for one week. Pain thresholds were assessed before and after modeling and treatment. Immunofluorescence staining was performed to observe the distribution astrocytes and neurons in the lumbar spinal cord, the innervation rate of neuromuscular junctions (NMJs), lymphatic endothelial cells (LECs) of lumbar aortic lymph nodes, and the percentage of M1 macrophages. The number of each type of immune cells in the lumbar aortic lymph nodes (LALNs) was measured by flow cytometry. Key findings The model group showed a significant decrease in pain threshold in the affected lower limb, while acupuncture treatment was able to significantly increase it. Acupuncture significantly repaired astrocytes and neurons in the lumbar spinal cord of the compressed segment, increased the innervation rate of nerve endings at NMJs, reduced LECs in the LALNs, reduced the proportion of M1 macrophages in the LALNs, and significantly reduced mononuclear neutrophils and monocytic neutrophils. Significance Acupuncture can reduce pain, promote nerve repair in mice with acute nerve root injury, and suppress immune responses in lumbar aortic lymph nodes.
Collapse
Affiliation(s)
- Jie Wang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian-ju Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Rehabilitation Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai, China
| | - Zhan-ying Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian-qian Liang
- Spine Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jia-wen Cui
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Rehabilitation Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
47
|
Zhang Y, Kang Q, He L, Chan KI, Gu H, Xue W, Zhong Z, Tan W. Exploring the immunometabolic potential of Danggui Buxue Decoction for the treatment of IBD-related colorectal cancer. Chin Med 2024; 19:117. [PMID: 39210410 PMCID: PMC11360867 DOI: 10.1186/s13020-024-00978-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Danggui Buxue (DGBX) decoction is a classical prescription composed of Astragali Radix (AR) and Angelicae Sinensis Radix (ASR), used to enrich blood, and nourish Qi in Chinese medicine, with the potential to recover energy and stimulate metabolism. Chronic inflammation is a risk factor in the development of inflammatory bowel disease (IBD)-related colorectal cancer (CRC). More importantly, AR and ASR have anti-inflammatory and anti-cancer activities, as well as prefiguring a potential effect on inflammation-cancer transformation. We, therefore, aimed to review the immunometabolism potential of DGBX decoction and its components in this malignant transformation, to provide a helpful complement to manage the risk of IBD-CRC. The present study investigates the multifaceted roles of DGBX decoction and its entire components AR and ASR, including anti-inflammation effects, anti-cancer properties, immune regulation, and metabolic regulation. This assessment is informed by a synthesis of scholarly literature, with more than two hundred articles retrieved from PubMed, Web of Science, and Scopus databases within the past two decades. The search strategy employed utilized keywords such as "Danggui Buxue", "Astragali Radix", "Angelicae Sinensis Radix", "Inflammation", and "Metabolism", alongside the related synonyms, with a particular emphasis on high-quality research and studies yielding significant findings. The potential of DGBX decoction in modulating immunometabolism holds promise for the treatment of IBD-related CRC. It is particularly relevant given the heterogeneity of CRC and the growing trend towards personalized medicine, but the precise and detailed mechanism necessitate further in vivo validation and extensive clinical studies to substantiate the immunometabolic modulation and delineate the pathways involved.
Collapse
Affiliation(s)
- Yang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Qianming Kang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Luying He
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Ka Iong Chan
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, SAR, China
| | - Hui Gu
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Wenjing Xue
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, SAR, China.
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
48
|
Cao G, Xu Q, Huang S, Dai D, Wang J, Li W, Zhao Y, Lin J, Han X. B10 cells regulate macrophage polarization to alleviate inflammation and bone loss in periodontitis. J Periodontol 2024. [PMID: 39210600 DOI: 10.1002/jper.24-0114] [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: 02/15/2024] [Revised: 05/21/2024] [Accepted: 06/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The polarization of macrophages into an anti-inflammatory phenotype is crucial for resolving periodontal inflammation. It has been reported that B10 cells can regulate the immune response of macrophages during inflammation and are also able to regulate inflammation in periodontitis. However, whether B10 cells' regulation function in periodontitis is related to macrophage polarization remains unclear. This study aims to investigate whether B10 cells can regulate macrophage polarization in periodontitis. METHODS Macrophages were cocultured with B10 cells in vitro for 5 days. After coculture, macrophages were obtained for analysis directly or followed by stimulation with Pg-LPS/IFN-γ or IL-4/IL-13. Flow cytometry and/or reverse transcriptase-polymerase chain reaction (RT-PCR) were employed to detect the expression of IL-1β, iNOS, TNF-α, CD206, and ARG-1 in macrophages. B10 cells were transferred on the 5th day after ligation in wild or macrophage-depletion mice. Toluidine blue and TRAP staining were used to evaluate alveolar bone resorption and osteoclast activation. Immunohistochemistry was employed to detect the expression of CD68, IL-1β, TNF-α, iNOS, ARG-1, and IL-10. Immunofluorescence was used to detect the expression of CD68+CD86+M1 macrophages and CD68+CD206+M2 macrophages. RESULTS In vitro, B10 cells inhibit the expression of IL-1β, iNOS, and TNF-α in macrophages while increasing the expression of CD206 and ARG-1. In experimental periodontitis, B10 cells inhibit the polarization of CD68+CD86+M1 macrophages and iNOS expression but enhance the polarization of CD68+CD206+M2 macrophages and ARG-1 expression. Importantly, the depletion of macrophages partially weakened the regulation function of B10 cells in periodontitis. CONCLUSIONS B10 cells promote M2 macrophage polarization, inhibit M1 macrophage polarization in periodontitis, and alleviate periodontitis partially by regulating macrophage polarization.
Collapse
Affiliation(s)
- Guoqin Cao
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qiuping Xu
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shengyuan Huang
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Dong Dai
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jilei Wang
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wei Li
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yue Zhao
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiang Lin
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaozhe Han
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, Florida, USA
| |
Collapse
|
49
|
Tang X, Ma C, Wu Q, Yu M. Ovarian cancer derived extracellular vesicles promote the cancer progression and angiogenesis by mediating M2 macrophages polarization. J Ovarian Res 2024; 17:172. [PMID: 39182150 PMCID: PMC11344305 DOI: 10.1186/s13048-024-01497-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are mediators between cancer cells and other types of cells, such as tumor-associated macrophages (TAMs), in the tumor microenvironment. EVs can remodel the tumor microenvironment and regulate tumor progression. However, the underlying molecular mechanism of these interactions remains unclear. METHODS First, we explored the effect of TAMs on the survival prognosis of patients with ovarian cancer. Next, we isolated EVs derived from ovarian cancer cells (OV-EVs) through ultracentrifugation and analyzed the capacity of OV-EVs to regulate macrophage polarization in ovarian tumors and in whole peripheral blood. Moreover, we explored the roles of OV-EVs-induced macrophages in tumor progression through in vitro and in vivo assays. RESULTS OV-EVs were encapsulated by macrophages and induced the polarization of macrophages toward the M2 phenotype. Moreover, OV-EVs-induced M2 macrophages promoted angiogenesis and cancer progression both in vitro and in vivo. In addition, OV-EVs-induced macrophages increased the expression level of VEGF and increased the expression level of VEGFR in tumors, which resulted in angiogenesis in ovarian cancer. CONCLUSIONS The present study demonstrated that OV-EVs induce M2 polarization in macrophages and promote the progression of ovarian cancer. This study provides novel insight into the mechanism of ovarian cancer progression.
Collapse
Affiliation(s)
- Xue Tang
- Department of Gynecology, Maternal and Child Healthcare Hospital of Changning District, Shanghai, 200135, China
| | - Chengbin Ma
- Department of Gynecology, Maternal and Child Healthcare Hospital of Changning District, Shanghai, 200135, China
| | - Qiongwei Wu
- Department of Gynecology, Maternal and Child Healthcare Hospital of Changning District, Shanghai, 200135, China
| | - Meng Yu
- Department of Gynecology, Maternal and Child Healthcare Hospital of Changning District, Shanghai, 200135, China.
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
50
|
He G, Li Y, Zeng Y, Zhang Y, Jiang Q, Zhang Q, Zhu J, Gong J. Advancements in melanoma immunotherapy: the emergence of Extracellular Vesicle Vaccines. Cell Death Discov 2024; 10:374. [PMID: 39174509 PMCID: PMC11341806 DOI: 10.1038/s41420-024-02150-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024] Open
Abstract
Malignant melanoma represents a particularly aggressive type of skin cancer, originating from the pathological transformation of melanocytes. While conventional interventions such as surgical resection, chemotherapy, and radiation therapy are available, their non-specificity and collateral damage to normal cells has shifted the focus towards immunotherapy as a notable approach. Extracellular vesicles (EVs) are naturally occurring transporters, and are capable of delivering tumor-specific antigens and directly engaging in the immune response. Multiple types of EVs have emerged as promising platforms for melanoma vaccination. The effectiveness of EV-based melanoma vaccines manifests their ability to potentiate the immune response, particularly by activating dendritic cells (DCs) and CD8+ T lymphocytes, through engineering a synergy of antigen presentation and targeted delivery. Here, this review mainly focuses on the construction strategies for EV vaccines from various sources, their effects, and immunological mechanisms in treating melanoma, as well as the shortcomings and future perspectives in this field. These findings will provide novel insights into the innovative exploitation of EV-based vaccines for melanoma immune therapy.
Collapse
Affiliation(s)
- Guijuan He
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yichuan Li
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuyang Zeng
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yong Zhang
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiong Jiang
- Department of Pharmacy, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Xianning Medical College, Hubei University of Science & Technology, Xianning, Hubei, China.
| | - Jinjin Zhu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jun Gong
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|