1
|
Wang J, Hu Y, Wang Z, Fan C, Liu Y, Xie Y, Liu L, Yang J, Xu Q. Exosomes Derived From Human Gingival Mesenchymal Stem Cells Induce Metabolic Reprogramming of Inflammatory Macrophages. J Clin Periodontol 2025. [PMID: 40388972 DOI: 10.1111/jcpe.14184] [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: 12/30/2024] [Revised: 04/22/2025] [Accepted: 05/09/2025] [Indexed: 05/21/2025]
Abstract
AIM To investigate the influence and mechanism of exosomes derived from human gingival mesenchymal stem cells (GMSC-Exo) regulating macrophage polarisation through metabolic reprogramming. MATERIALS AND METHODS Human acute monocytic leukaemia cells (THP-1)-derived macrophages were treated with GMSC-Exo or Porphyromonas gingivalis lipopolysaccharide (P.g-LPS) in vitro. Metabolic inhibitors were used to study the role of metabolic reprogramming in GMSC-Exo-induced polarisation, while the hypoxia-inducible factor-1 alpha (HIF-1α) modulators were employed to explore the HIF-1α signalling pathway's impact on macrophage metabolic reprogramming. The impact of GMSC-Exo on periodontitis and macrophage metabolism was assessed using a rat model in vivo. RESULTS In vitro experiments confirmed that GMSC-Exo promoted the polarisation of macrophages from pro-inflammatory M1 type (classically activated) to anti-inflammatory M2 type (alternatively activated) by promoting metabolic reprogramming (glycolysis to oxidative phosphorylation). In this process, the activation of the HIF-1α signalling pathway was inhibited. In vivo experiments revealed that GMSC-Exo could regulate the inflammatory microenvironment of periodontal tissue and the metabolic pattern of macrophages. CONCLUSION By inhibiting the activation of HIF-1α signalling pathway, GMSC-Exo trigger metabolic reprogramming in macrophages, thereby regulating the macrophage transformation from pro-inflammatory M1 phenotype to anti-inflammatory M2 phenotype. This change enhances the local inflammatory environment, aiding tissue repair and regeneration.
Collapse
Affiliation(s)
- Jialu Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Yingzhe Hu
- Department of Stomatology, Qingdao Huangdao Central Hospital, Qingdao, China
| | - Zhiguo Wang
- Department of Burn and Plastic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chun Fan
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ye Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yutong Xie
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Lubin Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Jingshu Yang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Stomatology, Qingdao University, Qingdao, China
| | - Quanchen Xu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Bakleh MZ, Al Haj Zen A. The Distinct Role of HIF-1α and HIF-2α in Hypoxia and Angiogenesis. Cells 2025; 14:673. [PMID: 40358197 PMCID: PMC12071368 DOI: 10.3390/cells14090673] [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: 04/02/2025] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Hypoxia results in a wide range of adaptive physiological responses, including metabolic reprogramming, erythropoiesis, and angiogenesis. The response to hypoxia at the cellular level is mainly regulated by hypoxia-inducible factors (HIFs): HIF1α and HIF2α isoforms. Although structurally similar and overlapping gene targets, both isoforms can exhibit distinct expression patterns and functions in some conditions of hypoxia. The interaction between these isoforms, known as the "HIF switch", determines their coordinated function under varying oxygen levels and exposure time. In angiogenesis, HIF-1α is rapidly stabilized under acute hypoxia, prompting a metabolic shift from oxidative phosphorylation to glycolysis and initiating angiogenesis by activating endothelial cells and extracellular matrix remodeling. Conversely, HIF-2α regulates cell responses to chronic hypoxia by sustaining genes critical for vascular remodeling and maturation. The current review highlights the different roles and regulatory mechanisms of HIF-1α and HIF-2α isoforms, focusing on their involvement in cell metabolism and the multi-step process of angiogenesis. Tuning the specific targeting of HIF isoforms and finding the right therapeutic window is essential to obtaining the best therapeutic effect in diseases such as cancer and vascular ischemic diseases.
Collapse
Affiliation(s)
| | - Ayman Al Haj Zen
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| |
Collapse
|
3
|
Wang Y, Zhou Q, Lu L, Xu J, Yang G, Sun X, Bao X, Kang L, Lv P, Liu R, Xu B, Yang Q, Mu D, Zhang B. Combining oxygen delivery and generation for targeted atherosclerosis therapy. J Control Release 2025; 380:1017-1030. [PMID: 39983924 DOI: 10.1016/j.jconrel.2025.02.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Hypoxia plays an important role in the progression of atherosclerosis. However, ameliorating hypoxia at atherosclerotic lesions remains a great challenge. To achieve targeted oxygen delivery to atherosclerotic plaques, Lipid 5-doped, platelet membrane-encapsulated magnetic mesoporous organosilicon nanoparticles loaded with perfluoro-15-crown ether (PFCE) (FMMON@PL) were prepared. PFCE worked as an oxygen carrier, while iron oxide nanoparticles (IONPs) acted as nanozymes with catalase-like activity to facilitate oxygen generation. To enhance plaque targeting, platelet membranes were coated onto mesoporous organosilicon nanoparticles containing PFCE and IONPs. Lipid 5 containing a tertiary amine was doped into the platelet membranes for lysosomal escape. Our results demonstrated that FMMON@PL specifically targeted macrophages in atherosclerotic plaques. FMMON@PL significantly reduced HIF-1α expression, ameliorated oxidative stress, inhibited foam cell formation, and reduced M1 macrophage polarization. In conclusion, FMMON@PL successfully achieved oxygen delivery within plaques and inhibited plaque progression, demonstrating the feasibility of hypoxia alleviation for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yujie Wang
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, No. 321 Zhongshan Road, Nanjing 210008, China
| | - Qianru Zhou
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, No. 321 Zhongshan Road, Nanjing 210008, China
| | - Le Lu
- Department of Transfusion Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing 210008, China
| | - Jianhua Xu
- Yizheng Hospital of Nanjing Drum Tower Hospital Group, 1 Huannan Road, Living Area of Sinopec Yizheng Chemical Fibre CO., LTD., Yizheng 211900, Jiangsu, China
| | - Gang Yang
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, No. 321 Zhongshan Road, Nanjing 210008, China
| | - Xuan Sun
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing 210008, China
| | - Xue Bao
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing 210008, China
| | - Lina Kang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing 210008, China
| | - Pin Lv
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing 210008, China
| | - Renyuan Liu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing 210008, China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing 210008, China.
| | - Qi Yang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Key Lab of Medical Engineering for Cardiovascular Disease, Ministry of Education, Beijing 100020, China.
| | - Dan Mu
- Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, No. 321 Zhongshan Road, Nanjing 210008, China; Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, No. 321 Zhongshan Road, Nanjing 210008, China; Yizheng Hospital of Nanjing Drum Tower Hospital Group, 1 Huannan Road, Living Area of Sinopec Yizheng Chemical Fibre CO., LTD., Yizheng 211900, Jiangsu, China; Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing 210008, China.
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing 210008, China
| |
Collapse
|
4
|
Park J, Wu Y, Le QV, Kim JS, Xu E, Lee J, Oh YK. Self-disassembling nanoparticles as oral nanotherapeutics targeting intestinal microenvironment. Nat Commun 2025; 16:3365. [PMID: 40204740 PMCID: PMC11982569 DOI: 10.1038/s41467-025-58513-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Inspired by the survival strategies of pyomelanin-producing microbes, we synthesize pyomelanin nanoparticles (PMNPs) from homogentisic acid- γ-lactone via auto-oxidation and investigate their biomedical potential. PMNPs possess distinct physicochemical properties, including reactive oxygen species scavenging and microenvironment-responsive self-disassembly. Under intestinal conditions, PMNPs self-disassemble and penetrate the nanoscale pores of the mucin layer. In an inflammatory bowel disease model, orally administered PMNPs withstand gastric acidity and, in their solubilized form, interact with macrophages and epithelial cells. They significantly reduce reactive oxygen species levels, exert anti-inflammatory effects, and restore gut microbiota composition. Compared to conventional nanoparticles and 5-aminosalicylic acid, PMNPs exhibit greater therapeutic efficacy. Clinical symptoms and intestinal inflammation are alleviated, and the gut microbiota is restored to near-normal levels. These findings underscore the therapeutic potential of PMNPs for inflammatory bowel disease treatment and suggest broader biomedical applications.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Jung Suk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Enzhen Xu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy, Korea University, Sejong, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Jafarzadeh S, Nemati M, Zandvakili R, Jafarzadeh A. Modulation of M1 and M2 macrophage polarization by metformin: Implications for inflammatory diseases and malignant tumors. Int Immunopharmacol 2025; 151:114345. [PMID: 40024215 DOI: 10.1016/j.intimp.2025.114345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
Macrophages perform an essential role in the body's defense mechanisms and tissue homeostasis. These cells exhibit plasticity and are categorized into two phenotypes, including classically activated/M1 pro-inflammatory and alternatively activated/M2 anti-inflammatory phenotypes. Functional deviation in macrophage polarization occurs in different pathological conditions that need correction. In addition to antidiabetic impacts, metformin also possesses multiple biological activities, including immunomodulatory, anti-inflammatory, anti-tumorigenic, anti-aging, cardioprotective, hepatoprotective, and tissue-regenerative properties. Metformin can influence the polarization of macrophages toward M1 and M2 phenotypes. The ability of metformin to support M2 polarization and suppress M1 polarization could enhance its anti-inflammatory properties and potentiate its protective effects in conditions such as chronic inflammatory diseases, atherosclerosis, and obesity. However, in metformin-treated tumors, the proportion of M2 macrophages is decreased, while the frequency ratio of M1 macrophages is increased, indicating that metformin can modulate macrophage polarization from a pro-tumoral M2 state to an anti-tumoral M1 phenotype in malignancies. Metformin affects macrophage polarization through AMPK-dependent and independent pathways involving factors, such as NF-κB, mTOR, ATF, AKT/AS160, SIRT1, STAT3, HO-1, PGC-1α/PPAR-γ, and NLRP3 inflammasome. By modulating cellular metabolism and apoptosis, metformin can also influence macrophage polarization. This review provides comprehensive evidence regarding metformin's effects on macrophage polarization and the underlying mechanisms. The polarization-inducing capabilities of metformin may provide significant therapeutic applications in various inflammatory diseases and malignant tumors.
Collapse
Affiliation(s)
- Sara Jafarzadeh
- Student Research Committee, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran; Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Raziyeh Zandvakili
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
6
|
Meyer SP, Bauer R, Brüne B, Schmid T. The role of type I interferon signaling in myeloid anti-tumor immunity. Front Immunol 2025; 16:1547466. [PMID: 40098954 PMCID: PMC11911529 DOI: 10.3389/fimmu.2025.1547466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Tumors often arise in chronically inflamed, and thus immunologically highly active niches. While immune cells are able to recognize and remove transformed cells, tumors eventually escape the control of the immune system by shaping their immediate microenvironment. In this context, macrophages are of major importance, as they initially exert anti-tumor functions before they adopt a tumor-associated phenotype that instead inhibits anti-tumor immune responses and even allows for sustaining a smoldering inflammatory, growth promoting tumor microenvironment (TME). Type I interferons (IFNs) are well established modulators of inflammatory reactions. While they have been shown to directly inhibit tumor growth, there is accumulating evidence that they also play an important role in altering immune cell functions within the TME. In the present review, we focus on the impact of type I IFNs on anti-tumor responses, driven by monocytes and macrophages. Specifically, we will provide an overview of tumor-intrinsic factors, which impinge on IFN-stimulated gene (ISG) expression, like the presence of nucleic acids, metabolites, or hypoxia. We will further summarize the current understanding of the consequences of altered IFN responses on macrophage phenotypes, i.e., differentiation, polarization, and functions. For the latter, we will focus on macrophage-mediated tumor cell killing and phagocytosis, as well as on how macrophages affect their environment by secreting cytokines and directly interacting with immune cells. Finally, we will discuss how type I IFN responses in macrophages might affect and should be considered for current and future tumor therapies.
Collapse
Affiliation(s)
- Sofie Patrizia Meyer
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Rebekka Bauer
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, Frankfurt, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| |
Collapse
|
7
|
Sentyabreva AV, Diatroptova MA, Miroshnichenko EA, Gerasimov AD, Kosyreva AM. Biomolecular and Functional Changes in a Culture of Microglial Cells Caused by Long-Term Exposure to AlCl 3. Bull Exp Biol Med 2025; 178:654-660. [PMID: 40295436 DOI: 10.1007/s10517-025-06392-0] [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: 06/11/2024] [Indexed: 04/30/2025]
Abstract
Inflammaging is one of the main risk factors for the development and progression of age-related diseases. The increase in the proinflammatory background and ROS production can lead to persistent activation and dysfunction of microglial cells. The biomolecular and functional changes in microglial cells after long-term exposure to prooxidant aluminum chloride were studied in in vitro experiment. BV2 cells were cultured in the presence of 0.5 and 1 mM AlCl3 for 70 or 140 h. The intensity of ROS production, apoptosis, and M1/M2 phenotype polarization of microglia were assessed by flow cytometry, qPCR-RT, and ELISA. Cells cultured with 0.5 mM AlCl3 showed signs of "healthy aging", while the higher concentration (1 mM) of AlCl3 led to persistent activation of microglia. The data obtained can be used for in vitro modeling of inflammaging and related physiological and pathological processes.
Collapse
Affiliation(s)
- A V Sentyabreva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia.
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia.
| | - M A Diatroptova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - E A Miroshnichenko
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - A D Gerasimov
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - A M Kosyreva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
8
|
Yang Y, Liu M, Dong X, Bai J, Shi W, Zhu Q, Liu J, Wang Z, Yi L, Yin X, Ni J, Qu C. Naringin Suppresses CoCl 2-Induced Ferroptosis in ARPE-19 Cells. Antioxidants (Basel) 2025; 14:236. [PMID: 40002420 PMCID: PMC11852185 DOI: 10.3390/antiox14020236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Hypoxic damage to retinal pigment epithelial (RPE) cells and subsequent neovascularization are key factors in the pathogenesis of branch retinal vein occlusion (BRVO). Naringin (NG), a naturally occurring flavanone glycoside, has demonstrated significant antioxidant and anti-neovascular activities. However, the regulatory effects and mechanisms of NG on ferroptosis in BRVO are yet to be explored. Our study aimed to investigate the protective effects of NG on RPE cells under hypoxic stress and to elucidate the underlying molecular mechanisms. Our findings revealed that NG significantly reduced cytotoxicity induced by cobaltous chloride (CoCl2) and also inhibited vascular proliferation in the retina, thereby attenuating choroidal neovascularization. NG pretreatment largely countered the overproduction of reactive oxygen species (ROS) and malondialdehyde (MDA) triggered by hypoxic damage, while also restoring levels of the antioxidants glutathione (GSH) and superoxide dismutase (SOD). Furthermore, NG pretreatment significantly activated the expression of hypoxia-inducible factor-1 alpha (HIF-1α) and its downstream heme oxygenase-1 (HO-1) and NADPH dehydrogenase (NQO1). In conclusion, NG not only inhibits neovascularization but also alleviates inflammation in RPE cells by modulating the HO-1/GPX4 pathway to inhibit ferroptosis. These findings highlight the potential of NG as a promising therapeutic agent for the treatment of BRVO.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jian Ni
- College of Chinese Herbal Medicine, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.Y.); (M.L.); (X.D.); (J.B.); (Q.Z.); (J.L.); (Z.W.); (L.Y.); (X.Y.)
| | - Changhai Qu
- College of Chinese Herbal Medicine, Beijing University of Chinese Medicine, Beijing 102488, China; (Y.Y.); (M.L.); (X.D.); (J.B.); (Q.Z.); (J.L.); (Z.W.); (L.Y.); (X.Y.)
| |
Collapse
|
9
|
Gil J, Kim D, Choi S, Bae ON. Cadmium-induced iron dysregulation contributes to functional impairment in brain endothelial cells via the ferroptosis pathway. Toxicol Appl Pharmacol 2025; 495:117233. [PMID: 39842614 DOI: 10.1016/j.taap.2025.117233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Cadmium (Cd2+) is a heavy metal that is a major hazardous environmental contaminant, ubiquitously present in the environment. Cd2+ exposure has been closely associated with an increased prevalence and severity of neurological and cardiovascular diseases (CVD). The blood-brain barrier (BBB) plays a crucial role in protecting the brain from external environmental factors. Mitochondria play an important role in maintaining the barrier function of brain endothelial cells by regulating energy metabolism and redox homeostasis. In this study, we aimed to assess the cytotoxic effects of Cd2+ on the integrity and function of brain endothelial cells. After 24 h of exposure, Cd2+ reduced cell survival, tight junction protein expression, and trans-endothelial electrical resistance (TEER) in bEnd.3 cells suggest a potential BBB integrity disruption by Cd2+ exposure. To clarify the underlying mechanism, we further investigated the role of mitochondria in iron overload-mediated cell death following Cd2+ exposure. Cd2+ induced a substantial reduction in mitochondrial basal respiration and ATP production in brain endothelial cells, suggesting mitochondrial dysfunction. In addition, Cd2+ exposure led to impaired autophagy, elevated iron levels, and increased lipid peroxidation, indicating the initiation of ferroptosis, a form of cell death triggered by iron. In summary, our research suggests that Cd2+ exposure can disrupt BBB function by causing mitochondrial dysfunction and disrupting iron homeostasis.
Collapse
Affiliation(s)
- Junkyung Gil
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Donghyun Kim
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Sungbin Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| | - Ok-Nam Bae
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA Campus, Ansan, South Korea.
| |
Collapse
|
10
|
Liu Z, Zheng X, Li N, Wang Z. Baicalein suppresses inflammation and attenuates acute lung injury by inhibiting glycolysis via HIF‑1α signaling. Mol Med Rep 2025; 31:18. [PMID: 39513601 PMCID: PMC11564906 DOI: 10.3892/mmr.2024.13383] [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: 06/09/2024] [Accepted: 09/18/2024] [Indexed: 11/15/2024] Open
Abstract
Baicalein, a flavonoid monomer compound isolated from the dried root of the traditional Chinese herb Scutellaria baicalensis, has several pharmacological activities, such as anti‑inflammatory, anti‑angiogenic, antitumor, antimicrobial and antiviral properties. Acute lung injury (ALI) is characterized by injury of the alveolar epithelium and capillary endothelium, which results in decreased lung volume, decreased lung compliance, ventilation/perfusion mismatch, intrapulmonary edema, alveolar edema and even acute hypoxemic respiratory failure. The present study aimed to investigate the effects of baicalein on lung injury and inflammation. Bioinformatics analysis using network pharmacology predicted that the hypoxia inducible factor‑1α (HIF‑1α) and glycolysis signaling pathways were involved in the mechanism underlying the therapeutic effects of baicalein. Further in vitro and in vivo experiments, such as immunohistochemistry, immunofluorescence and PCR, verified that baicalein could inhibit HIF‑1α signaling, thus suppressing glycolysis, and improving inflammatory responses and ALI. Taken together, the results of the present study suggested that the anti‑inflammatory effects of baicalein on treating ALI were associated with its ability to suppress glycolysis via the HIF‑1α signaling pathway.
Collapse
Affiliation(s)
- Zhongyou Liu
- Department of Respiratory Diseases, Zhumadian Hospital of Traditional Chinese Medicine, Zhumadian, Henan 463000, P.R. China
| | - Xiaona Zheng
- Department of Respiratory Diseases, Zhumadian Hospital of Traditional Chinese Medicine, Zhumadian, Henan 463000, P.R. China
| | - Ning Li
- Department of Scientific Research, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Zongyao Wang
- Department of Respiratory Diseases, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan 450007, P.R. China
| |
Collapse
|
11
|
Xie B, Li J, Lou Y, Chen Q, Yang Y, Zhang R, Liu Z, He L, Cheng Y. Reprogramming macrophage metabolism following myocardial infarction: A neglected piece of a therapeutic opportunity. Int Immunopharmacol 2024; 142:113019. [PMID: 39217876 DOI: 10.1016/j.intimp.2024.113019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Given the global prevalence of myocardial infarction (MI) as the leading cause of mortality, there is an urgent need to devise novel strategies that target reducing infarct size, accelerating cardiac tissue repair, and preventing detrimental left ventricular (LV) remodeling. Macrophages, as a predominant type of innate immune cells, undergo metabolic reprogramming following MI, resulting in alterations in function and phenotype that significantly impact the progression of MI size and LV remodeling. This article aimed to delineate the characteristics of macrophage metabolites during reprogramming in MI and elucidate their targets and functions in cardioprotection. Furthermore, we summarize the currently proposed regulatory mechanisms of macrophage metabolic reprogramming and identify the regulators derived from endogenous products and natural small molecules. Finally, we discussed the challenges of macrophage metabolic reprogramming in the treatment of MI, with the goal of inspiring further fundamental and clinical research into reprogramming macrophage metabolism and validating its potential therapeutic targets for MI.
Collapse
Affiliation(s)
- Baoping Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou 341000, China
| | - Jiahua Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Yanmei Lou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Qi Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Ying Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Rong Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China.
| | - Liu He
- Department of Endocrinology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong 510006, China.
| | - Yuanyuan Cheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China.
| |
Collapse
|
12
|
Santarsiero A, Convertini P, Iacobazzi D, Infantino V, Todisco S. Metabolic Crossroad Between Macrophages and Cancer Cells: Overview of Hepatocellular Carcinoma. Biomedicines 2024; 12:2684. [PMID: 39767591 PMCID: PMC11727080 DOI: 10.3390/biomedicines12122684] [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: 09/27/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 01/16/2025] Open
Abstract
The metabolic interplay between macrophages and cancer cells mirrors the plasticity of both kinds of cells, which adapt to the microenvironment by sustaining cell growth and proliferation. In this way, cancer cells induce macrophage polarization, and, on the other hand, tumor-associated macrophages (TAMs) contribute to the survival of cancer cells. In a simplified manner, macrophages can assume two opposite subtypes: M1, pro-inflammatory and anti-tumor phenotype, and M2, anti-inflammatory and protumor phenotype. How do cancer cells induce macrophage polarization? Any actor involved in tumor growth, including the mitochondria, releases molecules into the tumor microenvironment (TME) that trigger a subtype transition. These metabolic changes are the primary cause of this polarization. Hepatocellular carcinoma (HCC), the prevalent type of liver primary tumor, is characterized by cells with extensive metabolic adaptions due to high flexibility in different environmental conditions. This review focuses on the main metabolic features of M1 and M2 macrophages and HCC cells underlying their metabolic behavior in response to TME.
Collapse
Affiliation(s)
- Anna Santarsiero
- Department of Health Sciences, University of Basilicata, 85100 Potenza, Italy; (A.S.); (V.I.)
| | - Paolo Convertini
- Department of Basic and Applied Science, University of Basilicata, 85100 Potenza, Italy;
| | - Dominga Iacobazzi
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol BS2 8HW, UK;
| | - Vittoria Infantino
- Department of Health Sciences, University of Basilicata, 85100 Potenza, Italy; (A.S.); (V.I.)
| | - Simona Todisco
- Department of Basic and Applied Science, University of Basilicata, 85100 Potenza, Italy;
| |
Collapse
|
13
|
Xiong B, Fan M, Wang Z, Yang X, Cao S, Shen J, Fan B. Gentiopicroside-Induced gastric cancer necroptosis via the HIF-1 signaling pathway: A study involving molecular docking and experimental validation. PLoS One 2024; 19:e0311152. [PMID: 39570876 PMCID: PMC11581292 DOI: 10.1371/journal.pone.0311152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/13/2024] [Indexed: 11/24/2024] Open
Abstract
OBJECTIVES Gentiopicroside is an effective treatment for several types of cancer, inducing numerous forms of programmed cancer cell death. However, there are few investigations into the role of necroptosis. By utilizing molecular docking, and experimental validation, this study aims to investigate whether gentiopicroside elicits necroptosis in gastric cancer. METHODS Using software PyMOL and AutoDock, gentiopicroside was docked with RIPK1, RIPK3, MLKL and HIF-1α proteins. And a cell study was performed based on SGC7901 cells. The necroptosis-related proteins and HIF-1 signaling pathways were explored using western blot (WB) analysis. Finally, an animal study was performed to test the inhibitory effect in vivo. RESULTS Docking studies indicated that the docking energies of gentiopicroside to necroptosis-related proteins and necroptosis-characteristic proteins are all below -5 kcal/mol. Additionally, gentiopicroside cells reduce gastric cancer viability and inhibit proliferation. Results from the animal experiments indicated that gentiopicroside inhibits the growth of the gastric cancer xenograft tumor. Western blot and immunohistochemistry (IHC) staining demonstrated that gentiopicroside higher p-receptor-interacting protein kinase 3(p-RIPK3) levels in vitro and in vivo. CONCLUSION The findings of this study revealed that necroptosis is involved in the inhibitory effect of gentiopicroside toward gastric cancer.
Collapse
Affiliation(s)
- Bo Xiong
- Department of Clinical Pharmacy, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingjie Fan
- Department of Pharmacy, Shanghai Fourth Rehabilitation Hospital, Shanghai, China
| | - Zhihui Wang
- Department of Clinical Pharmacy, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaolu Yang
- Department of Clinical Pharmacy, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shan Cao
- Department of Clinical Pharmacy, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Shen
- Department of Pharmacy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Beibei Fan
- Department of Clinical Pharmacy, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Soroush A, Dunn JF. A Hypoxia-Inflammation Cycle and Multiple Sclerosis: Mechanisms and Therapeutic Implications. Curr Treat Options Neurol 2024; 27:6. [PMID: 39569339 PMCID: PMC11573864 DOI: 10.1007/s11940-024-00816-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/22/2024]
Abstract
Purpose of Review Multiple sclerosis (MS) is a complex neurodegenerative disease characterized by inflammation, demyelination, and neurodegeneration. Significant hypoxia exists in brain of people with MS (pwMS), likely contributing to inflammatory, neurodegenerative, and vascular impairments. In this review, we explore the concept of a negative feedback loop between hypoxia and inflammation, discussing its potential role in disease progression based on evidence of hypoxia, and its implications for therapeutic targets. Recent Findings In the experimental autoimmune encephalomyelitis (EAE) model, hypoxia has been detected in gray matter (GM) using histological stains, susceptibility MRI and implanted oxygen sensitive probes. In pwMS, hypoxia has been quantified using near-infrared spectroscopy (NIRS) to measure cortical tissue oxygen saturation (StO2), as well as through blood-based biomarkers such as Glucose Transporter-1 (GLUT-1). We outline the potential for the hypoxia-inflammation cycle to drive tissue damage even in the absence of plaques. Inflammation can drive hypoxia through blood-brain barrier (BBB) disruption and edema, mitochondrial dysfunction, oxidative stress, vessel blockage and vascular abnormalities. The hypoxia can, in turn, drive more inflammation. Summary The hypoxia-inflammation cycle could exacerbate neuroinflammation and disease progression. We explore therapeutic approaches that target this cycle, providing information about potential treatments in MS. There are many therapeutic approaches that could block this cycle, including inhibiting hypoxia-inducible factor 1-α (HIF-1α), blocking cell adhesion or using vasodilators or oxygen, which could reduce either inflammation or hypoxia. This review highlights the potential significance of the hypoxia-inflammation pathway in MS and suggests strategies to break the cycle. Such treatments could improve quality of life or reduce rates of progression.
Collapse
Affiliation(s)
- Ateyeh Soroush
- Department of Neuroscience, University of Calgary, Calgary, Alberta Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, Alberta Canada
- Experimental Imaging Center (EIC), Cal Wenzel Precision Health Building (CWPH Building) University of Calgary, 2500 University Dr NW, Calgary, AB T2N 1N4 Canada
| | - Jeff F Dunn
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, Alberta Canada
- Department of Radiology, University of Calgary, Calgary, Alberta Canada
- Experimental Imaging Center (EIC), Cal Wenzel Precision Health Building (CWPH Building) University of Calgary, 2500 University Dr NW, Calgary, AB T2N 1N4 Canada
| |
Collapse
|
15
|
Ajoolabady A, Pratico D, Lin L, Mantzoros CS, Bahijri S, Tuomilehto J, Ren J. Inflammation in atherosclerosis: pathophysiology and mechanisms. Cell Death Dis 2024; 15:817. [PMID: 39528464 PMCID: PMC11555284 DOI: 10.1038/s41419-024-07166-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/26/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Atherosclerosis imposes a heavy burden on cardiovascular health due to its indispensable role in the pathogenesis of cardiovascular disease (CVD) such as coronary artery disease and heart failure. Ample clinical and experimental evidence has corroborated the vital role of inflammation in the pathophysiology of atherosclerosis. Hence, the demand for preclinical research into atherosclerotic inflammation is on the horizon. Indeed, the acquisition of an in-depth knowledge of the molecular and cellular mechanisms of inflammation in atherosclerosis should allow us to identify novel therapeutic targets with translational merits. In this review, we aimed to critically discuss and speculate on the recently identified molecular and cellular mechanisms of inflammation in atherosclerosis. Moreover, we delineated various signaling cascades and proinflammatory responses in macrophages and other leukocytes that promote plaque inflammation and atherosclerosis. In the end, we highlighted potential therapeutic targets, the pros and cons of current interventions, as well as anti-inflammatory and atheroprotective mechanisms.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Ling Lin
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | | | - Suhad Bahijri
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Jaakko Tuomilehto
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia.
- Department of Public Health, University of Helsinki, Helsinki, Finland.
- Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
16
|
Xue JD, Gao J, Tang AF, Feng C. Shaping the immune landscape: Multidimensional environmental stimuli refine macrophage polarization and foster revolutionary approaches in tissue regeneration. Heliyon 2024; 10:e37192. [PMID: 39296009 PMCID: PMC11408064 DOI: 10.1016/j.heliyon.2024.e37192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
In immunology, the role of macrophages extends far beyond their traditional classification as mere phagocytes; they emerge as pivotal architects of the immune response, with their function being significantly influenced by multidimensional environmental stimuli. This review investigates the nuanced mechanisms by which diverse external signals ranging from chemical cues to physical stress orchestrate macrophage polarization, a process that is crucial for the modulation of immune responses. By transitioning between pro-inflammatory (M1) and anti-inflammatory (M2) states, macrophages exhibit remarkable plasticity, enabling them to adapt to and influence their surroundings effectively. The exploration of macrophage polarization provides a compelling narrative on how these cells can be manipulated to foster an immune environment conducive to tissue repair and regeneration. Highlighting cutting-edge research, this review presents innovative strategies that leverage the dynamic interplay between macrophages and their environment, proposing novel therapeutic avenues that harness the potential of macrophages in regenerative medicine. Moreover, this review critically evaluates the current challenges and future prospects of translating macrophage-centered strategies from the laboratory to clinical applications.
Collapse
Affiliation(s)
- Jing-Dong Xue
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jing Gao
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ai-Fang Tang
- Department of Geratology, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Chao Feng
- Department of Reproductive Medicine, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai 200030, China
| |
Collapse
|
17
|
Basheeruddin M, Qausain S. Hypoxia-Inducible Factor 1-Alpha (HIF-1α): An Essential Regulator in Cellular Metabolic Control. Cureus 2024; 16:e63852. [PMID: 39099978 PMCID: PMC11297807 DOI: 10.7759/cureus.63852] [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: 06/20/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024] Open
Abstract
The element that causes hypoxia when the von Hippel-Lindau (VHL) protein is not functioning is hypoxia-inducible factor 1-alpha (HIF-1α), which is the essential protein linked to cell control under hypoxia. Consequently, in situations where cells are oxygen-deficient, HIF-1α carries out a variety of essential functions. Citations to relevant literature support the notion that HIF-1α regulates the mitochondrial and glycolytic pathways, as well as the transition from the former to the latter. Cells with limited oxygen supply benefit from this change, which is especially beneficial for the inhibition of the mitochondrial electron transport chain and enhanced uptake of glucose and lactate. During hypoxic stress, HIF-1α also controls proline and glycolytic transporters such as lactate dehydrogenase A (LDHA) and glucose transporter 1 (GLUT1). These mechanisms help the cell return to homeostasis. Therefore, through metabolic change promoting adenosine triphosphate (ATP) synthesis and reducing reactive oxygen species (ROS) creation, HIF-1α may have a role in reducing oxidative stress in cells. This evidence, which describes the function of HIF-1α in many molecular pathways, further supports the notion that it is prognostic and that it contributes to hypoxic cell adaption. Understanding more about disorders, including inflammation, cancer, and ischemia, is possible because of HIF-1α's effect on metabolic changes. Gaining knowledge about the battle between metabolism, which is directed by HIF-1α, would help advance the research on pathophysiological situations involving dysregulated hypoxia and metabolism.
Collapse
Affiliation(s)
- Mohd Basheeruddin
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sana Qausain
- Biomedical Sciences, Allied Health Sciences, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
18
|
Li C, Wang J, Xu JF, Pi J, Zheng B. Roles of HIF-1α signaling in Mycobacterium tuberculosis infection: New targets for anti-TB therapeutics? Biochem Biophys Res Commun 2024; 711:149920. [PMID: 38615574 DOI: 10.1016/j.bbrc.2024.149920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Tuberculosis (TB), a deadly infectious disease induced by Mycobacterium tuberculosis (Mtb), continues to be a global public health issue that kill millions of patents every year. Despite significant efforts have been paid to identify effective TB treatments, the emergence of drug-resistant strains of the disease and the presence of comorbidities in TB patients urges us to explore the detailed mechanisms involved in TB immunity and develop more effective innovative anti-TB strategies. HIF-1α, a protein involved in regulating cellular immune responses during TB infection, has been highlighted as a promising target for the development of novel strategies for TB treatment due to its critical roles in anti-TB host immunity. This review provides a summary of current research progress on the roles of HIF-1α in TB infection, highlighting its importance in regulating the host immune response upon Mtb infection and summarizing the influences and mechanisms of HIF-1α on anti-TB immunological responses of host cells. This review also discusses the various challenges associated with developing HIF-1α as a target for anti-TB therapies, including ensuring specificity and avoiding off-target effects on normal cell function, determining the regulation and expression of HIF-1α in TB patients, and developing drugs that can inhibit HIF-1α. More deep understanding of the molecular mechanisms involved in HIF-1α signaling, its impact on TB host status, and systematic animal testing and clinical trials may benefit the optimization of HIF-1α as a novel therapeutic target for TB.
Collapse
Affiliation(s)
- Chaowei Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jiajun Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Biying Zheng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| |
Collapse
|
19
|
Zhao P, Hu HZ, Chen XT, Jiang QY, Yu XZ, Cen XL, Lin SQ, Mai SQ, Pang WL, Chen JX, Zhang Q. Mild hyperthermia enhanced synergistic uric acid degradation and multiple ROS elimination for an effective acute gout therapy. J Nanobiotechnology 2024; 22:275. [PMID: 38778401 PMCID: PMC11112921 DOI: 10.1186/s12951-024-02539-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Acute gouty is caused by the excessive accumulation of Monosodium Urate (MSU) crystals within various parts of the body, which leads to a deterioration of the local microenvironment. This degradation is marked by elevated levels of uric acid (UA), increased reactive oxygen species (ROS) production, hypoxic conditions, an upsurge in pro-inflammatory mediators, and mitochondrial dysfunction. RESULTS In this study, we developed a multifunctional nanoparticle of polydopamine-platinum (PDA@Pt) to combat acute gout by leveraging mild hyperthermia to synergistically enhance UA degradation and anti-inflammatory effect. Herein, PDA acts as a foundational template that facilitates the growth of a Pt shell on the surface of its nanospheres, leading to the formation of the PDA@Pt nanomedicine. Within this therapeutic agent, the Pt nanoparticle catalyzes the decomposition of UA and actively breaks down endogenous hydrogen peroxide (H2O2) to produce O2, which helps to alleviate hypoxic conditions. Concurrently, the PDA component possesses exceptional capacity for ROS scavenging. Most significantly, Both PDA and Pt shell exhibit absorption in the Near-Infrared-II (NIR-II) region, which not only endow PDA@Pt with superior photothermal conversion efficiency for effective photothermal therapy (PTT) but also substantially enhances the nanomedicine's capacity for UA degradation, O2 production and ROS scavenging enzymatic activities. This photothermally-enhanced approach effectively facilitates the repair of mitochondrial damage and downregulates the NF-κB signaling pathway to inhibit the expression of pro-inflammatory cytokines. CONCLUSIONS The multifunctional nanomedicine PDA@Pt exhibits exceptional efficacy in UA reduction and anti-inflammatory effects, presenting a promising potential therapeutic strategy for the management of acute gout.
Collapse
Affiliation(s)
- Pei Zhao
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Hua-Zhong Hu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Xiao-Tong Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qi-Yun Jiang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Xue-Zhao Yu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Xiao-Lin Cen
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Shi-Qing Lin
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Sui-Qing Mai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Wei-Lin Pang
- School of Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jin-Xiang Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Qun Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China.
| |
Collapse
|
20
|
Wang L, Zhang L, Chen F, Li Q, Zhu B, Tang Y, Yang Z, Cheng C, Qiu L, Ma L. Polymerized Network-Based Artificial Peroxisome Reprogramming Macrophages for Photoacoustic Imaging-Guided Treatment of Rheumatoid Arthritis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:25856-25868. [PMID: 38726921 DOI: 10.1021/acsami.4c04000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Artificial peroxisomes (AP) with enzyme-mimetic catalytic activity and recruitment ability have drawn a great deal of attention in fabricating protocell systems for scavenging reactive oxygen species (ROS), modulating the inflammatory microenvironment, and reprogramming macrophages, which is of great potential in treating inflammatory diseases such as rheumatoid arthritis (RA). Herein, a macrophage membrane-cloaked Cu-coordinated polyphthalocyanine-based AP (CuAP) is prepared with a macrocyclic conjugated polymerized network and embedded Cu-single atomic active center, which mimics the catalytic activity and coordination environment of natural superoxide dismutase and catalase, possesses the inflammatory recruitment ability of macrophages, and performs photoacoustic imaging (PAI)-guided treatment. The results of both in vitro cellular and in vivo animal experiments demonstrated that the CuAP under ultrasound and microbubbles could efficiently scavenge excess ROS in cells and tissues, modulate microenvironmental inflammatory cytokines such as interleukin-1β, tumor necrosis factor-α, and arginase-1, and reprogram macrophages by polarization of M1 (proinflammatory phenotype) to M2 (anti-inflammatory phenotype). We believe this study offers a proof of concept for engineering multifaceted AP and a promising approach for a PAI-guided treatment platform for RA.
Collapse
Affiliation(s)
- Liyun Wang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lingyan Zhang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fan Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Qian Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Bihui Zhu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuanjiao Tang
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengbao Yang
- Department of Mechanical and Aerospace Engineering, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong 999077, China
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Li Qiu
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lang Ma
- Department of Medical Ultrasound, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Li J, Ren H, Zhang Z, Zhang J, Wei F. Macrophage M2 polarization promotes pulpal inflammation resolution during orthodontic tooth movement. J Cell Mol Med 2024; 28:e18350. [PMID: 38700030 PMCID: PMC11066858 DOI: 10.1111/jcmm.18350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/10/2024] [Accepted: 04/11/2024] [Indexed: 05/05/2024] Open
Abstract
Mechanical force induces hypoxia in the pulpal area by compressing the apical blood vessels of the pulp, triggering pulpal inflammation during orthodontic tooth movement. However, this inflammation tends to be restorable. Macrophages are recognized as pivotal immunoreactive cells in the dental pulp. Whether they are involved in the resolution of pulpal inflammation in orthodontic teeth remains unclear. In this study, we investigated macrophage polarization and its effects during orthodontic tooth movement. It was demonstrated that macrophages within the dental pulp polarized to M2 type and actively participated in the process of pulpal inflammation resolution. Inflammatory reactions were generated and vascularization occurred in the pulp during orthodontic tooth movement. Macrophages in orthodontic pulp show a tendency to polarize towards M2 type as a result of pulpal hypoxia. Furthermore, by blocking M2 polarization, we found that macrophage M2 polarization inhibits dental pulp-secreting inflammatory factors and enhances VEGF production. In conclusion, our findings suggest that macrophages promote pulpal inflammation resolution by enhancing M2 polarization and maintaining dental health during orthodontic tooth movement.
Collapse
Affiliation(s)
- Jichang Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of MedicineShandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral DiseasesJinanShandongChina
| | - Huiying Ren
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of MedicineShandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral DiseasesJinanShandongChina
| | - Zijie Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of MedicineShandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral DiseasesJinanShandongChina
| | - Jin Zhang
- Department of Endodontics, School and Hospital of Stomatology, Cheeloo College of MedicineShandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral DiseasesJinanShandongChina
| | - Fulan Wei
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of MedicineShandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral DiseasesJinanShandongChina
| |
Collapse
|
22
|
Xiao J, Guo X, Wang Z. Crosstalk between hypoxia-inducible factor-1α and short-chain fatty acids in inflammatory bowel disease: key clues toward unraveling the mystery. Front Immunol 2024; 15:1385907. [PMID: 38605960 PMCID: PMC11007100 DOI: 10.3389/fimmu.2024.1385907] [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: 02/14/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024] Open
Abstract
The human intestinal tract constitutes a complex ecosystem, made up of countless gut microbiota, metabolites, and immune cells, with hypoxia being a fundamental environmental characteristic of this ecology. Under normal physiological conditions, a delicate balance exists among these complex "residents", with disruptions potentially leading to inflammatory bowel disease (IBD). The core pathology of IBD features a disrupted intestinal epithelial barrier, alongside evident immune and microecological disturbances. Central to these interconnected networks is hypoxia-inducible factor-1α (HIF-1α), which is a key regulator in gut cells for adapting to hypoxic conditions and maintaining gut homeostasis. Short-chain fatty acids (SCFAs), as pivotal gut metabolites, serve as vital mediators between the host and microbiota, and significantly influence intestinal ecosystem. Recent years have seen a surge in research on the roles and therapeutic potential of HIF-1α and SCFAs in IBD independently, yet reviews on HIF-1α-mediated SCFAs regulation of IBD under hypoxic conditions are scarce. This article summarizes evidence of the interplay and regulatory relationship between SCFAs and HIF-1α in IBD, pivotal for elucidating the disease's pathogenesis and offering promising therapeutic strategies.
Collapse
Affiliation(s)
- Jinyin Xiao
- Graduate School, Hunan University of Traditional Chinese Medicine, Changsha, China
- Department of Anorectal, the Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Xiajun Guo
- Department of Geriatric, the First People’s Hospital of Xiangtan City, Xiangtan, China
| | - Zhenquan Wang
- Department of Anorectal, the Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, China
| |
Collapse
|
23
|
Balan AI, Halațiu VB, Scridon A. Oxidative Stress, Inflammation, and Mitochondrial Dysfunction: A Link between Obesity and Atrial Fibrillation. Antioxidants (Basel) 2024; 13:117. [PMID: 38247541 PMCID: PMC10812976 DOI: 10.3390/antiox13010117] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
The adipose tissue has long been thought to represent a passive source of triglycerides and fatty acids. However, extensive data have demonstrated that the adipose tissue is also a major endocrine organ that directly or indirectly affects the physiological functions of almost all cell types. Obesity is recognized as a risk factor for multiple systemic conditions, including metabolic syndrome, type 2 diabetes mellitus, sleep apnea, cardiovascular disorders, and many others. Obesity-related changes in the adipose tissue induce functional and structural changes in cardiac myocytes, promoting a wide range of cardiovascular disorders, including atrial fibrillation (AF). Due to the wealth of epidemiologic data linking AF to obesity, the mechanisms underlying AF occurrence in obese patients are an area of rich ongoing investigation. However, progress has been somewhat slowed by the complex phenotypes of both obesity and AF. The triad inflammation, oxidative stress, and mitochondrial dysfunction are critical for AF pathogenesis in the setting of obesity via multiple structural and functional proarrhythmic changes at the level of the atria. The aim of this paper is to provide a comprehensive view of the close relationship between obesity-induced oxidative stress, inflammation, and mitochondrial dysfunction and the pathogenesis of AF. The clinical implications of these mechanistic insights are also discussed.
Collapse
Affiliation(s)
- Alkora Ioana Balan
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Vasile Bogdan Halațiu
- Physiology Department, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| | - Alina Scridon
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
- Physiology Department, University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș, 540142 Târgu Mureș, Romania;
| |
Collapse
|