1
|
Yibcharoenporn C, Muanprasat C, Moonwiriyakit A, Satitsri S, Pathomthongtaweechai N. AMPK in Intestinal Health and Disease: A Multifaceted Therapeutic Target for Metabolic and Inflammatory Disorders. Drug Des Devel Ther 2025; 19:3029-3058. [PMID: 40291159 PMCID: PMC12024487 DOI: 10.2147/dddt.s507489] [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: 11/27/2024] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
The intestines play essential roles in nutrient absorption and immune function and help maintain a protective barrier. Disruptions to its function can result in various diseases, including metabolic disorders, inflammation, and cancer. As a key regulator of cellular energy levels, 5'-adenosine monophosphate-activated protein kinase (AMPK) is essential for intestinal health. Beyond its established metabolic role, emerging evidence suggests that AMPK exerts profound effects on intestinal cell physiology, influencing cell proliferation and differentiation, inflammation, autophagy, barrier integrity, and smooth muscle contractility. Here, we explore the structure and regulation of AMPK, as well as its diverse roles in intestinal diseases and potential as a therapeutic target. Our findings reveal that AMPK is a multifaceted regulator of intestinal health, modulating various cellular processes and intestinal diseases. It plays a dual role in cancer, acting as both a tumor suppressor and promoter, and it regulates inflammatory pathways, autophagy, tight junction formation, and smooth muscle contractility. Both natural and synthetic AMPK activators offer promise as therapeutic agents. This review of AMPK's mechanisms and activators offers valuable insights for developing novel therapies for intestinal disorders. Further research is needed to fully define AMPK's roles and therapeutic potential.
Collapse
Affiliation(s)
- Chamnan Yibcharoenporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand
| | - Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand
| | - Saravut Satitsri
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand
| | - Nutthapoom Pathomthongtaweechai
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand
| |
Collapse
|
2
|
Guan Y, Zhang M, Song J, Negrete M, Adcock T, Kandel R, Racioppi L, Gerecht S. CaMKK2 Regulates Macrophage Polarization Induced by Matrix Stiffness: Implications for Shaping the Immune Response in Stiffened Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417778. [PMID: 40036145 PMCID: PMC12021110 DOI: 10.1002/advs.202417778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/13/2025] [Indexed: 03/06/2025]
Abstract
Macrophages are essential for immune responses and maintaining tissue homeostasis, exhibiting a wide range of phenotypes depending on their microenvironment. The extracellular matrix (ECM) is a vital component that provides structural support and organization to tissues, with matrix stiffness acting as a key regulator of macrophage behavior. Using physiologically relevant 3D stiffening hydrogel models, it is found that increased matrix stiffness alone promoted macrophage polarization toward a pro-regenerative phenotype, mimicking the effect of interleukin-4(IL-4) in softer matrices. Blocking Calcium/calmodulin-dependent kinase kinase 2 (CaMKK2) selectively inhibited stiffness-induced macrophage polarization without affecting IL-4-driven pro-regenerative pathways. In functional studies, CaMKK2 deletion prevented M2-like/pro-tumoral polarization caused by matrix stiffening, which in turn hindered tumor growth. In a murine wound healing model, loss of CaMKK2 impaired matrix stiffness-mediated macrophage accumulation, ultimately disrupting vascularization. These findings highlight the critical role of CaMKK2 in the macrophage mechanosensitive fate determination and gene expression program, positioning this kinase as a promising therapeutic target to selectively modulate macrophage responses in pathologically stiff tissues.
Collapse
Affiliation(s)
- Ya Guan
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Min Zhang
- Division of Hematological Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27708USA
| | - Jiyeon Song
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Marcos Negrete
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Tyler Adcock
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Reeva Kandel
- Division of Hematological Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27708USA
| | - Luigi Racioppi
- Division of Hematological Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27708USA
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples Federico IINaplesItaly
| | - Sharon Gerecht
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
3
|
Shen Z, Liu Z, Wang H, Landrock D, Noh JY, Zang QS, Lee C, Farnell YZ, Chen Z, Sun Y. Fructose induces inflammatory activation in macrophages and microglia through the nutrient-sensing ghrelin receptor. FASEB J 2025; 39:e70412. [PMID: 39985299 PMCID: PMC11846021 DOI: 10.1096/fj.202402531r] [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: 10/21/2024] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/24/2025]
Abstract
High fructose corn syrup (HFCS) is a commonly used sweetener in soft drinks and processed foods, and HFCS exacerbates inflammation when consumed in excess. Fructose, a primary component of HFCS; however, it is unclear whether fructose directly activates inflammatory signaling. Growth hormone secretagogue receptor (GHSR) is a receptor of the nutrient-sensing hormone ghrelin. We previously reported that GHSR ablation mitigates HFCS-induced inflammation in adipose tissue and liver, shifting macrophages toward an anti-inflammatory spectrum. Since inflammation is primarily governed by innate immune cells, such as macrophages in the peripheral tissues and microglia in the brain, this study aims to investigate whether GHSR autonomously regulates pro-inflammatory activation in macrophages and microglia upon fructose exposure. GHSR deletion mutants of RAW 264.7 macrophages and the immortalized microglial cell line (IMG) were generated using CRISPR-Cas9 gene editing. After treating the cells with equimolar concentrations of fructose or glucose for 24 h, fructose increased mRNA and protein expression of GHSR and pro-inflammatory cytokines (Il1β, Il6, and Tnfα) in both macrophages and microglia, suggesting that fructose activates Ghsr and induces inflammation directly in macrophages and microglia. Remarkably, GHSR deletion mutants (Ghsrmutant) of macrophages and microglia exhibited reduced inflammatory responses to fructose, indicating that GHSR mediates fructose-induced inflammation. Furthermore, we found that GHSR regulates fructose transport and fructose metabolism and mediates fructose-induced inflammatory activation through CREB-AKT-NF-κB and p38 MAPK signaling pathways. Our results underscore that fructose triggers inflammation, and reducing HFCS consumption would reduce disease risk. Moreover, these findings reveal for the first time that the nutrient-sensing receptor GHSR plays a crucial role in fructose-mediated inflammatory activation, suggesting that targeting GHSR may be a promising therapeutic approach to combat the immunotoxicity of foods that contain fructose.
Collapse
Affiliation(s)
- Zheng Shen
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Zeyu Liu
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Hongying Wang
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Danilo Landrock
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Ji Yeon Noh
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Qun Sophia Zang
- Department of SurgeryLoyola University Chicago Health Science CampusMaywoodIllinoisUSA
| | - Chih‐Hao Lee
- Genomics Research Center, Academia SinicaTaipeiTaiwan
| | - Yuhua Z. Farnell
- Department of Poultry ScienceTexas A&M UniversityCollege StationTexasUSA
| | - Zheng Chen
- Department of Biochemistry and Molecular BiologyThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Yuxiang Sun
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
- Department of Biochemistry & BiophysicsTexas A&M UniversityCollege StationTexasUSA
| |
Collapse
|
4
|
Li Y, Ai S, Li Y, Ye W, Li R, Xu X, Liu Q. The role of natural products targeting macrophage polarization in sepsis-induced lung injury. Chin Med 2025; 20:19. [PMID: 39910395 PMCID: PMC11800549 DOI: 10.1186/s13020-025-01067-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/17/2025] [Indexed: 02/07/2025] Open
Abstract
Sepsis-induced acute lung injury (SALI) is characterized by a dysregulated inflammatory and immune response. As a key component of the innate immune system, macrophages play a vital role in SALI, in which a macrophage phenotype imbalance caused by an increase in M1 macrophages or a decrease in M2 macrophages is common. Despite significant advances in SALI research, effective drug therapies are still lacking. Therefore, the development of new treatments for SALI is urgently needed. An increasing number of studies suggest that natural products (NPs) can alleviate SALI by modulating macrophage polarization through various targets and pathways. This review examines the regulatory mechanisms of macrophage polarization and their involvement in the progression of SALI. It highlights how NPs mitigate macrophage imbalances to alleviate SALI, focusing on key signaling pathways such as PI3K/AKT, TLR4/NF-κB, JAK/STAT, IRF, HIF, NRF2, HMGB1, TREM2, PKM2, and exosome-mediated signaling. NPs influencing macrophage polarization are classified into five groups: terpenoids, polyphenols, alkaloids, flavonoids, and others. This work provides valuable insights into the therapeutic potential of NPs in targeting macrophage polarization to treat SALI.
Collapse
Affiliation(s)
- Yake Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Beijing Institute of Chinese Medicine, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100010, China
| | - Sinan Ai
- China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yuan Li
- Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Wangyu Ye
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Rui Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Beijing Institute of Chinese Medicine, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100010, China
| | - Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100010, China.
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100010, China.
| |
Collapse
|
5
|
Cheon J, Kwon S, Kim M. Exerkines mitigating Alzheimer's disease progression by regulating inflammation: Focusing on macrophage/microglial NLRP3 inflammasome pathway. Alzheimers Dement 2025; 21:e14432. [PMID: 39641407 PMCID: PMC11848186 DOI: 10.1002/alz.14432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/16/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Recent research highlights the critical role of inflammation in accelerating amyloid beta and phosphorylated tubulin-associated protein tau cascade and Alzheimer's disease (AD) progression. Emerging evidence suggests that exercise influences AD by modulating inflammatory responses. We conducted a comprehensive search across multiple online databases. Our approach focused on previous and recent studies exploring the links among inflammation, AD, and the effects of exercise, specifically targeting research articles and books published in English. We pointed out that inflammation extends from the periphery to the central nervous system, facilitated by macrophage/microglial NLRP3 (nucleotide-binding domain, leucine rich-containing family, pyrin domain-containing protein 3) inflammasome signaling, which exacerbates classical AD mechanisms. Moreover, we provided further insights into the modulation of inflammasome signaling through exercise and exerkines, which may contribute to mitigating AD development. These insights deepen our understanding of AD mechanisms and offer the potential for identifying key therapeutic targets and biomarkers crucial for effective disease management and treatment. HIGHLIGHTS: Inflammation is potentially linked to the acceleration of classical Alzheimer's disease (AD) pathogenesis, including the pathways involving amyloid beta and phosphorylated tau, mediated by pro-inflammatory cytokines. Inflammation, initiated by the nucleotide-binding domain, leucine rich-containing family, pyrin domain-containing protein 3 (NLRP3) inflammasome signaling pathway within M1-type macrophages/microglia, may contribute to neuroinflammation and AD progression. Exercise has the potential to reduce inflammation and the development of AD by influencing NLRP3 inflammasome signaling via exerkines.
Collapse
Affiliation(s)
- Jaehwan Cheon
- Department of Biomedical ScienceKorea University College of MedicineSeongbuk‐guSeoulRepublic of Korea
- Uimyung Research Institute for NeuroscienceDepartment of PharmacySahmyook UniversityNowon‐guSeoulRepublic of Korea
| | - Soonyong Kwon
- Uimyung Research Institute for NeuroscienceDepartment of PharmacySahmyook UniversityNowon‐guSeoulRepublic of Korea
- Department of Chemistry & Life ScienceSahmyook UniversityNowon‐guSeoulRepublic of Korea
| | - Mikyung Kim
- Uimyung Research Institute for NeuroscienceDepartment of PharmacySahmyook UniversityNowon‐guSeoulRepublic of Korea
- Department of Chemistry & Life ScienceSahmyook UniversityNowon‐guSeoulRepublic of Korea
| |
Collapse
|
6
|
Yadav S, Ganta VC, Varadarajan S, Ong V, Shi Y, Das A, Ash D, Nagarkoti S, McMenamin M, Kelley S, Fukai T, Ushio-Fukai M. Myeloid DRP1 deficiency limits revascularization in ischemic muscles via inflammatory macrophage polarization and metabolic reprogramming. JCI Insight 2025; 10:e177334. [PMID: 39589842 PMCID: PMC11721294 DOI: 10.1172/jci.insight.177334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Macrophages play a crucial role in promoting perfusion recovery and revascularization after ischemia through antiinflammatory polarization, a process essential for the treatment of peripheral artery disease (PAD). Mitochondrial dynamics, particularly regulated by the fission protein DRP1, are closely linked to macrophage metabolism and inflammation. However, the role of DRP1 in reparative neovascularization remains unexplored. Here, we show that DRP1 expression was increased in F4/80+ macrophages within ischemic muscle on day 3 after hind limb ischemia (HLI), an animal model of PAD. Mice lacking Drp1 in myeloid cells exhibited impaired limb perfusion recovery, angiogenesis, and muscle regeneration after HLI. These effects were associated with increased proinflammatory M1-like macrophages, p-NF-κB, and TNF-α, and reduced antiinflammatory M2-like macrophages and p-AMPK in ischemic muscle of myeloid Drp1-/- mice. In vitro, Drp1-deficient macrophages under hypoxia serum starvation (HSS), an in vitro PAD model, demonstrated enhanced glycolysis via reducing p-AMPK as well as mitochondrial dysfunction, and excessive mitochondrial ROS production, resulting in increased proinflammatory M1-gene and reduced antiinflammatory M2-gene expression. Conditioned media from HSS-treated Drp1-/- macrophages exhibited increased proinflammatory cytokine secretion, leading to suppressed angiogenesis in endothelial cells. Thus, macrophage DRP1 deficiency under ischemia drives proinflammatory metabolic reprogramming and macrophage polarization, limiting revascularization in experimental PAD.
Collapse
Affiliation(s)
| | - Vijay C. Ganta
- Vascular Biology Center
- Department of Medicine (Cardiology), and
| | - Sudhahar Varadarajan
- Vascular Biology Center
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Vy Ong
- Biostatistics and Bioinformatics Core, Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yang Shi
- Biostatistics and Bioinformatics Core, Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Archita Das
- Vascular Biology Center
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Dipankar Ash
- Vascular Biology Center
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | | | - Malgorzata McMenamin
- Vascular Biology Center
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | | | - Tohru Fukai
- Vascular Biology Center
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | | |
Collapse
|
7
|
Ding R, Kan Q, Wang T, Xiao R, Song Y, Li D. Ginsenoside Rh2 regulates triple-negative breast cancer proliferation and apoptosis via the IL-6/JAK2/STAT3 pathway. Front Pharmacol 2025; 15:1483896. [PMID: 39845783 PMCID: PMC11751231 DOI: 10.3389/fphar.2024.1483896] [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: 08/22/2024] [Accepted: 12/05/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) is the most challenging subtype of breast cancer to treat. While previous studies have demonstrated that ginsenoside Rh2 induces apoptosis in TNBC cells, the specific molecular targets and underlying mechanisms remain poorly understood. This study aims to uncover the molecular mechanisms through which ginsenoside Rh2 regulates apoptosis and proliferation in TNBC, offering new insights into its therapeutic potential. Methods Network analysis and transcriptome sequencing were utilized to explore the potential mechanisms of ginsenoside Rh2 in treating TNBC. In vivo imaging and immunohistochemistry were employed to examine the effects of ginsenoside Rh2 in a TNBC mouse model. Functional assays were conducted to assess the impact of ginsenoside Rh2 on TNBC cell behavior. Additionally, ELISA, Western blot, and quantitative real-time PCR were used to further investigate the mechanisms of ginsenoside Rh2-induced apoptosis in TNBC cells. Results Through network analysis, 47 common targets were identified, and Gene Ontology (GO) enrichment analysis suggested that ginsenoside Rh2 may exert therapeutic effects in TNBC by influencing apoptosis, cell proliferation, and protein kinase activity. Both transcriptomic analysis and network analysis revealed the JAK/STAT signaling pathway as a key mechanism. Ginsenoside Rh2 inhibited tumor growth in TNBC mice and reduced the expression of IL- 6, IL-6R, STAT3, Bcl-2, and Bcl-xL in tumor tissues. The ability of ginsenoside Rh2 to inhibit TNBC cell proliferation was further confirmed by attenuating the activation of the IL-6/JAK2/STAT3 apoptosis pathway and reducing the expression of protein kinases AMPK-α1 and PKA-Cα. Conclusion Based on network analysis and experimental validation, our findings demonstrate that ginsenoside Rh2 regulates TNBC proliferation and apoptosis through suppression of the IL-6/JAK2/STAT3 pathway, both in vitro and in vivo. This comprehensive approach represents a significant advancement in understanding the therapeutic potential of ginsenoside Rh2 in treating TNBC.
Collapse
Affiliation(s)
| | | | | | | | | | - Duolu Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Liu J, Li X, Li Y, Gong Q, Luo K. Metformin-based nanomedicines for reprogramming tumor immune microenvironment. Theranostics 2025; 15:993-1016. [PMID: 39776799 PMCID: PMC11700864 DOI: 10.7150/thno.104872] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
Immunotherapy has transformed current cancer management, and it has achieved significant progress over last decades. However, an immunosuppressive tumor microenvironment (TME) diminishes the effectiveness of immunotherapy by suppressing the activity of immune cells and facilitating tumor immune-evasion. Adenosine monophosphate-activated protein kinase (AMPK), a key modulator of cellular energy metabolism and homeostasis, has gained growing attention in anti-tumor immunity. Metformin is usually considered as a cornerstone in diabetes management, and its role in activating the AMPK pathway has also been extensively explored in cancer therapy although the findings on its role remain inconsistent. Metformin in a nanomedicine formulation has been found to hold potential in reprogramming the immunosuppressive TME through immunometabolic modulation of both tumor and immune cells. This review elaborates the foundation and progress of immunometabolic reprogramming of the TME via metformin-based nanomedicines, offering valuable insights for the next generation of cancer therapy.
Collapse
Affiliation(s)
- Jieyu Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoling Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinggang Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, NHC Key Laboratory of Transplant Engineering and Immunology, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Xiamen Key Lab of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, NHC Key Laboratory of Transplant Engineering and Immunology, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
9
|
Tam E, Ouimet M, Sweeney G. Cardioprotective Effects of Adiponectin-Stimulated Autophagy. J Lipid Atheroscler 2025; 14:40-53. [PMID: 39911962 PMCID: PMC11791421 DOI: 10.12997/jla.2025.14.1.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 02/07/2025] Open
Abstract
Cardiovascular diseases (CVDs), including heart failure, pose a significant economic and health burden worldwide. Current treatment strategies for heart failure are greatly limited, in that they mainly mitigate symptoms or delay further progression. In contrast, therapies aimed at proactively preventing the onset of heart failure could greatly improve outcomes. Adiponectin is an adipocyte-derived hormone that confers an array of cardioprotective effects. It exerts anti-inflammatory effects, improves metabolic function, mitigates endothelial cell dysfunction, and reduce cardiomyocyte cell death. Furthermore, it has gained increasing attention for its ability to activate autophagy, a conserved cellular pathway that facilitates the degradation and recycling of cell components. The disruption of autophagy has been linked to CVDs including heart failure. Additionally, growing evidence also points to specific forms of autophagy, namely mitophagy and lipophagy, as crucial adaptive responses in protection against CVDs. The protective effects of adiponectin, autophagy, mitophagy, and lipophagy against CVDs along with potential therapeutic implications will be discussed.
Collapse
Affiliation(s)
- Eddie Tam
- Department of Biology, York University, Toronto, ON, Canada
| | - Mireille Ouimet
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
10
|
Dong H, Chen J, Zhang H, Zhao M, Yue Y, Wang S. Palmitic acid inhibits macrophage-mediated chemotherapy resistance in multiple myeloma via ALOX12 signaling. Int Immunopharmacol 2024; 143:113320. [PMID: 39378653 DOI: 10.1016/j.intimp.2024.113320] [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: 04/24/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Abstract
We previously discovered that macrophages (MΦs), especially tumor-associated MΦs (tMΦs), contribute to chemotherapy resistance in multiple myeloma (MM). However, the mechanism underlying MΦ-mediated chemotherapy resistance in MM needs further elucidation, and the identification of factors that preferentially abrogate MΦ-induced inhibition of MM chemotherapy may have important clinical significance. In this study, we showed that the expression of FASN and SCD2, the enzymes that synthesize palmitic acid and convert it to palmitoleic acid, was decreased in tMΦs compared with MΦs. Interestingly, palmitic acid abrogated the MΦ-mediated protection of MM cells from the effects of bortezomib and melphalan in vitro. Combination treatment with palmitic acid and bortezomib or melphalan further inhibited MM tumor growth in vivo. Mechanistically, palmitic acid treatment increased ALOX12 expression in MΦs. ALOX12 inhibition partially abrogated the palmitic acid-induced decrease in MΦ-mediated MM cell survival. Palmitic acid treatment inhibited AMPK signaling in MΦs, and ALOX12 knockdown activated the AMPK signaling pathway in MΦs. AMPK inhibition decreased the MΦ-mediated protection of drug-treated MM cells, and AMPK activation partially abolished the palmitic acid-induced inhibition of MΦ-mediated protection. ALOX12 converts arachidonic acid (AA) to 12-HETE. Moreover, treatment with AA but not 12-HETE partially abrogated the inhibitory effect of palmitic acid on MΦ-mediated MM cell survival in response to bortezomib or melphalan. Overall, we identified palmitic acid as a factor that inhibits MΦ-mediated resistance to bortezomib and melphalan in MM, which may have clinical significance.
Collapse
Affiliation(s)
- He Dong
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Jintong Chen
- Department of Hematology, The First Hospital of Jilin University, Changchun 130021, China
| | - Hua Zhang
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Munan Zhao
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Ying Yue
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Siqing Wang
- Department of Cancer Immunology, The First Hospital of Jilin University, Changchun 130061, China.
| |
Collapse
|
11
|
Akhter N, Contreras J, Ansari MA, Ducruet AF, Hoda MN, Ahmad AS, Gangwani LD, Bhatia K, Ahmad S. Remote Ischemic Post-Conditioning (RIC) Mediates Anti-Inflammatory Signaling via Myeloid AMPKα1 in Murine Traumatic Optic Neuropathy (TON). Int J Mol Sci 2024; 25:13626. [PMID: 39769388 PMCID: PMC11728166 DOI: 10.3390/ijms252413626] [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: 10/25/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Traumatic optic neuropathy (TON) has been regarded a vision-threatening condition caused by either ocular or blunt/penetrating head trauma, which is characterized by direct or indirect TON. Injury happens during sports, vehicle accidents and mainly in military war and combat exposure. Earlier, we have demonstrated that remote ischemic post-conditioning (RIC) therapy is protective in TON, and here we report that AMPKα1 activation is crucial. AMPKα1 is the catalytic subunit of the heterotrimeric enzyme AMPK, the master regulator of cellular energetics and metabolism. The α1 isoform predominates in immune cells including macrophages (Mφs). Myeloid-specific AMPKα1 KO mice were generated by crossing AMPKα1Flox/Flox and LysMcre to carry out the study. We induced TON in mice by using a controlled impact system. Mice (mixed sex) were randomized in six experimental groups for Sham (mock); Sham (RIC); AMPKα1F/F (TON); AMPKα1F/F (TON+RIC); AMPKα1F/F LysMCre (TON); AMPKα1F/F LysMCre (TON+RIC). RIC therapy was given every day (5-7 days following TON). Data were generated by using Western blotting (pAMPKα1, ICAM1, Brn3 and GAP43), immunofluorescence (pAMPKα1, cd11b, TMEM119 and ICAM1), flow cytometry (CD11b, F4/80, CD68, CD206, IL-10 and LY6G), ELISA (TNF-α and IL-10) and transmission electron microscopy (TEM, for demyelination and axonal degeneration), and retinal oxygenation was measured by a Unisense sensor system. First, we observed retinal morphology with funduscopic images and found TON has vascular inflammation. H&E staining data suggested that TON increased retinal inflammation and RIC attenuates retinal ganglion cell death. Immunofluorescence and Western blot data showed increased microglial activation and decreased retinal ganglion cell (RGCs) marker Brn3 and axonal regeneration marker GAP43 expression in the TON [AMPKα1F/F] vs. Sham group, but TON+RIC [AMPKα1F/F] attenuated the expression level of these markers. Interestingly, higher microglia activation was observed in the myeloid AMPKα1F/F KO group following TON, and RIC therapy did not attenuate microglial expression. Flow cytometry, ELISA and retinal tissue oxygen data revealed that RIC therapy significantly reduced the pro-inflammatory signaling markers, increased anti-inflammatory macrophage polarization and improved oxygen level in the TON+RIC [AMPKα1F/F] group; however, RIC therapy did not reduce inflammatory signaling activation in the myeloid AMPKα1 KO mice. The transmission electron microscopy (TEM) data of the optic nerve showed increased demyelination and axonal degeneration in the TON [AMPKα1F/F] group, and RIC improved the myelination process in TON [AMPKα1F/F], but RIC had no significant effect in the AMPKα1 KO mice. The myeloid AMPKα1c deletion attenuated RIC induced anti-inflammatory macrophage polarization, and that suggests a molecular link between RIC and immune activation. Overall, these data suggest that RIC therapy provided protection against inflammation and neurodegeneration via myeloid AMPKα1 activation, but the deletion of myeloid AMPKα1 is not protective in TON. Further investigation of RIC and AMPKα1 signaling is warranted in TON.
Collapse
Affiliation(s)
- Naseem Akhter
- Department of Biology, Arizona State University, Lake Havasu City, AZ 86403, USA
| | - Jessica Contreras
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center (SJHMC), Phoenix, AZ 85013, USA (K.B.)
| | - Mairaj A. Ansari
- Department of Biotechnology, Centre for Virology, Hamdard University, New Delhi 110062, India
| | - Andrew F. Ducruet
- Department of Neurosurgery, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center (SJHMC), Phoenix, AZ 85013, USA
| | - Md Nasrul Hoda
- Department of Neurology, Henry Ford Medical Center, Detroit, MI 48202, USA
| | - Abdullah S. Ahmad
- Department of Neurology, Henry Ford Medical Center, Detroit, MI 48202, USA
| | - Laxman D. Gangwani
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Kanchan Bhatia
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center (SJHMC), Phoenix, AZ 85013, USA (K.B.)
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA
| | - Saif Ahmad
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center (SJHMC), Phoenix, AZ 85013, USA (K.B.)
- Phoenix Veteran Affairs (VA), Phoenix, AZ 85012, USA
| |
Collapse
|
12
|
Singh S, Singh PK, Ahmad Z, Das S, Foretz M, Viollet B, Giri S, Kumar A. Myeloid Cell-Specific Deletion of AMPKα1 Worsens Ocular Bacterial Infection by Skewing Macrophage Phenotypes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1656-1665. [PMID: 39413004 PMCID: PMC11573643 DOI: 10.4049/jimmunol.2400282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/11/2024] [Indexed: 10/18/2024]
Abstract
AMP-activated protein kinase (AMPK) plays a crucial role in governing essential cellular functions such as growth, proliferation, and survival. Previously, we observed increased vulnerability to bacterial (Staphylococcus aureus) endophthalmitis in global AMPKα1 knockout mice. In this study, we investigated the specific involvement of AMPKα1 in myeloid cells using LysMCre;AMPKα1fl mice. Our findings revealed that whereas endophthalmitis resolved in wild-type C57BL/6 mice, the severity of the disease progressively worsened in AMPKα1-deficient mice over time. Moreover, the intraocular bacterial load and inflammatory mediators (e.g., IL-1β, TNF-α, IL-6, and CXCL2) were markedly elevated in the LysMCre;AMPKα1fl mice. Mechanistically, the deletion of AMPKα1 in myeloid cells skewed macrophage polarization toward the inflammatory M1 phenotype and impaired the phagocytic clearance of S. aureus by macrophages. Notably, transferring AMPK-competent bone marrow from wild-type mice to AMPKα1 knockout mice preserved retinal function and mitigated the severity of endophthalmitis. Overall, our study underscores the role of myeloid-specific AMPKα1 in promoting the resolution of inflammation in the eye during bacterial infection. Hence, therapeutic strategies aimed at restoring or enhancing AMPKα1 activity could improve visual outcomes in endophthalmitis and other ocular infections.
Collapse
Affiliation(s)
- Sukhvinder Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Pawan Kumar Singh
- Department of Ophthalmology/ Mason Eye Institute, University of Missouri School of Medicine, Columbia, MO, USA
| | - Zeeshan Ahmad
- Department of Ophthalmology, Visual and Anatomical Sciences, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Susmita Das
- Department of Ophthalmology, Visual and Anatomical Sciences, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Marc Foretz
- Université Paris cité, CNRS, Inserm, Institut Cochin, Paris 75014, France
| | - Benoit Viollet
- Université Paris cité, CNRS, Inserm, Institut Cochin, Paris 75014, France
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | - Ashok Kumar
- Department of Ophthalmology, Visual and Anatomical Sciences, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
13
|
Takahashi N, Eltalkhawy YM, Nasu K, Abdelnaser RA, Monde K, Habash SA, Nasser H, Hiyoshi M, Ishimoto T, Suzu S. IL-10 induces activated phenotypes of monocytes observed in virally-suppressed HIV-1-infected individuals. Biochem Biophys Res Commun 2024; 729:150342. [PMID: 38981402 DOI: 10.1016/j.bbrc.2024.150342] [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: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
Despite viral suppression by effective combined antiretroviral therapy, HIV-1-infected individuals have an increased risk of non-AIDS-related overall morbidity, which is due to the persistent chronic inflammation exemplified by the activation of monocytes, such as increased CD16high subset, and elevated plasma level of soluble CD163 (sCD163) and soluble CD14 (sCD14). Here, we show that IL-10, which has been recognized as anti-inflammatory, induces these activated phenotypes of monocytes in vitro. IL-10 increased CD16high monocytes, which was due to the upregulation of CD16 mRNA expression and completely canceled by an inhibitor of Stat3. Moreover, IL-10 increased the production of sCD163 and sCD14 by monocytes, which was consistent with the upregulation of cell surface expression of CD163 and CD14, and mRNA expression of CD163. However, unlike the IL-10-indeuced upregulation of CD16, that of CD14 was minimally affected by the Stat3 inhibitor. Furthermore, the IL-10-induced upregulation of CD163 protein and mRNA was partially inhibited by the Stat3 inhibitor, but completely canceled by an inhibitor of AMPK, an upstream kinase of Stat3 and PI3K/Akt/mTORC1 pathways. In this study, we also found that HIV-1 pathogenic protein Nef, which is known to persist in plasma of virally-suppressed individuals, induced IL-10 production in monocyte-derived macrophages. Our results may suggest that IL-10, which is inducible by Nef-activated macrophages, is one of drivers for activated phenotypes of monocytes in virally-suppressed individuals, and that IL-10 induces the increased CD16high monocytes and elevated level of sCD163 and sCD14 through the activation of different signaling pathways.
Collapse
MESH Headings
- Humans
- Interleukin-10/metabolism
- Monocytes/metabolism
- Monocytes/immunology
- HIV Infections/immunology
- HIV Infections/virology
- HIV Infections/metabolism
- HIV Infections/blood
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- HIV-1
- Receptors, IgG/metabolism
- Lipopolysaccharide Receptors/metabolism
- STAT3 Transcription Factor/metabolism
- Phenotype
- Up-Regulation
- Cells, Cultured
Collapse
Affiliation(s)
- Naofumi Takahashi
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
| | - Youssef M Eltalkhawy
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Kanako Nasu
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Randa A Abdelnaser
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Kazuaki Monde
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Sara A Habash
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Hesham Nasser
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Masateru Hiyoshi
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shinya Suzu
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
14
|
Lopresti L, Tatangelo V, Baldari CT, Patrussi L. Rewiring the T cell-suppressive cytokine landscape of the tumor microenvironment: a new frontier for precision anti-cancer therapy. Front Immunol 2024; 15:1418527. [PMID: 39281678 PMCID: PMC11392891 DOI: 10.3389/fimmu.2024.1418527] [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: 04/16/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024] Open
Abstract
T lymphocytes that infiltrate the tumor microenvironment (TME) often fail to function as effective anti-cancer agents. Within the TME, cell-to-cell inhibitory interactions play significant roles in dampening their anti-tumor activities. Recent studies have revealed that soluble factors released in the TME by immune and non-immune cells, as well as by tumor cells themselves, contribute to the exacerbation of T cell exhaustion. Our understanding of the cytokine landscape of the TME, their interrelationships, and their impact on cancer development is still at its early stages. In this review, we aim to shed light on Interleukin (IL) -6, IL-9, and IL-10, a small group of JAK/STAT signaling-dependent cytokines harboring T cell-suppressive effects in the TME and summarize their mechanisms of action. Additionally, we will explore how advancements in scientific research can help us overcoming the obstacles posed by cytokines that suppress T cells in tumors, with the ultimate objective of stimulating further investigations for the development of novel therapeutic strategies to counteract their tumor-promoting activities.
Collapse
Affiliation(s)
| | | | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
15
|
Wixler V, Boergeling Y, Leite Dantas R, Varga G, Ludwig S. Conversion of dendritic cells into tolerogenic or inflammatory cells depends on the activation threshold and kinetics of the mTOR signaling pathway. Cell Commun Signal 2024; 22:281. [PMID: 38773618 PMCID: PMC11106905 DOI: 10.1186/s12964-024-01655-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND Restoring impaired peripheral immune tolerance is the primary challenge in treating autoimmune diseases. Our previous research demonstrated the effectiveness of small spleen peptides (SSPs), a fraction of low molecular weight proteins, in inhibiting the progression of psoriatic arthritis, even in the presence of high levels of the proinflammatory cytokine TNFα in the bloodstream. When specifically targeting dendritic cells (DCs), SSPs transform them into tolerogenic cells, which efficiently induce the development of regulatory Foxp3+ Treg cells. In this study, we provide further insights into the mechanism of action of SSPs. RESULTS We found that SSPs stimulate the activation of the mTOR signaling pathway in dendritic cells, albeit in a different manner than the classical immunogenic stimulus LPS. While LPS-induced activation is rapid, strong, and sustained, the activity induced by SSPs is delayed, less intense, yet still significant. These distinct patterns of activation, as measured by phosphorylation of key components of the pathway are also observed in response to other immunogenic and tolerogenic stimuli such as GM-CSF + IL-4 or IL-10 and TGFβ. The disparity in mTOR activation between immunogenic and tolerogenic stimuli is quantitative rather than qualitative. In both cases, mTOR activation primarily occurs through the PI3K/Akt signaling axis and involves ERK and GSK3β kinases, with minimal involvement of AMPK or NF-kB pathways. Furthermore, in the case of SSPs, mTOR activation seems to involve adenosine receptors. Additionally, we observed that DCs treated with SSPs exhibit an energy metabolism with high plasticity, which is typical of tolerogenic cells rather than immunogenic cells. CONCLUSION Hence, the decision whether dendritic cells enter an inflammatory or tolerogenic state seems to rely on varying activation thresholds and kinetics of the mTOR signaling pathway.
Collapse
Affiliation(s)
- Viktor Wixler
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms- University, Von-Esmarch-Str. 56, 48149, Muenster, Germany.
| | - Yvonne Boergeling
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms- University, Von-Esmarch-Str. 56, 48149, Muenster, Germany
| | - Rafael Leite Dantas
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms- University, Von-Esmarch-Str. 56, 48149, Muenster, Germany
- Department of Mental Health, Westfaelische Wilhelms-University, 48149, Muenster, Germany
| | - Georg Varga
- Pediatric Rheumatology and Immunology, University Children's Hospital Muenster, 48149, Muenster, Germany
| | - Stephan Ludwig
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms- University, Von-Esmarch-Str. 56, 48149, Muenster, Germany
| |
Collapse
|
16
|
Huang Q, Wang Y, Chen S, Liang F. Glycometabolic Reprogramming of Microglia in Neurodegenerative Diseases: Insights from Neuroinflammation. Aging Dis 2024; 15:1155-1175. [PMID: 37611905 PMCID: PMC11081147 DOI: 10.14336/ad.2023.0807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023] Open
Abstract
Neurodegenerative diseases (ND) are conditions defined by progressive deterioration of the structure and function of the nervous system. Some major examples include Alzheimer's disease (AD), Parkinson's disease (PD), and Amyotrophic lateral sclerosis (ALS). These diseases lead to various dysfunctions, like impaired cognition, memory, and movement. Chronic neuroinflammation may underlie numerous neurodegenerative disorders. Microglia, an important immunocell in the brain, plays a vital role in defending against neuroinflammation. When exposed to different stimuli, microglia are activated and assume different phenotypes, participating in immune regulation of the nervous system and maintaining tissue homeostasis. The immunological activity of activated microglia is affected by glucose metabolic alterations. However, in the context of chronic neuroinflammation, specific alterations of microglial glucose metabolism and their mechanisms of action remain unclear. Thus, in this paper, we review the glycometabolic reprogramming of microglia in ND. The key molecular targets and main metabolic pathways are the focus of this research. Additionally, this study explores the mechanisms underlying microglial glucose metabolism reprogramming in ND and offers an analysis of the most recent therapeutic advancements. The ultimate aim is to provide insights into the development of potential treatments for ND.
Collapse
Affiliation(s)
- Qi Huang
- Department of Rehabilitation, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| | - Yanfu Wang
- Department of Rehabilitation, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| | - Shanshan Chen
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Fengxia Liang
- Department of Acupuncture and Moxibustion, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
17
|
Hu W, Zhang X, Sheng H, Liu Z, Chen Y, Huang Y, He W, Luo G. The mutual regulation between γδ T cells and macrophages during wound healing. J Leukoc Biol 2024; 115:840-851. [PMID: 37493223 DOI: 10.1093/jleuko/qiad087] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/08/2023] [Accepted: 07/20/2023] [Indexed: 07/27/2023] Open
Abstract
Macrophages are the main cells shaping the local microenvironment during wound healing. As the prime T cells in the skin, γδ T cells participate in regulating microenvironment construction, determining their mutual regulation helps to understand the mechanisms of wound healing, and explore innovative therapeutic options for wound repair. This review introduced their respective role in wound healing firstly, and then summarized the regulatory effect of γδ T cells on macrophages, including chemotaxis, polarization, apoptosis, and pyroptosis. Last, the retrograde regulation on γδ T cells by macrophages was also discussed. The main purpose is to excavate novel interventions for treating wound and provide new thought for further research.
Collapse
Affiliation(s)
- Wengang Hu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), ShaPingBa District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, ShaPingBa District, Chongqing 400038, China
| | - Xiaorong Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), ShaPingBa District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, ShaPingBa District, Chongqing 400038, China
| | - Hao Sheng
- Urology Department, Second Affiliated Hospital, Third Military Medical University (Army Medical University), XinQiao District, Chongqing 400037, China
| | - Zhongyang Liu
- Department of Plastic Surgery, First Affiliated Hospital, Zhengzhou University, ErQi District, Zhengzhou, Henan 450000, China
| | - Yunxia Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), ShaPingBa District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, ShaPingBa District, Chongqing 400038, China
| | - Yong Huang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), ShaPingBa District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, ShaPingBa District, Chongqing 400038, China
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), ShaPingBa District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, ShaPingBa District, Chongqing 400038, China
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), ShaPingBa District, Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, ShaPingBa District, Chongqing 400038, China
| |
Collapse
|
18
|
Liu T, Zhuang XX, Tang YY, Gao YC, Gao JR. Mechanistic insights into Qiteng Xiaozhuo Granules' regulation of autophagy for chronic glomerulonephritis treatment: Serum pharmacochemistry, network pharmacology, and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117819. [PMID: 38286158 DOI: 10.1016/j.jep.2024.117819] [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: 12/12/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qiteng Xiaozhuo Granules (QTXZG), a traditional Chinese medicine prescription, is widely acknowledged for its therapeutic efficacy and lack of discernible toxicity in clinical practice, substantiating its potential in the treatment of chronic glomerulonephritis (CGN). Nevertheless, the specific effectiveness and underlying mechanisms of QTXZG remain insufficiently explored. AIM OF THE STUDY The purpose of this study was to explore the mechanism of the QTXZG in the treatment of CGN via targeting autophagy based on serum pharmacochemistry, network pharmacology, and experimental validation. METHODS Serum samples from SD rats orally administered QTXZG were analyzed using UPLC-QE/MS to identify contained compounds. Network and functional enrichment analyses elucidated QTXZG's targets and biological mechanisms. Reliability was ensured through molecular docking, in vivo and in vitro experiments. RESULTS After oral administration of QTXZG, 39 enriched compounds in serum samples collected 1 h later were identified as potential active agents, with 508 potential targets recognized as QTXZG-specific targets. Through integration of various databases, intersection analysis of QTXZG targets, CGN-related genes, and autophagy-related targets identified 10 core autophagy-related targets for QTXZG in CGN. GO and KEGG analyses emphasized their roles in autophagy, inflammation, and immune processes, particularly emphasizing the enrichment of the AMPK/mTOR signaling pathway. Molecular docking results demonstrated strong binding affinities between QTXZG's key compounds and the predicted core targets. In animal experiments, QTXZG was found to ameliorate renal tissue damage in CGN model mice, significantly reducing serum creatinine (Scr) and blood urea nitrogen (BUN) levels. Importantly, both animal and cell experiments revealed QTXZG's ability to decrease excessive ROS and inflammatory factor release in mesangial cells. Furthermore, in vitro and in vivo experiments confirmed QTXZG's capacity to upregulate Beclin1 and LC3II/I expression, decrease p62 expression, and induce CGN autophagy through modulation of the AMPK/mTOR pathway. CONCLUSIONS This study indicated that QTXZG can induce autophagy in CGN by affecting the AMPK/mTOR pathway, and induction of autophagy may be one of the possible mechanisms of QTXZG's anti-CGN.
Collapse
Affiliation(s)
- Tao Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230011, Anhui, China.
| | - Xing Xing Zhuang
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University, Chaohu, 238000, Anhui, China.
| | - Yong Yan Tang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230011, Anhui, China.
| | - Ya Chen Gao
- Nephrology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Jia Rong Gao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| |
Collapse
|
19
|
Bongartz H, Bradfield C, Gross J, Fraser I, Nita-Lazar A, Meier-Schellersheim M. IL-10 dependent adaptation allows macrophages to adjust inflammatory responses to TLR4 stimulation history. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587272. [PMID: 38654826 PMCID: PMC11037870 DOI: 10.1101/2024.03.28.587272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
During an infection, innate immune cells must adjust nature and strength of their responses to changing pathogen abundances. To determine how stimulation of the pathogen sensing TLR4 shapes subsequent macrophage responses, we systematically varied priming and restimulation concentrations of its ligand KLA. We find that different priming strengths have very distinct effects at multiple stages of the signaling response, including receptor internalization, MAPK activation, cytokine and chemokine production, and nuclear translocation and chromatin association of NFκB and IκB members. In particular, restimulation-induced TNF-α production required KLA doses equal to or greater than those used for prior exposure, indicating that macrophages can detect and adaptively respond to changing TLR4 stimuli. Interestingly, while such adaptation was dependent on the anti-inflammatory cytokine IL-10, exogenous concentrations of IL-10 corresponding to those secreted after strong priming did not exert suppressive effects on TNF-α without such prior priming, confirming the critical role of TLR4 stimulation history.
Collapse
Affiliation(s)
- H. Bongartz
- Computational Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - C. Bradfield
- Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J. Gross
- Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - I.D.C. Fraser
- Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - A. Nita-Lazar
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M. Meier-Schellersheim
- Computational Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
20
|
Arosa L, Camba-Gómez M, Lorenzo-Martín LF, Clavaín L, López M, Conde-Aranda J. RNA Expression of MMP12 Is Strongly Associated with Inflammatory Bowel Disease and Is Regulated by Metabolic Pathways in RAW 264.7 Macrophages. Int J Mol Sci 2024; 25:3167. [PMID: 38542140 PMCID: PMC10970096 DOI: 10.3390/ijms25063167] [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: 02/02/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Macrophage metalloelastase or matrix metalloproteinase-12 (MMP12) is a macrophage-specific proteolytic enzyme involved in the physiopathology of many inflammatory diseases, including inflammatory bowel disease. Although previously published data suggested that the modulation of MMP12 in macrophages could be a determinant for the development of intestinal inflammation, scarce information is available on the mechanisms underlying the regulation of MMP12 expression in those phagocytes. Therefore, in this study, we aimed to delineate the association of MMP12 with inflammatory bowel disease and the molecular events leading to the transcriptional control of this metalloproteinase. For that, we used publicly available transcriptional data. Also, we worked with the RAW 264.7 macrophage cell line for functional experiments. Our results showed a strong association of MMP12 expression with the severity of inflammatory bowel disease and the response to relevant biological therapies. In vitro assays revealed that the inhibition of mechanistic target of rapamycin complex 1 (mTORC1) and the stimulation of the AMP-activated protein kinase (AMPK) signaling pathway potentiated the expression of Mmp12. Additionally, AMPK and mTOR required a functional downstream glycolytic pathway to fully engage with Mmp12 expression. Finally, the pharmacological inhibition of MMP12 abolished the expression of the proinflammatory cytokine Interleukin-6 (Il6) in macrophages. Overall, our findings provide a better understanding of the mechanistic regulation of MMP12 in macrophages and its relationship with inflammation.
Collapse
Affiliation(s)
- Laura Arosa
- Molecular and Cellular Gastroenterology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (L.A.); (M.C.-G.)
| | - Miguel Camba-Gómez
- Molecular and Cellular Gastroenterology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (L.A.); (M.C.-G.)
| | | | - Laura Clavaín
- EGO Genomics, Scientific Park of the University of Salamanca, Adaja Street 4, Building M2, 37185 Villamayor, Spain;
| | - Miguel López
- NeurObesity Group, Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Javier Conde-Aranda
- Molecular and Cellular Gastroenterology Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (L.A.); (M.C.-G.)
| |
Collapse
|
21
|
Liao Z, Huang L, Chen J, Chen T, Kong D, Wei Q, Chen Q, Deng B, Li Y, Zhong S, Huang Z. Liraglutide Improves Nonalcoholic Fatty Liver Disease in Diabetic Mice by Activating Autophagy Through AMPK/mTOR Signaling Pathway. Diabetes Metab Syndr Obes 2024; 17:575-584. [PMID: 38343582 PMCID: PMC10854402 DOI: 10.2147/dmso.s447182] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/11/2024] [Indexed: 01/05/2025] Open
Abstract
BACKGROUND Type 2 diabetes (T2DM) combined nonalcoholic fatty liver disease (NAFLD) are characterized by metabolic disruptions. Liraglutide has been proved to be effective in T2DM. If LRG could regulate NAFLD combined T2DM has not been reported. METHODS Intraperitoneal injection of 1% streptozotocin (STZ) plus high-sugar and high-fat diet was used to induce NAFLD combined T2DM animal model. Palmitic acid (200 µmol/L) and glucose (25 mmol/L) incubation were used to induce cell model. The cell apoptosis, mRNA and protein expression were measured through flow cytometry, PCR, and Western blotting, respectively. RESULTS Liraglutide significantly improved the liver injury of NAFLD combined T2DM rats, but Com-C reversed the effect of liraglutide. The decreased AMPK/mTOR signaling pathway in the NAFLD combined T2DM animals was greatly activated by liraglutide. Com-C reversed the protection effects of liraglutide on palmitic acid+glucose induced cell damage. CONCLUSION Liraglutide could greatly alleviate the damage caused by NAFLD+T2DM and palmitic acid+glucose. The protection effects of liraglutide were greatly inhibited by suppressing AMPK/mTOR signaling pathway. This research might provide a novel therapeutic strategy for the prevention and treatment of NAFLD combined T2DM disease.
Collapse
Affiliation(s)
- Zhanlin Liao
- Department of Endocrine, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| | - Liangzhi Huang
- Department of Endocrine, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| | - Jun Chen
- Department of Ophthalmology, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| | - Ting Chen
- Department of Endocrine, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| | - Dezhi Kong
- Department of Endocrine, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| | - Qifeng Wei
- Department of Endocrine, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| | - Qiao Chen
- Department of Endocrine, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| | - Bin Deng
- Department of Endocrine, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| | - Yanyan Li
- Department of Endocrine, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| | - Shuai Zhong
- Department of Endocrine, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| | - Zugui Huang
- Department of Endocrine, Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, 353006, People’s Republic of China
| |
Collapse
|
22
|
Berglund R, Cheng Y, Piket E, Adzemovic MZ, Zeitelhofer M, Olsson T, Guerreiro-Cacais AO, Jagodic M. The aging mouse CNS is protected by an autophagy-dependent microglia population promoted by IL-34. Nat Commun 2024; 15:383. [PMID: 38195627 PMCID: PMC10776874 DOI: 10.1038/s41467-023-44556-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024] Open
Abstract
Microglia harness an unutilized health-promoting potential in age-related neurodegenerative and neuroinflammatory diseases, conditions like progressive multiple sclerosis (MS). Our research unveils an microglia population emerging in the cortical brain regions of aging mice, marked by ERK1/2, Akt, and AMPK phosphorylation patterns and a transcriptome indicative of activated autophagy - a process critical for cellular adaptability. By deleting the core autophagy gene Ulk1 in microglia, we reduce this population in the central nervous system of aged mice. Notably, this population is found dependent on IL-34, rather than CSF1, although both are ligands for CSF1R. When aging mice are exposed to autoimmune neuroinflammation, the loss of autophagy-dependent microglia leads to neural and glial cell death and increased mortality. Conversely, microglial expansion mediated by IL-34 exhibits a protective effect. These findings shed light on an autophagy-dependent neuroprotective microglia population as a potential target for treating age-related neuroinflammatory conditions, including progressive MS.
Collapse
Affiliation(s)
- Rasmus Berglund
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden.
| | - Yufei Cheng
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Eliane Piket
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Milena Z Adzemovic
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Manuel Zeitelhofer
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, 171 65, Solna, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Andre Ortlieb Guerreiro-Cacais
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Division of Neuro, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| |
Collapse
|
23
|
Athari SZ, Mirzaei Bavil F, Keyhanmanesh R, Lotfi H, Sajed Y, Delkhosh A, Ghiasi F. Voluntary exercise improves pulmonary inflammation through NF-κB and Nrf2 in type 2 diabetic male rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:74-80. [PMID: 38164478 PMCID: PMC10722479 DOI: 10.22038/ijbms.2023.70416.15307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 08/09/2023] [Indexed: 01/03/2024]
Abstract
Objectives This study aimed to evaluate the effects of voluntary exercise as an anti-inflammatory intervention on the pulmonary levels of inflammatory cytokines in type 2 diabetic male rats. Materials and Methods Twenty-eight male Wistar rats were divided into four groups (n=7), including control (Col), diabetic (Dia), voluntary exercise (Exe), and diabetic with voluntary exercise (Dia+Exe). Diabetes was induced by a high-fat diet (4 weeks) and intraperitoneal injection of streptozotocin (35 mg/kg), and animals did training on the running wheel for 10 weeks as voluntary exercise. Finally, the rats were euthanized and the lung tissues were sampled for the evaluation of the levels of pulmonary interleukin (IL)-10, IL-11, and TNF-α using ELISA, and the protein levels of Nrf-2 and NF-κB using western blotting and tissue histopathological analysis. Results Diabetes reduced the IL-10, IL-11, and Nrf2 levels (P<0.001 to P<0.01) and increased the levels of TNF-α and NF-κB compared to the Col group (P<0.001). Lung tissue levels of IL-10, IL-11, and Nrf2 in the Dia+Exe group enhanced compared to the Dia group (P<0.001 to P<0.05), however; the TNF-α and NF-κB levels decreased (P<0.001). The level of pulmonary Nrf2 in the Dia+Exe group was lower than that of the Exe group while the NF-κB level increased (P<0.001). Moreover, diabetes caused histopathological changes in lung tissue which improved with exercise in the Dia+Exe group. Conclusion These findings showed that voluntary exercise could improve diabetes-induced pulmonary complications by ameliorating inflammatory conditions.
Collapse
Affiliation(s)
- Seyed Zanyar Athari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Mirzaei Bavil
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajie Lotfi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Yousef Sajed
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aref Delkhosh
- Department of Pathobiology, Faculty of Veterinary Medicine, Division of Pathology, Urmia University, Urmia, Iran
| | - Fariba Ghiasi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
24
|
Cui Y, Chen J, Zhang Z, Shi H, Sun W, Yi Q. The role of AMPK in macrophage metabolism, function and polarisation. J Transl Med 2023; 21:892. [PMID: 38066566 PMCID: PMC10709986 DOI: 10.1186/s12967-023-04772-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is a ubiquitous sensor of energy and nutritional status in eukaryotic cells. It plays a key role in regulating cellular energy homeostasis and multiple aspects of cell metabolism. During macrophage polarisation, AMPK not only guides the metabolic programming of macrophages, but also counter-regulates the inflammatory function of macrophages and promotes their polarisation toward the anti-inflammatory phenotype. AMPK is located at the intersection of macrophage metabolism and inflammation. The metabolic characteristics of macrophages are closely related to immune-related diseases, infectious diseases, cancer progression and immunotherapy. This review discusses the structure of AMPK and its role in the metabolism, function and polarisation of macrophages. In addition, it summarises the important role of the AMPK pathway and AMPK activators in the development of macrophage-related diseases.
Collapse
Affiliation(s)
- Yinxing Cui
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Junhua Chen
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Zhao Zhang
- Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Houyin Shi
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Weichao Sun
- Department of Bone Joint and Bone Oncology, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong, China.
- The Central Laboratory, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong, China.
| | - Qian Yi
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
25
|
Assar S, Dastbaz M, Amini K, Roghani SA, Lotfi R, Taghadosi M, Kafi H, Abdan Z, Allahyari H, Rostampour R, Shahrokhvand SZ. Assessing the gene expression of the adenosine 5'-monophosphate-activated protein kinase (AMPK) and its relation with the IL-6 and IL-10 plasma levels in COVID-19 patients. Mol Biol Rep 2023; 50:9925-9933. [PMID: 37874507 DOI: 10.1007/s11033-023-08835-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/19/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND Metabolic dysregulation and excessive inflammation are implicated in the pathogenesis of the highly infectious disease of coronavirus disease 2019 (COVID-19), which is caused by a newly emerging coronavirus (i.e., severe acute respiratory syndrome-coronavirus 2; SARS-CoV-2). The adenosine 5'-monophosphate-activated protein kinase (AMPK), an energy sensor regulating the metabolic pathways in diverse cells, exerts a regulatory role in the immune system. This study aims to examine the mRNA expression level of AMPK and the plasma levels of interleukin-6 (IL-6) and IL-10 cytokines in patients with different grades of COVID-19. METHODS Peripheral blood was collected from 60 patients with COVID-19 (Moderate, severe, and critical). The plasma levels of IL-6 and IL-10 were quantified by enzyme-linked immunosorbent assay (ELISA), and the mRNA expression level of AMPK was determined using real-time PCR. RESULTS The results showed that the plasma levels of IL-6 increased significantly in critical and severe patients compared to moderate cases of COVID-19 (P < 0.001). Moreover, IL-10 plasma concentrations were significantly higher in critical and severe cases than in moderate cases of COVID-19 (P < 0.01 and P < 0.05, respectively). Also, the gene expression of AMPK was meaningfully enhanced in critical patients relative to moderate and severe cases of COVID-19, in order (P < 0.001 and P < 0.01, respectively). There was a positive association between AMPK gene expression and plasma levels of IL-6 and IL-10 (P = 0.006, r = 0.348, P = 0.028, r = 0.283, respectively). CONCLUSION Increasing AMPK gene expression is likely a necessary effort of the immune system to inhibit inflammation in critical COVID-19. However, this effort seems to be inadequate, probably due to factors that induce inflammation, like erythrocyte sedimentation rate (ESR) and IL-6.
Collapse
Affiliation(s)
- Shirin Assar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Dastbaz
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Komail Amini
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Askar Roghani
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ramin Lotfi
- Clinical Research Development Center, Tohid Hospital, Kurdistan University of Medical Sciences, Sanandaj, Iran.
- Lung Diseases and Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, 6617713446, Iran.
| | - Mahdi Taghadosi
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hamidreza Kafi
- Medical Department, Orchid Pharmed Company, Tehran, Iran
| | - Zahra Abdan
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hosna Allahyari
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Rezvan Rostampour
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyedeh Zahra Shahrokhvand
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
26
|
Yadav S, Ganta V, Sudhahar V, Ash D, Nagarkoti S, Das A, McMenamin M, Kelley S, Fukai T, Ushio-Fukai M. Myeloid Drp1 Deficiency Limits Revascularization in Ischemic Muscles via Inflammatory Macrophage Polarization and Metabolic Reprograming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.04.565656. [PMID: 37961122 PMCID: PMC10635146 DOI: 10.1101/2023.11.04.565656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In the preclinical model of peripheral arterial disease (PAD), M2-like anti-inflammatory macrophage polarization and angiogenesis are required for revascularization. The regulation of cell metabolism and inflammation in macrophages is tightly linked to mitochondrial dynamics. Drp1, a mitochondrial fission protein, has shown context-dependent macrophage phenotypes with both pro- and anti-inflammatory characteristics. However, the role of macrophage Drp1 in reparative neovascularization remains unexplored. Here we show that Drp1 expression was significantly increased in F4/80+ macrophages within ischemic muscle at day 3 following hindlimb ischemia (HLI), an animal model of PAD. Myeloid-specific Drp1 -/- mice exhibited reduced limb perfusion recovery, angiogenesis and muscle regeneration after HLI. These effects were concomitant with enhancement of pro-inflammatory M1-like macrophages, p-NFkB, and TNFα levels, while showing reduction in anti-inflammatory M2-like macrophages and p-AMPK in ischemic muscle of myeloid Drp1 -/- mice. In vitro, Drp1 -/- macrophages under hypoxia serum starvation (HSS), an in vitro PAD model, demonstrated enhanced glycolysis via reducing p-AMPK as well as mitochondrial dysfunction and excessive mitochondrial ROS, resulting in increased M1-gene and reduced M2-gene expression. Conditioned media from HSS-treated Drp1 -/- macrophages exhibited increased secretion of pro-inflammatory cytokines and suppressed angiogenic responses in cultured endothelial cells. Thus, Drp1 deficiency in macrophages under ischemia drives inflammatory metabolic reprogramming and macrophage polarization, thereby limiting revascularization in experimental PAD.
Collapse
|
27
|
Yang L, Yang L, Lu K, Su N, Li X, Guo S, Xue S, Lian F, Feng C. 3D Chiral Self-Assembling Matrixes for Regulating Polarization of Macrophages and Enhance Repair of Myocardial Infarction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304627. [PMID: 37767946 PMCID: PMC10646248 DOI: 10.1002/advs.202304627] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/22/2023] [Indexed: 09/29/2023]
Abstract
The regulation of inflammatory response at the site of injury and macrophage immunotherapy is critical for tissue repair. Chiral self-assemblies are one of the most ubiquitous life cues, which is closely related to biological functions, life processes, and even the pathogenesis of diseases. However, the role of supramolecular chiral self-assemblies in the regulation of immune functions in the internal environment of tissues has not been fully explored yet. Herein, 3D supramolecular chiral self-assembling matrixes are prepared to regulate the polarization of macrophages and further enhance the repair of myocardial infarction (MI). Experiments studies show that M-type (left-handed) self-assembling matrixes significantly inhibit inflammation and promote damaged myocardium repair by upregulating M2 macrophage polarization and downstream immune signaling compared with P-type (right-handed), and R-type (non-chirality) self-assembling matrixes. Classical molecular dynamics (MD) simulation demonstrates that M-type self-assembling matrixes display higher stereo-affinity to cellular binding, which enhances the clustering of mechanosensitive integrin β1 (Itgβ1) and activates focal adhesion kinase (FAK) and Rho-associated protein kinase (ROCK), as well as downstream PI3K/Akt1/mTOR signaling axes to promote M2 polarization. This study of designing a 3D chiral self-assembling matrixes microenvironment suitable for regulating the polarization of macrophages will provide devise basis for immunotherapy with biomimetic materials.
Collapse
Affiliation(s)
- Lei Yang
- Department of Cardiovascular SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
| | - Li Yang
- State Key Lab of Metal Matrix CompositesSchool of Materials Science and EngineeringShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Kongli Lu
- Department of Cardiovascular SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
| | - Nan Su
- State Key Lab of Metal Matrix CompositesSchool of Materials Science and EngineeringShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Xueqin Li
- Department of Cardiovascular SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
| | - Shuoxiang Guo
- Department of Cardiovascular SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
| | - Song Xue
- Department of Cardiovascular SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
| | - Feng Lian
- Department of Cardiovascular SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong University160 Pujian RoadShanghai200127P. R. China
| | - Chuanliang Feng
- State Key Lab of Metal Matrix CompositesSchool of Materials Science and EngineeringShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| |
Collapse
|
28
|
Sharda D, Kaur P, Choudhury D. Protein-modified nanomaterials: emerging trends in skin wound healing. DISCOVER NANO 2023; 18:127. [PMID: 37843732 PMCID: PMC10579214 DOI: 10.1186/s11671-023-03903-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 09/23/2023] [Indexed: 10/17/2023]
Abstract
Prolonged inflammation can impede wound healing, which is regulated by several proteins and cytokines, including IL-4, IL-10, IL-13, and TGF-β. Concentration-dependent effects of these molecules at the target site have been investigated by researchers to develop them as wound-healing agents by regulating signaling strength. Nanotechnology has provided a promising approach to achieve tissue-targeted delivery and increased effective concentration by developing protein-functionalized nanoparticles with growth factors (EGF, IGF, FGF, PDGF, TGF-β, TNF-α, and VEGF), antidiabetic wound-healing agents (insulin), and extracellular proteins (keratin, heparin, and silk fibroin). These molecules play critical roles in promoting cell proliferation, migration, ECM production, angiogenesis, and inflammation regulation. Therefore, protein-functionalized nanoparticles have emerged as a potential strategy for improving wound healing in delayed or impaired healing cases. This review summarizes the preparation and applications of these nanoparticles for normal or diabetic wound healing and highlights their potential to enhance wound healing.
Collapse
Affiliation(s)
- Deepinder Sharda
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Pawandeep Kaur
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Diptiman Choudhury
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
- Thapar Institute of Engineering and Technology-Virginia Tech Centre of Excellence for Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
29
|
Zeng W, Li F, Jin S, Ho PC, Liu PS, Xie X. Functional polarization of tumor-associated macrophages dictated by metabolic reprogramming. J Exp Clin Cancer Res 2023; 42:245. [PMID: 37740232 PMCID: PMC10517486 DOI: 10.1186/s13046-023-02832-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023] Open
Abstract
Macrophages are highly plastic in different tissues and can differentiate into functional subpopulations under different stimuli. Tumor-associated macrophages (TAMs) are one of the most important innate immune cells implicated in the establishment of an immunosuppressive tumor microenvironment (TME). Recent evidence pinpoints the critical role of metabolic reprogramming in dictating pro-tumorigenic functions of TAMs. Both tumor cells and macrophages undergo metabolic reprogramming to meet energy demands in the TME. Understanding the metabolic rewiring in TAMs can shed light on immune escape mechanisms and provide insights into repolarizing TAMs towards anti-tumorigenic function. Here, we discuss how metabolism impinges on the functional divergence of macrophages and its relevance to macrophage polarization in the TME.
Collapse
Affiliation(s)
- Wentao Zeng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Fei Li
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Shikai Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Ping-Chih Ho
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Ludwig Lausanne Branch, Lausanne, Switzerland
| | - Pu-Ste Liu
- Institute of Cellular and System Medicine, National Health Research Institute, Miaoli, Taiwan, ROC
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
30
|
Townsend LK, Steinberg GR. AMPK and the Endocrine Control of Metabolism. Endocr Rev 2023; 44:910-933. [PMID: 37115289 DOI: 10.1210/endrev/bnad012] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/10/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
Complex multicellular organisms require a coordinated response from multiple tissues to maintain whole-body homeostasis in the face of energetic stressors such as fasting, cold, and exercise. It is also essential that energy is stored efficiently with feeding and the chronic nutrient surplus that occurs with obesity. Mammals have adapted several endocrine signals that regulate metabolism in response to changes in nutrient availability and energy demand. These include hormones altered by fasting and refeeding including insulin, glucagon, glucagon-like peptide-1, catecholamines, ghrelin, and fibroblast growth factor 21; adipokines such as leptin and adiponectin; cell stress-induced cytokines like tumor necrosis factor alpha and growth differentiating factor 15, and lastly exerkines such as interleukin-6 and irisin. Over the last 2 decades, it has become apparent that many of these endocrine factors control metabolism by regulating the activity of the AMPK (adenosine monophosphate-activated protein kinase). AMPK is a master regulator of nutrient homeostasis, phosphorylating over 100 distinct substrates that are critical for controlling autophagy, carbohydrate, fatty acid, cholesterol, and protein metabolism. In this review, we discuss how AMPK integrates endocrine signals to maintain energy balance in response to diverse homeostatic challenges. We also present some considerations with respect to experimental design which should enhance reproducibility and the fidelity of the conclusions.
Collapse
Affiliation(s)
- Logan K Townsend
- Centre for Metabolism Obesity and Diabetes Research, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Gregory R Steinberg
- Centre for Metabolism Obesity and Diabetes Research, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
31
|
Kobayashi T, Toyama-Sorimachi N. Metabolic control from the endolysosome: lysosome-resident amino acid transporters open novel therapeutic possibilities. Front Immunol 2023; 14:1243104. [PMID: 37781390 PMCID: PMC10540624 DOI: 10.3389/fimmu.2023.1243104] [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: 06/20/2023] [Accepted: 08/23/2023] [Indexed: 10/03/2023] Open
Abstract
Amino acid transporters are generally recognized as machinery that transport amino acids from the extracellular environment into the cytoplasm. Although their primary function is the uptake of amino acids to supply the cell with nutrients and energy, endolysosome-resident amino acid (EL-aa) transporters possess several unique functions in accordance with their localization in intracellular vesicular membranes. They play pivotal roles in the maintenance of metabolic homeostasis via direct involvement in the amino acid sensing pathway, which regulates the activity of mechanistic target of rapamycin complex 1 (mTORC1), a master regulator of cellular metabolism. Additionally, some EL-aa transporters contribute to the maintenance of dynamic homeostasis of endolysosomes, including the regulation of endolysosomal acidity, by carrying amino acids out of endolysosomes. In addition, EL-aa transporters act as a scaffold to gather signaling molecules and multiple enzymes to control cellular metabolism on the endolysosomal membrane. Among EL-aa transporters, solute carrier family 15 member 4 (SLC15A4) is preferentially expressed in immune cells, including macrophages, dendritic cells, and B cells, and plays a key role in the integration of metabolic and inflammatory signals. In this review, we summarize our recent findings on EL-aa transporter contributions to inflammatory and metabolic signaling in the endolysosomes of immune cells by focusing on the SLC15 family, including SLC15A4 and SLC15A3, and discuss their uniqueness and universality. We also discuss the potential of targeting these EL-aa transporters in immune cells for the development of novel therapeutic strategies for inflammatory diseases. Because these transporters are highly expressed in immune cells and significantly alter the functions of immune cells, targeting them would provide a great advantage in ensuring a wide safety margin.
Collapse
Affiliation(s)
| | - Noriko Toyama-Sorimachi
- Division of Human Immunology, International Research and Development Center for Vaccines, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
| |
Collapse
|
32
|
Banerjee A, Dass D, Dhotre K, Wakchoure P, More A, Rana S, Khan AA, Mukherjee A. Combinatorial Effects of miRNAs in HSV-2 Infection of Macrophages: An In Silico and In Vitro Integration Approach. Vaccines (Basel) 2023; 11:1488. [PMID: 37766164 PMCID: PMC10537408 DOI: 10.3390/vaccines11091488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The rising issues of herpes simplex virus (HSV)-2 drug ramifications have encouraged the researchers to look for new and alternative approaches that pose minimum adversities in the host while efficiently reducing the HSV-2 infection. Although microRNAs (miRNAs), as unorthodox approaches, are gaining popularity due to eliciting highly reduced immunogenic reactions, their implications in HSV-2 research have been rarely explored. In this study, a pool of cellular miRNAs with significance in HSV-2-induced inflammatory and immune responses have been identified. Computationally recognizing the host targets of these miRNAs through network biology and machine learning, in vitro validation has been addressed along with the identification of their regulation in the HSV-2 infection. To signify the role of these identified miRNAs, they have been individually ectopically expressed in macrophages. The ectopic expression of the individual miRNAs was able to suppress HSV-2 viral gene expression. Taking a step forward, this study also highlights the Box-Behnken design-based combinatorial effect of ectopically expressed miRNAs on maximum suppression of HSV-2 infectivity. Therefore, the concentrations of each of the miRNAs optimized in a combination, predicted through expert systems biology tools were validated in vitro to not only recover the target expressions but also inhibit the HSV-2 infection in the macrophages. Overall, the study offers miRNAs as intriguing alternatives to commercially available medications against HSV-2. Moreover, the study illuminates the prophylactic potentiality of the miRNAs, which is significant since there are currently no vaccines available for HSV-2. Moving forward, the miRNAs are employed in an innovative strategy that incorporates intricate biological system models and in vitro confirmation methods to deliver a prospective combinatorial miRNA therapeutic against HSV-2 infection.
Collapse
Affiliation(s)
- Anwesha Banerjee
- Division of Virology, ICMR-National AIDS Research Institute, Pune 411026, MH, India; (A.B.); (D.D.); (K.D.); (A.M.)
| | - Debashree Dass
- Division of Virology, ICMR-National AIDS Research Institute, Pune 411026, MH, India; (A.B.); (D.D.); (K.D.); (A.M.)
| | - Kishore Dhotre
- Division of Virology, ICMR-National AIDS Research Institute, Pune 411026, MH, India; (A.B.); (D.D.); (K.D.); (A.M.)
| | - Pooja Wakchoure
- Division of Microbiology, ICMR-National AIDS Research Institute, Pune 411026, MH, India;
| | - Ashwini More
- Division of Virology, ICMR-National AIDS Research Institute, Pune 411026, MH, India; (A.B.); (D.D.); (K.D.); (A.M.)
| | - Santanu Rana
- Department of Zoology, RPMC-University of Calcutta, Uttarpara 712258, WB, India;
| | - Abdul A. Khan
- Division of Microbiology, ICMR-National AIDS Research Institute, Pune 411026, MH, India;
| | - Anupam Mukherjee
- Division of Virology, ICMR-National AIDS Research Institute, Pune 411026, MH, India; (A.B.); (D.D.); (K.D.); (A.M.)
| |
Collapse
|
33
|
Xiao C, Xiong W, Xu Y, Zou J, Zeng Y, Liu J, Peng Y, Hu C, Wu F. Immunometabolism: a new dimension in immunotherapy resistance. Front Med 2023; 17:585-616. [PMID: 37725232 DOI: 10.1007/s11684-023-1012-z] [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: 12/26/2022] [Accepted: 05/19/2023] [Indexed: 09/21/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated unparalleled clinical responses and revolutionized the paradigm of tumor treatment, while substantial patients remain unresponsive or develop resistance to ICIs as a single agent, which is traceable to cellular metabolic dysfunction. Although dysregulated metabolism has long been adjudged as a hallmark of tumor, it is now increasingly accepted that metabolic reprogramming is not exclusive to tumor cells but is also characteristic of immunocytes. Correspondingly, people used to pay more attention to the effect of tumor cell metabolism on immunocytes, but in practice immunocytes interact intimately with their own metabolic function in a way that has never been realized before during their activation and differentiation, which opens up a whole new frontier called immunometabolism. The metabolic intervention for tumor-infiltrating immunocytes could offer fresh opportunities to break the resistance and ameliorate existing ICI immunotherapy, whose crux might be to ascertain synergistic combinations of metabolic intervention with ICIs to reap synergic benefits and facilitate an adjusted anti-tumor immune response. Herein, we elaborate potential mechanisms underlying immunotherapy resistance from a novel dimension of metabolic reprogramming in diverse tumor-infiltrating immunocytes, and related metabolic intervention in the hope of offering a reference for targeting metabolic vulnerabilities to circumvent immunotherapeutic resistance.
Collapse
Affiliation(s)
- Chaoyue Xiao
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, China
| | - Yiting Xu
- Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Ji'an Zou
- Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Yue Zeng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Junqi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yurong Peng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Chunhong Hu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Hunan Cancer Mega-Data Intelligent Application and Engineering Research Centre, Changsha, 410011, China
| | - Fang Wu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Hunan Cancer Mega-Data Intelligent Application and Engineering Research Centre, Changsha, 410011, China.
- Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
34
|
Montano M, Correa-de-Araujo R. Maladaptive Immune Activation in Age-Related Decline of Muscle Function. J Gerontol A Biol Sci Med Sci 2023; 78:19-24. [PMID: 37325961 PMCID: PMC10272988 DOI: 10.1093/gerona/glad036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Indexed: 06/17/2023] Open
Abstract
Age-related changes in immune competency and inflammation play a role in the decline of physical function. In this review of the conference on Function-Promoting Therapies held in March 2022, we discuss the biology of aging and geroscience with an emphasis on decline in physical function and the role of age-related changes in immune competence and inflammation. More recent studies in skeletal muscle and aging highlighting a crosstalk between skeletal muscle, neuromuscular feedback, and immune cell subsets are also discussed. The value of strategies targeting specific pathways that affect skeletal muscle and more systems-wide approaches that provide benefits in muscle homeostasis with aging are underscored. Goals in clinical trial design and the need for incorporating differences in life history when interpreting results from these intervention strategies are important. Where applicable, references are made to papers presented at the conference. We conclude by underscoring the need to incorporate age-related immune competency and inflammation when interpreting results from interventions that target specific pathways predicted to promote skeletal muscle function and tissue homeostasis.
Collapse
Affiliation(s)
- Monty Montano
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Rosaly Correa-de-Araujo
- Division of Geriatrics and Clinical Gerontology, National Institute on Aging, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, USA
| |
Collapse
|
35
|
Laux J, Martorelli M, Späth N, Maier F, Burnet M, Laufer SA. Selective Inhibitors of Janus Kinase 3 Modify Responses to Lipopolysaccharides by Increasing the Interleukin-10-to-Tumor Necrosis Factor α Ratio. ACS Pharmacol Transl Sci 2023; 6:892-906. [PMID: 37325444 PMCID: PMC10262334 DOI: 10.1021/acsptsci.3c00043] [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: 03/02/2023] [Indexed: 06/17/2023]
Abstract
Janus kinase (JAK) inhibitors act at low doses (e.g., tofacitinib, 0.2-0.4 μmol/kg bid) in clinical use, suggesting an efficient underlying mode of action. We hypothesized that their effectiveness is due to their ability to raise the ratio of IL-10 to TNFα. Unlike other JAK isoforms, JAK3 is expressed mainly in hematopoietic cells and is essential for immune function. We used JAK3 selective inhibitors with preferential distribution to immune cells. Inhibition of JAK3 in human leukocytes reduced TNFα and IL-6 but maintained levels of IL-10, while pan-JAK inhibitors increased TNFα, IL-6, and IL-10. JAK1 is required for IL-10 receptor signaling, which suggests that, at exposure above the IC50 (55 nM for tofacitinib on JAK1), there is less feedback control of TNFα levels. This leads to self-limiting effects of JAK1 inhibitors and could place an upper limit on appropriate doses. In vivo, treating mice with JAK3 inhibitors before LPS administration decreased plasma TNFα and increased IL-10 above vehicle levels, suggesting that JAK3 inhibition may limit TNFα release by increasing IL-10 while leaving the IL-10 receptor functional. This mechanism should have general utility in controlling autoimmune diseases and can be conveniently observed by measuring the ratio of IL-10 to TNFα. In summary, our targeted, "leukotropic" inhibitors more effectively increased IL-10/TNFα ratios than unselective control compounds and could, therefore, be ideal for autoimmune therapy.
Collapse
Affiliation(s)
- Julian Laux
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
- Department
of Pharmaceutical/Medicinal Chemistry, Eberhard
Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE, Germany
| | - Mariella Martorelli
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
- Department
of Pharmaceutical/Medicinal Chemistry, Eberhard
Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE, Germany
| | - Nadja Späth
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
| | - Florian Maier
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
| | - Michael Burnet
- Synovo
GmbH, Paul-Ehrlich-Straße
15, 72076 Tübingen, DE, Germany
| | - Stefan A. Laufer
- Department
of Pharmaceutical/Medicinal Chemistry, Eberhard
Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE, Germany
- Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, University
of Tübingen, 72076 Tübingen, Germany
- Tübingen
Center for Academic Drug Discovery & Development (TüCAD2), 72076 Tübingen, Germany
| |
Collapse
|
36
|
Callegari IOM, Rocha GZ, Oliveira AG. Physical exercise, health, and disease treatment: The role of macrophages. Front Physiol 2023; 14:1061353. [PMID: 37179836 PMCID: PMC10166825 DOI: 10.3389/fphys.2023.1061353] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
Subclinical inflammation is linked to comorbidities and risk factors, consolidating the diagnosis of chronic non-communicable diseases, such as insulin resistance, atherosclerosis, hepatic steatosis, and some types of cancer. In this context, the role of macrophages is highlighted as a marker of inflammation as well as for the high power of plasticity of these cells. Macrophages can be activated in a wide range between classical or proinflammatory, named M1, and alternative or anti-inflammatory, also known as M2 polarization. All nuances between M1 and M2 macrophages orchestrate the immune response by secreting different sets of chemokines, while M1 cells promote Th1 response, the M2 macrophages recruit Th2 and Tregs lymphocytes. In turn, physical exercise has been a faithful tool in combating the proinflammatory phenotype of macrophages. This review proposes to investigate the cellular and molecular mechanisms in which physical exercise can help control inflammation and infiltration of macrophages within the non-communicable diseases scope. During obesity progress, proinflammatory macrophages predominate in adipose tissue inflammation, which reduces insulin sensitivity until the development of type 2 diabetes, progression of atherosclerosis, and diagnosis of non-alcoholic fatty liver disease. In this case, physical activity restores the balance between the proinflammatory/anti-inflammatory macrophage ratio, reducing the level of meta-inflammation. In the case of cancer, the tumor microenvironment is compatible with a high level of hypoxia, which contributes to the advancement of the disease. However, exercise increases the level of oxygen supply, favoring macrophage polarization in favor of disease regression.
Collapse
Affiliation(s)
- Irineu O. M. Callegari
- Department of Physical Education, Bioscience Institute, São Paulo State University (UNESP), São Paulo, Brazil
| | - Guilherme Z. Rocha
- Department of Internal Medicine, State University of Campinas, Campinas, Brazil
| | - Alexandre G. Oliveira
- Department of Physical Education, Bioscience Institute, São Paulo State University (UNESP), São Paulo, Brazil
| |
Collapse
|
37
|
Chen M, Lan H, Yao S, Jin K, Chen Y. Metabolic Interventions in Tumor Immunity: Focus on Dual Pathway Inhibitors. Cancers (Basel) 2023; 15:cancers15072043. [PMID: 37046703 PMCID: PMC10093048 DOI: 10.3390/cancers15072043] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
The metabolism of tumors and immune cells in the tumor microenvironment (TME) can affect the fate of cancer and immune responses. Metabolic reprogramming can occur following the activation of metabolic-related signaling pathways, such as phosphoinositide 3-kinases (PI3Ks) and the mammalian target of rapamycin (mTOR). Moreover, various tumor-derived immunosuppressive metabolites following metabolic reprogramming also affect antitumor immune responses. Evidence shows that intervention in the metabolic pathways of tumors or immune cells can be an attractive and novel treatment option for cancer. For instance, administrating inhibitors of various signaling pathways, such as phosphoinositide 3-kinases (PI3Ks), can improve T cell-mediated antitumor immune responses. However, dual pathway inhibitors can significantly suppress tumor growth more than they inhibit each pathway separately. This review discusses the latest metabolic interventions by dual pathway inhibitors as well as the advantages and disadvantages of this therapeutic approach.
Collapse
Affiliation(s)
- Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Huanrong Lan
- Department of Surgical Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310002, China
| | - Shiya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Yun Chen
- Department of Colorectal Surgery, Xinchang People's Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang 312500, China
| |
Collapse
|
38
|
Abdelmalak MFL, Abdelrahim DS, George Michael TMA, Abdel-Maksoud OM, Labib JMW. Vitamin D and lactoferrin attenuate stress-induced colitis in Wistar rats via enhancing AMPK expression with inhibiting mTOR-STAT3 signaling and modulating autophagy. Cell Biochem Funct 2023; 41:211-222. [PMID: 36588325 DOI: 10.1002/cbf.3774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/02/2022] [Accepted: 12/16/2022] [Indexed: 01/03/2023]
Abstract
Irritable bowel syndrome (IBS) is a global gastrointestinal disorder closely related to psychological stress exposure and local colonic inflammation. Herein, we investigated the effect of wrap-restraint stress (WRS) on rat behavior, on adenosine monophosphate-activated protein kinase-mammalian/mechanistic target of rapamycin-signal transducer and activator of transcription 3 (AMPK-mTOR-STAT3) signaling, and autophagy in colonic mucosa. The impact of chronic administration of vitamin D3 and lactoferrin was compared. Twenty-four male Wistar rats were randomly divided into four groups. Chronic WRS protocol was applied as a rodent model of IBS. Group I: naïve animals, Group II: WRS animals, Group III: WRS-exposed and treated with vitamin D3 (500 IU/kg/day), and Group IV: WRS-exposed and treated with lactoferrin (300 mg/kg/day). In this study, we found that chronic administration of each of vitamin D3 and lactoferrin resulted in a significant increase in social interaction test, interleukin-10, AMPK, optical density of LC3B, goblet cell count and marked decrease in serum cortisol level, STAT3, inflammatory cell count, and optical density of mTOR in comparison to the WRS rats. Our findings suggest that both vitamin D3 and Lactoferrin could augment colonic autophagy through enhanced AMPK expression and inhibition of mTOR-STAT3 signaling, which offers practical insights into their clinical use in the prevention and therapy of IBS. However, lactoferrin intake as a nutritional supplement could be more helpful for stress-induced colitis treatment than vitamin D3.
Collapse
Affiliation(s)
- Marian F L Abdelmalak
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Dina S Abdelrahim
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Jolly M W Labib
- Histology and Cell Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
39
|
Zhou R, Guo T, Li J. Research progress on the antitumor effects of astragaloside IV. Eur J Pharmacol 2023; 938:175449. [PMID: 36473596 DOI: 10.1016/j.ejphar.2022.175449] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/15/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
One of the most important and effective components of Astragalus membranaceus is astragaloside IV (AS-IV), which can exert anti-tumor effects through various pathways. For instance, AS-IV exerts an anti-tumor effect by acting at the cellular level, regulating the phenotype switch of tumor-associated macrophages, or inhibiting the development of tumor cells. Furthermore, AS-IV inhibits tumor cell progression by enhancing its sensitivity to antitumor drugs or reversing the drug resistance of tumor cells. This article reviews the different mechanisms of AS-IV inhibition of epithelial-mesenchymal transition (EMT), migration, proliferation, and invasion of tumor cells, inducing apoptosis and improving the sensitivity of anti-tumor drugs. This review summarizes recent progress in the current research into AS-IV anti-tumor effect and provides insight on the next anti-tumor research of AS-IV.
Collapse
Affiliation(s)
- Ruixi Zhou
- The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou 730030, China
| | - Tiankang Guo
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730030, China
| | - Junliang Li
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou 730030, China; The First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou 730030, China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730030, China.
| |
Collapse
|
40
|
Keerthana CK, Rayginia TP, Shifana SC, Anto NP, Kalimuthu K, Isakov N, Anto RJ. The role of AMPK in cancer metabolism and its impact on the immunomodulation of the tumor microenvironment. Front Immunol 2023; 14:1114582. [PMID: 36875093 PMCID: PMC9975160 DOI: 10.3389/fimmu.2023.1114582] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a key metabolic sensor that is pivotal for the maintenance of cellular energy homeostasis. AMPK contributes to diverse metabolic and physiological effects besides its fundamental role in glucose and lipid metabolism. Aberrancy in AMPK signaling is one of the determining factors which lead to the development of chronic diseases such as obesity, inflammation, diabetes, and cancer. The activation of AMPK and its downstream signaling cascades orchestrate dynamic changes in the tumor cellular bioenergetics. It is well documented that AMPK possesses a suppressor role in the context of tumor development and progression by modulating the inflammatory and metabolic pathways. In addition, AMPK plays a central role in potentiating the phenotypic and functional reprogramming of various classes of immune cells which reside in the tumor microenvironment (TME). Furthermore, AMPK-mediated inflammatory responses facilitate the recruitment of certain types of immune cells to the TME, which impedes the development, progression, and metastasis of cancer. Thus, AMPK appears to play an important role in the regulation of anti-tumor immune response by regulating the metabolic plasticity of various immune cells. AMPK effectuates the metabolic modulation of anti-tumor immunity via nutrient regulation in the TME and by virtue of its molecular crosstalk with major immune checkpoints. Several studies including that from our lab emphasize on the role of AMPK in regulating the anticancer effects of several phytochemicals, which are potential anticancer drug candidates. The scope of this review encompasses the significance of the AMPK signaling in cancer metabolism and its influence on the key drivers of immune responses within the TME, with a special emphasis on the potential use of phytochemicals to target AMPK and combat cancer by modulating the tumor metabolism.
Collapse
Affiliation(s)
- Chenicheri Kizhakkeveettil Keerthana
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Department of Biotechnology, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Tennyson Prakash Rayginia
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Department of Biotechnology, University of Kerala, Thiruvananthapuram, Kerala, India
| | | | - Nikhil Ponnoor Anto
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kalishwaralal Kalimuthu
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Noah Isakov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
41
|
Wang F, Yan X, Hua Y, Song J, Liu D, Yang C, Peng F, Kang F, Hui Y. PI3K/AKT/mTOR pathway and its related molecules participate in PROK1 silence-induced anti-tumor effects on pancreatic cancer. Open Life Sci 2023; 18:20220538. [PMID: 37070074 PMCID: PMC10105552 DOI: 10.1515/biol-2022-0538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/28/2022] [Accepted: 11/21/2022] [Indexed: 04/19/2023] Open
Abstract
The PI3K/AKT/mTOR (phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin) pathway can be initiated by PROK1 (prokineticin 1), but its effect and mechanism of action in pancreatic carcinoma (PC) are not fully understood. In this study, we elucidated the roles of PROK1 and its related molecules in PC in vivo. PANC-1 cells with PROK1 knockdown were injected into BALB/c nude mice. The growth and weight of the tumor were monitored and measured, which was followed by TUNEL (terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling), immunohistochemical staining, and hematoxylin and eosin staining. The key proteins related to proliferation, apoptosis, and the PI3K/AKT/mTOR pathway were determined by Western blotting. We also used public databases to identify the molecules related to PROK1. The reduction of PROK1 inhibited angiopoiesis and promoted apoptosis in vivo. PCNA-1, cyclin D1, and Bcl-2 decreased considerably, while Bax and cleaved caspase-3 increased significantly after PROK1 inhibition. The PI3K/AKT/mTOR signal inhibition was also closely associated with PROK1 knockdown. The possible related molecules of PROK1, such as von Willebrand factor, were screened and considered to be involved in the aberrant activation of PI3K/AKT. In conclusion, PROK1 knockdown significantly prevented tumor growth and promoted apoptosis of human PC cells in vivo, where the PI3K/AKT/mTOR pathway was probably inhibited. Therefore, PROK1, along with its related molecules, might be important targets for PC therapy.
Collapse
Affiliation(s)
- Feng Wang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Xiaogang Yan
- Department of Surgical Oncology, The First People’s Hospital of Yinchuan, Yinchuan750001, China
| | - Yongqiang Hua
- Minimally Invasive Treatment Center, Fudan University Shanghai Cancer Center, Shanghai200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Jianjun Song
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Di Liu
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Chun Yang
- Department of Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan750001, China
| | - Fei Peng
- Department of Hepatobiliary Pancreatic Surgery, Edong Healthcare Huangshi Central Hospital, Huangshi435002, Hubei, China
| | - Fuping Kang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| | - Yongfeng Hui
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan750001, Ningxia, China
- Ningxia Clinical Medical Research Center of Hepatobiliary and Pancreatic Surgical Diseases, Yinchuan750001, China
| |
Collapse
|
42
|
Vaez H, Soraya H, Garjani A, Gholikhani T. Toll-Like Receptor 4 (TLR4) and AMPK Relevance in Cardiovascular Disease. Adv Pharm Bull 2023; 13:36-47. [PMID: 36721803 PMCID: PMC9871286 DOI: 10.34172/apb.2023.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/04/2021] [Accepted: 09/28/2021] [Indexed: 02/03/2023] Open
Abstract
Toll-like receptors (TLRs) are essential receptors of the innate immune system, playing a significant role in cardiovascular diseases. TLR4, with the highest expression among TLRs in the heart, has been investigated extensively for its critical role in different myocardial inflammatory conditions. Studies suggest that inhibition of TLR4 signaling pathways reduces inflammatory responses and even prevents additional injuries to the already damaged myocardium. Recent research results have led to a hypothesis that there may be a relation between TLR4 expression and 5' adenosine monophosphate-activated protein kinase (AMPK) signaling in various inflammatory conditions, including cardiovascular diseases. AMPK, as a cellular energy sensor, has been reported to show anti-inflammatory effects in various models of inflammatory diseases. AMPK, in addition to its physiological acts in the heart, plays an essential role in myocardial ischemia and hypoxia by activating various energy production pathways. Herein we will discuss the role of TLR4 and AMPK in cardiovascular diseases and a possible relation between TLRs and AMPK as a novel therapeutic target. In our opinion, AMPK-related TLR modulators will find application in treating different immune-mediated inflammatory disorders, especially inflammatory cardiac diseases, and present an option that will be widely used in clinical practice in the future.
Collapse
Affiliation(s)
- Haleh Vaez
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Corresponding Author: Haleh Vaez, Tel:+984133344798, Fax:+984133344798,
| | - Hamid Soraya
- Department of Pharmacology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Alireza Garjani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tooba Gholikhani
- Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Nanora Pharmaceuticals Ltd, Tabriz, Iran
| |
Collapse
|
43
|
Immunoregulatory signal networks and tumor immune evasion mechanisms: insights into therapeutic targets and agents in clinical development. Biochem J 2022; 479:2219-2260. [DOI: 10.1042/bcj20210233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022]
Abstract
Through activation of immune cells, the immune system is responsible for identifying and destroying infected or otherwise damaged cells including tumorigenic cells that can be recognized as foreign, thus maintaining homeostasis. However, tumor cells have evolved several mechanisms to avoid immune cell detection and killing, resulting in tumor growth and progression. In the tumor microenvironment, tumor infiltrating immune cells are inactivated by soluble factors or tumor promoting conditions and lose their effects on tumor cells. Analysis of signaling and crosstalk between immune cells and tumor cells have helped us to understand in more detail the mechanisms of tumor immune evasion and this forms basis for drug development strategies in the area of cancer immunotherapy. In this review, we will summarize the dominant signaling networks involved in immune escape and describe the status of development of therapeutic strategies to target tumor immune evasion mechanisms with focus on how the tumor microenvironment interacts with T cells.
Collapse
|
44
|
IL10/AMPK pathway was associated with the hippocampal anti-inflammatory response to high-sugar and high-fat diet withdrawal. Inflamm Res 2022; 71:1365-1374. [PMID: 36083322 DOI: 10.1007/s00011-022-01632-9] [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: 04/28/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE The present experimental study aimed to evaluate the effect of consuming an obesogenic diet (OD) on serum and hippocampal inflammation and proteins related to energy metabolism, alongside, we evaluated how the same parameters responded to an OD withdrawal. SUBJECTS Thirty male 60-days-old Wistar rats were used. METHODS The control group (n = 10) was fed the control diet across the whole experiment. The remaining animals were fed a high-sugar/high-fat (HSHF) diet for 30 days (n = 20) and half of them were placed on the control diet for 48 h (n = 10) afterwards. RESULTS OD intake decreased hippocampal AMPK phosphorylation, although, it did not increase serum inflammation and only increased hippocampal pNFκBp65 levels without any increase in the cytokines assessed. Moreover, OD withdrawal led to higher inflammatory markers in the serum and hippocampus and higher hippocampal AMPK phosphorylation. The mediation models applied suggested that the effect of OD withdrawal on hippocampal inflammation was driven by serum inflammation, which activated the hippocampal IL10/AMPK anti-inflammatory pathway as a response. CONCLUSION Our analyses suggest that OD withdrawal increases serum inflammation with hippocampal consequent inflammatory alterations. Despite the general assumption that improving diet improves health, this may not be immediate.
Collapse
|
45
|
Morris G, Gevezova M, Sarafian V, Maes M. Redox regulation of the immune response. Cell Mol Immunol 2022; 19:1079-1101. [PMID: 36056148 PMCID: PMC9508259 DOI: 10.1038/s41423-022-00902-0] [Citation(s) in RCA: 213] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/29/2022] [Indexed: 12/20/2022] Open
Abstract
AbstractThe immune-inflammatory response is associated with increased nitro-oxidative stress. The aim of this mechanistic review is to examine: (a) the role of redox-sensitive transcription factors and enzymes, ROS/RNS production, and the activity of cellular antioxidants in the activation and performance of macrophages, dendritic cells, neutrophils, T-cells, B-cells, and natural killer cells; (b) the involvement of high-density lipoprotein (HDL), apolipoprotein A1 (ApoA1), paraoxonase-1 (PON1), and oxidized phospholipids in regulating the immune response; and (c) the detrimental effects of hypernitrosylation and chronic nitro-oxidative stress on the immune response. The redox changes during immune-inflammatory responses are orchestrated by the actions of nuclear factor-κB, HIF1α, the mechanistic target of rapamycin, the phosphatidylinositol 3-kinase/protein kinase B signaling pathway, mitogen-activated protein kinases, 5' AMP-activated protein kinase, and peroxisome proliferator-activated receptor. The performance and survival of individual immune cells is under redox control and depends on intracellular and extracellular levels of ROS/RNS. They are heavily influenced by cellular antioxidants including the glutathione and thioredoxin systems, nuclear factor erythroid 2-related factor 2, and the HDL/ApoA1/PON1 complex. Chronic nitro-oxidative stress and hypernitrosylation inhibit the activity of those antioxidant systems, the tricarboxylic acid cycle, mitochondrial functions, and the metabolism of immune cells. In conclusion, redox-associated mechanisms modulate metabolic reprogramming of immune cells, macrophage and T helper cell polarization, phagocytosis, production of pro- versus anti-inflammatory cytokines, immune training and tolerance, chemotaxis, pathogen sensing, antiviral and antibacterial effects, Toll-like receptor activity, and endotoxin tolerance.
Collapse
|
46
|
Yadav S, Dwivedi A, Tripathi A. Biology of macrophage fate decision: Implication in inflammatory disorders. Cell Biol Int 2022; 46:1539-1556. [PMID: 35842768 DOI: 10.1002/cbin.11854] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/04/2022] [Accepted: 06/18/2022] [Indexed: 11/11/2022]
Abstract
The activation of immune cells in response to stimuli present in their microenvironment is regulated by their metabolic profile. Unlike the signal transduction events, which overlap to a huge degree in diverse cellular processes, the metabolome of a cell reflects a more precise picture of cell physiology and function. Different factors governing the cellular metabolome include receptor signaling, macro and micronutrients, normoxic and hypoxic conditions, energy needs, and biomass demand. Macrophages have enormous plasticity and can perform diverse functions depending upon their phenotypic state. This review presents recent updates on the cellular metabolome and molecular patterns associated with M1 and M2 macrophages, also termed "classically activated macrophages" and "alternatively activated macrophages," respectively. M1 macrophages are proinflammatory in nature and predominantly Th1-specific immune responses induce their polarization. On the contrary, M2 macrophages are anti-inflammatory in nature and primarily participate in Th2-specific responses. Interestingly, the same macrophage cell can adapt to the M1 or M2 phenotype depending upon the clues from its microenvironment. We elaborate on the various tissue niche-specific factors, which govern macrophage metabolism and heterogeneity. Furthermore, the current review provides an in-depth account of deregulated macrophage metabolism associated with pathological disorders such as cancer, obesity, and atherosclerosis. We further highlight significant differences in various metabolic pathways governing the cellular bioenergetics and their impact on macrophage effector functions and associated disorders.
Collapse
Affiliation(s)
- Sarika Yadav
- Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Ashish Dwivedi
- Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Anurag Tripathi
- Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
47
|
Stuart AKDC, Furuie JL, Cataldi TR, Stuart RM, Zawadneak MAC, Labate CA, Pimentel IC. Fungal consortium of two Beauveria bassiana strains increases their virulence, growth, and resistance to stress: A metabolomic approach. PLoS One 2022; 17:e0271460. [PMID: 35834517 PMCID: PMC9282594 DOI: 10.1371/journal.pone.0271460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 06/28/2022] [Indexed: 11/18/2022] Open
Abstract
The use of two or more microorganisms in a microbial consortium has been increasingly applied in the biological control of diseases and pests. Beauveria bassiana is one of the most widely studied fungal species in biological control, yet little is known about its role in fungal consortiums. In a previous study, our group found that a consortium formed by two strains of B. bassiana had significantly greater biocontrol potential against the polyphagous caterpillars Duponchelia fovealis (Lepidoptera: Crambidae) than either strain on its own. In this study, we use GC-MS and LC-MS/MS to evaluate and discuss the metabolomics of the consortium. A total of 21 consortium biomarkers were identified, corresponding to 14 detected by LC-MS/MS and seven by GC-MS. Antioxidant and anti-inflammatory mechanisms are the main properties of the metabolites produced by the consortium. These metabolites can depress the insect’s immune system, increasing its vulnerability and, hence, the fungal virulence of the consortium. In light of these results, we propose an action model of insect mortality due to the metabolites secreted by the consortium. The model includes the inhibition of defense mechanisms such as pro-inflammatory interleukin secretion, cell migration, cell aggregation, Dif, Dorsal and Relish gene transcription, and JAK/STAT and JNK signaling pathways. It also promotes the cleaning of oxidative molecules, like ROS, NOS, and H2O2, and the induction of virulence factors.
Collapse
Affiliation(s)
- Andressa Katiski da Costa Stuart
- Departamento de Patologia Básica, Setor de Ciências Biológicas, Laboratório de Microbiologia e Biologia Molecular (LabMicro), Universidade Federal do Paraná, Curitiba, Paraná, Brazil
- * E-mail:
| | - Jason Lee Furuie
- Departamento de Patologia Básica, Setor de Ciências Biológicas, Laboratório de Microbiologia e Biologia Molecular (LabMicro), Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Thais Regiani Cataldi
- Departamento de Genética, Laboratório de Genética de Plantas Max Feffer, Escola Superior de Agronomia Luiz de Queiroz – Esalq/USP, Piracicaba, São Paulo, Brazil
| | - Rodrigo Makowiecky Stuart
- Departamento de Patologia Básica, Setor de Ciências Biológicas, Laboratório de Microbiologia e Biologia Molecular (LabMicro), Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Maria Aparecida Cassilha Zawadneak
- Departamento de Patologia Básica, Setor de Ciências Biológicas, Laboratório de Microbiologia e Biologia Molecular (LabMicro), Universidade Federal do Paraná, Curitiba, Paraná, Brazil
- Departamento de Fitotecnia e Fitossanitaríssimo, Programa de Pós-graduação em Agronomia Produção Vegetal, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Carlos Alberto Labate
- Departamento de Genética, Laboratório de Genética de Plantas Max Feffer, Escola Superior de Agronomia Luiz de Queiroz – Esalq/USP, Piracicaba, São Paulo, Brazil
| | - Ida Chapaval Pimentel
- Departamento de Patologia Básica, Setor de Ciências Biológicas, Laboratório de Microbiologia e Biologia Molecular (LabMicro), Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| |
Collapse
|
48
|
Wang S, Liu G, Li Y, Pan Y. Metabolic Reprogramming Induces Macrophage Polarization in the Tumor Microenvironment. Front Immunol 2022; 13:840029. [PMID: 35874739 PMCID: PMC9302576 DOI: 10.3389/fimmu.2022.840029] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/09/2022] [Indexed: 12/18/2022] Open
Abstract
Macrophages are one of the most important cells in the innate immune system, they are converted into two distinct subtypes with completely different molecular phenotypes and functional features under different stimuli of the microenvironment: M1 macrophages induced by IFN-γ/lipopolysaccharides(LPS) and M2 macrophages induced by IL-4/IL-10/IL-13. Tumor-associated macrophages (TAMs) differentiate from macrophages through various factors in the tumor microenvironment (TME). TAMs have the phenotype and function of M2 macrophages and are capable of secreting multiple cytokines to promote tumor progression. Both tumor cells and macrophages can meet the energy needs for rapid cell growth and proliferation through metabolic reprogramming, so a comprehensive understanding of pro-tumor and antitumor metabolic switches in TAM is essential to understanding immune escape mechanisms. This paper focuses on the functions of relevant signaling pathways and cytokines during macrophage polarization and metabolic reprogramming, and briefly discusses the effects of different microenvironments and macrophage pathogenicity, in addition to describing the research progress of inhibitory drugs for certain metabolic and polarized signaling pathways.
Collapse
Affiliation(s)
- Shilin Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Guohong Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yirong Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
49
|
Luis LB, Ana GT, Carlos GE, Abraham GG, Iris EG, Martha ML, Vianney ON. Salmonella Promotes Its Own Survival in B Cells by Inhibiting Autophagy. Cells 2022; 11:cells11132061. [PMID: 35805144 PMCID: PMC9266210 DOI: 10.3390/cells11132061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/18/2022] [Accepted: 06/27/2022] [Indexed: 02/01/2023] Open
Abstract
Salmonella is a Gram-negative bacterium known to be the major cause of gastrointestinal diseases and systemic infections. During infection of murine B cells, Salmonella activates the PI3K/Akt pathway through its effector, SopB. This signaling pathway induces the downregulation of NLRC4 transcription, resulting in reduced secretion of IL-1β. Thus, Salmonella-infected B cells do not progress to pyroptosis; consequently, the bacteria can survive inside these cells. However, the mechanism by which Salmonella evades the control of B cells has not yet been elucidated. In this study, we found that SopB activates mTORC1, which is necessary for bacterial survival, since B cells cultured with the mTORC1 inhibitor rapamycin and B cells lacking raptor can control Salmonella infection. A similar result was observed in B cells when they were infected with the Salmonella SopB mutant (Δsopb). Salmonella also promoted the phosphorylation of the ULK1 complex at serine 757 (Ser757) by mTORC1, resulting in decreased levels of LC3-II in infected B cells. In this study, we did not observe these results when B cells were infected with Δsopb Salmonella. Our results demonstrated that Salmonella survival within B cells depends on the inhibition of autophagy by mTORC1 activation.
Collapse
Affiliation(s)
- Lopez-Bailon Luis
- Departamento y Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional (ENCB-IPN), Mexico 11350, Mexico; (L.-B.L.); (E.-G.I.); (M.-L.M.)
| | - Gonzalez-Telona Ana
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico 07360, Mexico; (G.-T.A.); (G.-E.C.); (G.-G.A.)
| | - Galán-Enríquez Carlos
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico 07360, Mexico; (G.-T.A.); (G.-E.C.); (G.-G.A.)
| | - García-Gil Abraham
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico 07360, Mexico; (G.-T.A.); (G.-E.C.); (G.-G.A.)
| | - Estrada-García Iris
- Departamento y Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional (ENCB-IPN), Mexico 11350, Mexico; (L.-B.L.); (E.-G.I.); (M.-L.M.)
| | - Moreno-Lafont Martha
- Departamento y Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional (ENCB-IPN), Mexico 11350, Mexico; (L.-B.L.); (E.-G.I.); (M.-L.M.)
| | - Ortiz-Navarrete Vianney
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico 07360, Mexico; (G.-T.A.); (G.-E.C.); (G.-G.A.)
- Correspondence:
| |
Collapse
|
50
|
Parapoxvirus Interleukin-10 Homologues Vary in Their Receptor Binding, Anti-Inflammatory, and Stimulatory Activities. Pathogens 2022; 11:pathogens11050507. [PMID: 35631028 PMCID: PMC9143231 DOI: 10.3390/pathogens11050507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 01/11/2023] Open
Abstract
Homologues of interleukin (IL)-10, a pleiotropic immunomodulatory cytokine, have been identified in the Parapoxvirus genus. The first identified, Orf virus (ORFV) IL-10, greatly enhanced infection of its host, exhibiting immune modulatory effects equivalent to human IL-10. IL-10-like genes were then identified in Bovine papular stomatitis virus (BPSV), Pseudocowpox virus (PCPV), Red deerpox virus (RDPV) and Grey sealpox virus (GSPV). This study aimed to produce and characterise recombinant parapoxvirus IL-10s, then quantitatively compare their receptor binding and immunomodulatory activities. Recombinant IL-10s were expressed, purified, then characterised using bioinformatic, biochemical and enzymatic analyses. Anti-inflammatory effects were assessed in lipoteichoic acid-activated THP-1 monocytes, and stimulatory effects in MC/9 mast cells. IL-10 receptor (IL-10R)1 binding was detected in a competitive displacement assay. BPSV IL-10 inhibited production of monocyte chemoattractant protein (MCP)-1, IL-8 and IL-1β, induced mast cell proliferation, and bound IL-10R1 similarly to ORFV IL-10. PCPV IL-10 showed reduced MCP-1 inhibition, mast cell proliferation, and IL-10R1 binding. RDPV IL-10 displayed reduced inhibition of IL-8 and MCP-1 production. GSPV IL-10 showed limited inhibition of IL-1β production and stimulation of mast cell proliferation. These findings provide valuable insight into IL-10 receptor interactions, and suggest that the parapoxvirus IL-10s play similar pathogenic roles during infection of their hosts.
Collapse
|