1
|
Alshahrani AS, Saber S, Alruwaili OS, Al-Majdoub ZM, Hamad RS, Abdel-Reheim MA, Khaled BEA, Alibrahim A, Ramadan A, El-Kott AF, Alshehri AS, Negm S, Elmorsy EA, Khalifa AK, Abdelhady R. Modulation of FOXO3a Nuclear Localization by Linagliptin (BI-1356) reveals a new therapeutic target in chronic ulcerative colitis. Eur J Pharm Sci 2025; 209:107100. [PMID: 40221059 DOI: 10.1016/j.ejps.2025.107100] [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: 12/22/2024] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
Globally, the incidence and prevalence rates of ulcerative colitis (UC) show a rising pattern. The limited efficacy and significant adverse effects associated with current treatment options underscore the need for novel therapeutic approaches. It has been found that linagliptin, a dipeptidyl peptidase-4 inhibitor, activates AMPK in different disease conditions. The main objective of the present work was to elucidate the potential implications of the AMPK/FOXO3a mediated by linagliptin in rats with chronic colitis. The findings of the current report revealed the first robust in-vivo evidence advocating the coloprotective effect of linagliptin against dextran sodium sulfate-induced chronic UC in rats. It has demonstrated potential beyond its antidiabetic effects by modulating FOXO3a localization. By shifting FOXO3a from the cytosol to the nucleus, linagliptin enhanced the transcription of genes involved in attenuation of pro-inflammatory events and restoration of redox homeostasis. Nuclear FOXO3a also impacted NFκB activity, reducing inflammation. This conclusion was fundamentally supported by the documented improvements in histopathological changes evidenced by reduced inflammation, edema, crypt atrophy, and submucosal fibrosis. Moreover, decreased colon weight/length ratio, as well as reduced scores of disease activity and macroscopic damage indices, were observed. Furthermore, it corrected body weight loss during the time frame of the experiment. These findings underscore the anti-inflammatory potential of therapies that promote the nuclear localization of FOXO3a in inflammatory conditions. Linagliptin's ability to modulate FOXO3a localization might be particularly useful for diabetic patients suffering from inflammatory bowel diseases. However, further molecular investigations are required to validate the findings and to assess the clinical application of this approach as a valid tool for alleviating UC.
Collapse
Affiliation(s)
- Abdulaziz Saad Alshahrani
- Department of Internal Medicine, Medicine and Gastroenterologist Consultant, Najran University Hospital, Najran University, Saudi Arabia.
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | | | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK.
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia.
| | | | - Bahaa Eldin Ali Khaled
- Anatomy Department, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Alaa Alibrahim
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia.
| | - Asmaa Ramadan
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia; Department of Zoology, Faculty of Science, Damanhour University, Egypt.
| | - Ali S Alshehri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.
| | - Sally Negm
- Applied College, Health Specialities, Basic Sciences and Their Applications Unit, Mahayil Asir, King Khalid University, Abha, 62529, Saudi Arabia.
| | - Elsayed A Elmorsy
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah, 51452, Saudi Arabia.
| | - Amira Karam Khalifa
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt; Department of Medical Pharmacology, Faculty of Medicine, Nahda University, New Beni Suef 62521, Egypt.
| | - Rasha Abdelhady
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt; Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Chinese University, Cairo, Egypt.
| |
Collapse
|
2
|
Sen MK, Liao E, Ni D, Ge A, Piccio L. Immunomodulatory effects of calorie restriction and its mimetics: A new potential therapeutic approach for autoimmune diseases. Pharmacol Rev 2025; 77:100063. [PMID: 40449126 DOI: 10.1016/j.pharmr.2025.100063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 04/22/2025] [Indexed: 06/02/2025] Open
Abstract
Calorie restriction (CR) is a well known intervention associated with multifaceted anti-aging and pro-longevity health benefits. It induces complex physiological cellular and molecular adaptations, resulting in the fine-tuning of metabolic and immune responses in both homeostatic and diseased states. It has thus been extensively studied both preclinically and clinically, uncovering its therapeutic potential against inflammatory conditions, particularly autoimmune diseases. CR mimetics (CRMs), that is, molecules that mimic CR's effects, have also been widely investigated to counteract inflammatory states associated with numerous diseases, including autoimmunity. However, a comprehensive overview of how CR and CRMs modulate different aspects of immune responses, thereby potentially modifying autoimmunity, is still lacking. Here, we reviewed the latest progress on the impacts of CR and CRMs on the immune system and the current evidence on their potential translation in the clinical management of people with autoimmune diseases. First, we summarized different types of CR and CRMs and their main mechanisms of action. We next reviewed comprehensively how CR and CRMs modulate immune cells and discussed up-to-date preclinical and clinical advances in using CR and CRMs in the context of some of the most common autoimmune diseases. Finally, challenges faced in CR-related research and its translation into the clinic are discussed. SIGNIFICANCE STATEMENT: Calorie restriction (CR) encompasses various approaches for daily or intermittent reduction in calorie intake while maintaining adequate nutrient intake. It acts through cell-intrinsic and -extrinsic pathways to modulate immune cell functions. CR is emerging as a strategy for autoimmune disease management. CR's effects could be partially mimicked by molecules called CR mimetics, which are proposed to achieve CR's effects without reducing food intake. CR and CR mimetics have been tested as promising potential therapeutics in preclinical and clinical autoimmune disease studies.
Collapse
Affiliation(s)
- Monokesh K Sen
- Charles Perkins Centre, Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Eileen Liao
- Charles Perkins Centre, Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Duan Ni
- Charles Perkins Centre, Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anjie Ge
- Charles Perkins Centre, Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Laura Piccio
- Charles Perkins Centre, Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
3
|
Ma X, Lin Y, Zhang L, Huang Z, Zhang Y, Fu X, Li P. The dual missions of FoxO3a in inflammatory diseases: Regulation of antioxidant enzymes and involvement in programmed cell death. Int Immunopharmacol 2025; 151:114369. [PMID: 40031428 DOI: 10.1016/j.intimp.2025.114369] [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: 01/20/2025] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
The transcription factor FoxO3a plays a crucial role in the process of cells adapting to various stress conditions. Multiple post - translational modifications and epigenetic mechanisms work together to precisely regulate the activity of FoxO3a, influencing its subcellular localization, stability, interactions with other proteins, DNA - binding affinity, and transcriptional regulatory capacity. Under different chemical signal stimuli and subcellular environments, the activation of FoxO3a triggered by oxidative stress can initiate diverse transcriptional programs, which are essential for the body to resist oxidative damage. In the development and progression of inflammatory diseases, FoxO3a exerts an important function by regulating the expression levels of antioxidant enzymes and participating in key physiological processes such as programmed cell death. This article comprehensively reviews the structural characteristics, mechanism of action of FoxO3a, as well as its functions in regulating antioxidant enzymes and programmed cell death. The aim is to deeply explore the potential of FoxO3a as a potential therapeutic target for preventing and treating damages such as inflammatory diseases caused by cellular stress.
Collapse
Affiliation(s)
- Xiangli Ma
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Yujie Lin
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Ling Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhenzhen Huang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Yurong Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Xu Fu
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Peiwu Li
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
4
|
Wu Z, Zhan W, Wu L, Yu L, Xie X, Yu F, Kong W, Bi S, Liu S, Yin G, Zhou J. The Roles of Forkhead Box O3a (FOXO3a) in Bone and Cartilage Diseases - A Narrative Review. Drug Des Devel Ther 2025; 19:1357-1375. [PMID: 40034405 PMCID: PMC11874768 DOI: 10.2147/dddt.s494841] [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: 09/05/2024] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
Bone and cartilage diseases are significantly associated with musculoskeletal disability. However, no effective drugs are available to cure them. FOXO3a, a member of the FOXO family, has been implicated in cell proliferation, ROS detoxification, autophagy, and apoptosis. The biological functions of FOXO3a can be modulated by post-translational modifications (PTMs), such as phosphorylation and acetylation. Several signaling pathways, such as MAPK, NF-κB, PI3K/AKT, and AMPK/Sirt1 pathways, have been implicated in the development of bone and cartilage diseases by mediating the expression of FOXO3a. In particular, FOXO3a acts as a transcriptional factor in mediating the expression of various genes, such as MnSOD, CAT, BIM, BBC3, and CDK6. FOXO3a plays a critical role in the metabolism of bone and cartilage. In this article, we mainly discussed the biological functions of FOXO3a in bone and cartilage diseases, such as osteoporosis (OP), osteoarthritis (OA), rheumatoid arthritis (RA), ankylosing spondylitis (AS), and intervertebral disc degeneration (IDD). FOXO3a can promote osteogenic differentiation, induce osteoblast proliferation, inhibit osteoclast activity, suppress chondrocyte apoptosis, and reduce inflammatory responses. Collectively, up-regulation of FOXO3a expression shows beneficial effects, and FOXO3a has become a potential target for bone and cartilage diseases.
Collapse
Affiliation(s)
- Zhenyu Wu
- Department of Medical Imaging, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People’s Republic of China
- First Clinical Medical College, Gannan Medical University, Ganzhou, 341000, People’s Republic of China
| | - Wang Zhan
- First Clinical Medical College, Gannan Medical University, Ganzhou, 341000, People’s Republic of China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou, 341000, People’s Republic of China
| | - Luhu Yu
- Department of Clinical Laboratory, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Xunlu Xie
- Department of Pathology, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Fang Yu
- Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Weihao Kong
- Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Shengrong Bi
- Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Shiwei Liu
- Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Guoqiang Yin
- Department of Joint Surgery, Ganzhou Hospital Affiliated to Nanchang University, Ganzhou, 341000, People’s Republic of China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| |
Collapse
|
5
|
Zou P, Li J, Li J, Wang J. Inhibition of DNA Methyltransferase DNMT1 Reverses Th2 Response Polarisation and Alleviates Allergic Rhinitis. Clin Exp Pharmacol Physiol 2025; 52:e70015. [PMID: 39667746 DOI: 10.1111/1440-1681.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/28/2024] [Accepted: 11/24/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Type 2 T helper (Th2) cells-mediated immune response plays vital roles in allergic rhinitis (AR), and DNA methylation is previously found to be closely related to AR development. AIMS Our study aims to reveal the detail mechanism of DNA methylation affecting Th2 response in AR. METHODS Mice were stimulated with ovalbumin (OVA) to induce AR symptoms, and CD4+ T cells were subjected to Th2 induction culture. Real-time quantitative PCR, western blot, flow cytometry, and enzyme-linked immunosorbent assay were performed to analyse the activation of Th2 response. RESULTS DNA methyltransferase 1 (DNMT1) was significantly upregulated in OVA-induced AR model mice, and DNMT1 knockdown alleviated AR symptoms and pathological changes of nasal mucosa tissues in the model mice. DNMT1 knockdown obviously reduced the expression of GATA binding protein 3 (GATA3), the ratio of interleukin (IL)-4+CD4+ cells and the release of Th2 cytokines, but elevated the expression of T-box expressed in T cells (T-bet), the ratio of interferon (IFN)-γ+CD4+ cells and the levels of Th1 cytokines to improve Th1/Th2 imbalance in the model mice and Th2-induced CD4+T cells. Mechanistically, DNMT1 promoted promoter methylation of forkhead box O3 (FOXO3), inhibited FOXO3 expression and activated the nuclear factor kappa-B (NF-κB)/GATA3 signalling. FOXO3 overexpression remarkably inactivated the NF-κB/GATA3 pathway and mitigated Th2 polarisation in DNMT1-deficient and Th2-conditined CD4+T cells, which was reversed by a NF-κB inhibitor. CONCLUSION Altogether, DNMT1 downregulated FOXO3 expression to activate the NF-κB/GATA3 pathway and promote Th2 response in AR.
Collapse
Affiliation(s)
- Peng Zou
- Otolaryngology Head and Neck Surgery, The First People's Hospital of Kunming, Kunming, China
| | - Jianguo Li
- Ophthalmology Department, The First People's Hospital of Kunming, Kunming, China
| | - Jia Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, China
| | - Jian Wang
- Otolaryngology Head and Neck Surgery, The First People's Hospital of Kunming, Kunming, China
| |
Collapse
|
6
|
Wang X, Wang T, Zhu D, Wang J, Han W. From acute lung injury to cerebral ischemia: a unified concept involving intercellular communication through extracellular vesicle-associated miRNAs released by macrophages/microglia. Clin Exp Immunol 2025; 219:uxae105. [PMID: 39658101 PMCID: PMC11773807 DOI: 10.1093/cei/uxae105] [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: 04/30/2024] [Revised: 08/30/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024] Open
Abstract
Ischemic stroke and acute lung injury are prevalent life-threatening conditions marked by intricate molecular mechanisms and elevated mortality rates. Despite evident pathophysiological distinctions, a notable similarity exists in the gene responses to tissue injury observed in both pathologies. This similarity extends to both protein-encoding RNAs and non-coding RNAs. Extracellular vesicles (EVs) are nano-scale vesicles derived through cell secretion, possessing unique advantages such as high biocompatibility, low immunogenicity, intrinsic cell targeting, and facile chemical and genetic manipulation. Importantly, miRNAs, the most prevalent non-coding RNAs, are selectively concentrated within EVs. Macrophages/microglia serve as immune defense and homeostatic cells, deriving from progenitor cells in the bone marrow. They can be classified into two contrasting types: classical proinflammatory M1 phenotype or alternative anti-inflammatory M2 phenotype. However, there exists a continuum of various intermediate phenotypes between M1 and M2, and macrophages/microglia can transition from one phenotype to another. This review will investigate recent discoveries concerning the impact of EVs derived from macrophages/microglia under various states on the progression of ischemic stroke and acute lung injury. The focus will be on the involvement of miRNAs within these vesicles. The concluding remarks of this review will underscore the clinical possibilities linked to EV-miRNAs, accentuating their potential as both biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Xianbin Wang
- Department of Emergency Medicine, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Ting Wang
- Department of Radiology, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Dong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Jing Wang
- Graduate School of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Weijie Han
- Department of General Surgery, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| |
Collapse
|
7
|
Chen X, Yu L, Meng S, Zhao J, Huang X, Wang Z, Zhou Z, Huang Y, Hong T, Duan J, Su T, Cao Z, Chi Y, Huang T, Wang H. Inhibition of TREM-1 ameliorates angiotensin II-induced atrial fibrillation by attenuating macrophage infiltration and inflammation through the PI3K/AKT/FoxO3a signaling pathway. Cell Signal 2024; 124:111458. [PMID: 39384003 DOI: 10.1016/j.cellsig.2024.111458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
Inflammation and infiltration of immune cells are intricately linked to the pathogenesis of atrial fibrillation (AF). Triggering receptor expressed on myeloid cells-1 (TREM-1), an enhancer of inflammation, is implicated in various cardiovascular disorders. However, the precise role and potential mechanisms of TREM-1 in the development of AF remain ambiguous. Atrial samples from patients with AF were used to assess the expression levels of TREM-1. An angiotensin II (Ang II)-induced AF mouse model was established to assess the functionality of TREM-1. Cardiac function and AF inducibility were assessed through echocardiography, programmed transvenous cardiac pacing, and atrial electrophysiological mapping. Peripheral blood and atrial inflammatory cells were assessed using flow cytometry. Using histology, bulk RNA sequencing, biochemical analyses, and cell cultures, the mechanistic role of TREM-1 in AF was elucidated. TREM-1 expression was upregulated and co-localized with macrophages in the atria of patients with AF. Pharmacological inhibition of TREM-1 decreased Ang II-induced atrial enlargement and electrical remodeling. TREM-1 inhibition also ameliorated Ang II-induced NLRP3 inflammasome activation, inflammatory factor release, atrial fibrosis, and macrophage infiltration. Transcriptomic analysis revealed that TREM-1 modulates Ang II-induced inflammation through the PI3K/AKT/FoxO3a signaling pathway. In vitro studies further supported these findings, demonstrating that TREM-1 activation exacerbates Ang II-induced inflammation, while overexpression of FoxO3a counteracts this effect. This study discovered the critical role of TREM-1 in the pathogenesis of AF and its underlying molecular mechanisms. Inhibition of TREM-1 provides a new therapeutic strategy for the treatment of AF.
Collapse
Affiliation(s)
- Xin Chen
- Postgraduate Training Base of General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Liming Yu
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Shan Meng
- Postgraduate Training Base of General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jikai Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Xinyi Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Zhishang Wang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Zijun Zhou
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Yuting Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Tao Hong
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Postgraduate College, Dalian Medical University, Dalian, Liaoning 116000, PR China; Pediatric Surgery Ward, Fuwai Hospital Chinese Academy of Medical Sciences, ShenZhen 518000, PR China
| | - Jinfeng Duan
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Postgraduate College, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Tong Su
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110167, PR China
| | - Zijun Cao
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Postgraduate College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, PR China
| | - Yanbang Chi
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China; Department of Obstetrics and Gynecology, General Hospital of Northern Theater Command, Shenyang 110016, PR China
| | - Tao Huang
- State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| | - Huishan Wang
- Postgraduate Training Base of General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China; State Key Laboratory of Frigid Zone Cardiovascular Disease, Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
8
|
Kuo JF, Wu HY, Tung CW, Huang WH, Lin CS, Wang CC. Induction of Thymus Atrophy and Disruption of Thymocyte Development by Fipronil through Dysregulation of IL-7-Associated Genes. Chem Res Toxicol 2024; 37:1488-1500. [PMID: 39141674 PMCID: PMC11409377 DOI: 10.1021/acs.chemrestox.4c00060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
The susceptibility of the immune system to immunotoxic chemicals is evident, particularly in the thymus, a vital primary immune organ prone to atrophy due to exposure to toxicants. Fipronil (FPN), a widely used insecticide, is of concern due to its potential neurotoxicity, hepatotoxicity, and immunotoxicity. Our previous study showed that FPN disturbed the antigen-specific T-cell functionality in vivo. As T-cell lineage commitment and thymopoiesis are closely interconnected with the normal function of the T-cell-mediated immune responses, this study aims to further examine the toxic effects of FPN on thymocyte development. In this study, 4-week-old BALB/c mice received seven doses of FPN (1, 5, 10 mg/kg) by gavage. Thymus size, medulla/cortex ratio, total thymocyte counts, double-positive thymocyte population, and IL-7-positive cells decreased dose-dependently. IL-7 aids the differentiation of early T-cell precursors into mature T cells, and several essential genes contribute to the maturation of T cells in the thymus. Foxn1 ensures that the thymic microenvironment is suitable for the maturation of T-cell precursors. Lyl1 is involved in specifying lymphoid cells and maintaining T-cell development in the thymus. The c-Kit/SCF collaboration fosters a supportive thymic milieu to promote the formation of functional T cells. The expression of IL-7, IL-7R, c-Kit, SCF, Foxn1, and Lyl1 genes in the thymus was significantly diminished in FPN-treated groups with the concordance with the reduction of IL-7 signaling proteins (IL-7, IL-7R, c-KIT, SCF, LYL1, FOXO3A, and GABPA), suggesting that the dysregulation of T-cell lineage-related genes may contribute to the thymic atrophy induced by FPN. In addition, FPN disturbed the functionality of thymocytes with an increase of IL-4 and IFN-γ production and a decrease of IL-2 secretion after T-cell mitogen stimulation ex vivo. Collectively, FPN significantly deregulated genes related to T-cell progenitor differentiation, survival, and expansion, potentially leading to impaired thymopoiesis.
Collapse
Affiliation(s)
- Jui-Fang Kuo
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan
| | - Hsin-Ying Wu
- Laboratory Animal Center, National Health Research Institutes, Miaoli County 350, Taiwan
| | - Chun-Wei Tung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan
| | - Wei-Hsiang Huang
- Graduate Institute of Molecular and Comparative Pathobiology, National Taiwan University, Taipei 106, Taiwan
| | - Chen-Si Lin
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan
| | - Chia-Chi Wang
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
9
|
Maalouly G, Martin CMA, Baz Y, Saliba Y, Baramili AM, Fares N. Antioxidant and Anti-Apoptotic Neuroprotective Effects of Cinnamon in Imiquimod-Induced Lupus. Antioxidants (Basel) 2024; 13:880. [PMID: 39061948 PMCID: PMC11274315 DOI: 10.3390/antiox13070880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Despite accumulating evidence correlating oxidative stress with lupus disease activity, the brain redox pathways are still poorly investigated. Cinnamomum cassia, a widely used spice with powerful antioxidant properties, could be a novel therapeutic candidate in lupus. METHODS C57BL/6J female mice were divided into five groups: sham, sham-cinnamon, lupus, lupus-cinnamon starting from induction, and lupus-cinnamon starting two weeks before induction. Lupus was induced by skin application on the right ear with 1.25 mg of 5% imiquimod cream three times per week for six weeks. Cinnamomum cassia was given orally, five days per week, at 200 mg/kg. RESULTS Concomitant to TLR7-MYD88 pathway activation, the p-NRF2/NRF2 and p-FOXO3/FOXO3 ratios were increased in the hippocampus and alleviated by cinnamon treatment. BCL-2 positivity was enhanced in hippocampal neurons and reversed only by preventive cinnamon administration. In vitro, exposure of hippocampal cells to the plasma of different groups induced a surge in oxidative stress. This was associated with an increased t-BID/BID ratio. Cinnamon treatment, particularly in the preventive arm, normalized these modifications. CONCLUSIONS Our study shows a neuroprotective effect of cinnamon by rescuing brain redox and apoptosis homeostasis in lupus, paving the way for its use as a natural therapeutic compound in the clinical management of lupus.
Collapse
Affiliation(s)
| | | | | | | | | | - Nassim Fares
- Laboratory of Research in Physiology and Pathophysiology, Pole of Technology and Health, Faculty of Medicine, Saint Joseph University of Beirut, Beirut 1104 2020, Lebanon; (G.M.); (C.-M.-A.M.); (Y.B.); (Y.S.); (A.-M.B.)
| |
Collapse
|
10
|
Hu R, Xue X, Sun X, Mi Y, Wen H, Xi H, Li F, Zheng P, Liu S. Revealing the role of metformin in gastric intestinal metaplasia treatment. Front Pharmacol 2024; 15:1340309. [PMID: 39101145 PMCID: PMC11294171 DOI: 10.3389/fphar.2024.1340309] [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: 12/13/2023] [Accepted: 07/01/2024] [Indexed: 08/06/2024] Open
Abstract
Objective Gastric intestinal metaplasia (IM) is a precancerous stage associated with gastric cancer. Despite the observed beneficial effects of metformin on IM, its molecular mechanism remains not fully elucidated. This study aims to reveal the effects and potential mechanisms of metformin in treating IM based on both bioinformatics and in vivo investigations. Methods The seven public databases (GeneCards, DisGeNET, OMIM, SuperPred, Pharm Mapper, Swiss Target Prediction, TargetNet) were used in this work to identify targeted genes related to intestinal metaplasia (IM) and metformin. The shared targeted genes between metformin and IM were further analyzed by network pharmacology, while the interactions in-between were investigated by molecular docking. In parallel, the therapeutic effect of metformin was evaluated in IM mice model, while the core targets and pathways effected by metformin were verified in vivo. Results We screened out 1,751 IM-related genes and 318 metformin-targeted genes, 99 common genes identified in between were visualized by constructing the protein-protein interaction (PPI) network. The top ten core targeted genes were EGFR, MMP9, HIF1A, HSP90AA1, SIRT1, IL2, MAPK8, STAT1, PIK3CA, and ICAM1. The functional enrichment analysis confirmed that carcinogenesis and HIF-1 signaling pathways were primarily involved in the metformin treatment of IM. Based on molecular docking and dynamics, we found metformin affected the function of its targets by inhibiting receptor binding. Furthermore, metformin administration reduced the progression of IM lesions in Atp4a-/- mice model significantly. Notably, metformin enhanced the expression level of MUC5AC, while inhibited the expression level of CDX2. Our results also showed that metformin modulated the expression of core targets in vivo by reducing the activity of NF-κB and the PI3K/AKT/mTOR/HIF-1α signaling pathway. Conclusion This study confirms that metformin improves the efficacy of IM treatment by regulating a complex molecular network. Metformin plays a functional role in inhibiting inflammation/apoptosis-related pathways of further IM progression. Our work provides a molecular foundation for understanding metformin and other guanidine medicines in IM treatment.
Collapse
Affiliation(s)
- Ruoyu Hu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xia Xue
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangdong Sun
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Mi
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huijuan Wen
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huayuan Xi
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fuhao Li
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Simeng Liu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
11
|
Li W, Wang Z, Liang J, Xia B, Chen R, Chen T. Role of Medaka ( Oryzias latipes) Foxo3 in Resistance to Nervous Necrosis Virus Infection. Animals (Basel) 2024; 14:1587. [PMID: 38891634 PMCID: PMC11171044 DOI: 10.3390/ani14111587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Upon encountering a virus, fish initiate an innate immune response, guided by IFNs. Foxo3 plays a part in the body's immune response; however, its specific role in the IFN-guided immune response in fish is yet to be clarified. In this study, we characterized foxo3 in Japanese medaka (Oryzias latipes) and examined its role in the IFN-dependent immune response upon infection with the RGNNV. The results show that the coding region of the medaka foxo3 gene is 2007 base pairs long, encoding 668 amino acids, and possesses a typical forkhead protein family structural domain. The product of this gene shares high homology with foxo3 in other fish species and is widely expressed, especially in the brain, eyes, testes, and heart. Upon RGNNV infection, foxo3-/- mutant larvae showed a lower mortality rate, and adults exhibited a significant reduction in virus replication. Moreover, the absence of foxo3 expression led to an increase in the expression of irf3, and a decrease in the expression of other IFN-related genes such as tbk1 and mapk9, implying that foxo3 may function as a negative regulator in the antiviral signaling pathway. These findings provide crucial insights for disease-resistant breeding in the aquaculture industry.
Collapse
Affiliation(s)
- Wen Li
- State Key Laboratory of Mariculture Breeding, Fisheries College of Jimei University, Xiamen 361021, China; (W.L.); (J.L.); (R.C.)
- Engineering Research Center of the Modern Technology for Eel Industry, Xiamen 361021, China
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Xiamen 361021, China
| | - Zhi Wang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (Z.W.); (B.X.)
| | - Jingjie Liang
- State Key Laboratory of Mariculture Breeding, Fisheries College of Jimei University, Xiamen 361021, China; (W.L.); (J.L.); (R.C.)
- Engineering Research Center of the Modern Technology for Eel Industry, Xiamen 361021, China
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Xiamen 361021, China
| | - Bilin Xia
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (Z.W.); (B.X.)
| | - Ruoxue Chen
- State Key Laboratory of Mariculture Breeding, Fisheries College of Jimei University, Xiamen 361021, China; (W.L.); (J.L.); (R.C.)
- Engineering Research Center of the Modern Technology for Eel Industry, Xiamen 361021, China
| | - Tiansheng Chen
- State Key Laboratory of Mariculture Breeding, Fisheries College of Jimei University, Xiamen 361021, China; (W.L.); (J.L.); (R.C.)
- Engineering Research Center of the Modern Technology for Eel Industry, Xiamen 361021, China
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Xiamen 361021, China
| |
Collapse
|
12
|
Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, Zeng C, Zhou T, Zhang J. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther 2024; 9:53. [PMID: 38433280 PMCID: PMC10910037 DOI: 10.1038/s41392-024-01757-9] [Citation(s) in RCA: 427] [Impact Index Per Article: 427.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 03/05/2024] Open
Abstract
NF-κB signaling has been discovered for nearly 40 years. Initially, NF-κB signaling was identified as a pivotal pathway in mediating inflammatory responses. However, with extensive and in-depth investigations, researchers have discovered that its role can be expanded to a variety of signaling mechanisms, biological processes, human diseases, and treatment options. In this review, we first scrutinize the research process of NF-κB signaling, and summarize the composition, activation, and regulatory mechanism of NF-κB signaling. We investigate the interaction of NF-κB signaling with other important pathways, including PI3K/AKT, MAPK, JAK-STAT, TGF-β, Wnt, Notch, Hedgehog, and TLR signaling. The physiological and pathological states of NF-κB signaling, as well as its intricate involvement in inflammation, immune regulation, and tumor microenvironment, are also explicated. Additionally, we illustrate how NF-κB signaling is involved in a variety of human diseases, including cancers, inflammatory and autoimmune diseases, cardiovascular diseases, metabolic diseases, neurological diseases, and COVID-19. Further, we discuss the therapeutic approaches targeting NF-κB signaling, including IKK inhibitors, monoclonal antibodies, proteasome inhibitors, nuclear translocation inhibitors, DNA binding inhibitors, TKIs, non-coding RNAs, immunotherapy, and CAR-T. Finally, we provide an outlook for research in the field of NF-κB signaling. We hope to present a stereoscopic, comprehensive NF-κB signaling that will inform future research and clinical practice.
Collapse
Affiliation(s)
- Qing Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyu Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Xiaomin Ye
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xin Shen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Zeng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Teng Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Snarski P, Ghimire J, Savkovic SD. FOXO3: at the crossroads of metabolic, inflammatory, and tumorigenic remodeling in the colon. Am J Physiol Gastrointest Liver Physiol 2024; 326:G247-G251. [PMID: 38193202 PMCID: PMC11211034 DOI: 10.1152/ajpgi.00201.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/10/2024]
Abstract
The Forkhead box O3 (FOXO3) transcription factor regulates the expression of genes critical for diverse cellular functions in homeostasis. Diminished FOXO3 activity is associated with human diseases such as obesity, metabolic diseases, inflammatory diseases, and cancer. In the mouse colon, FOXO3 deficiency leads to an inflammatory immune landscape and dysregulated molecular pathways, which, under various insults, exacerbates inflammation and tumor burden, mimicking characteristics of human diseases. This deficiency also results in dysregulated lipid metabolism, and consequently, the accumulation of intracellular lipid droplets (LDs) in colonic epithelial cells and infiltrated immune cells. FOXO3 and LDs form a self-reinforcing negative regulatory loop in colonic epithelial cells, neutrophils, and macrophages, which is associated with inflammatory bowel disease and colon cancer, particularly in the context of obesity.
Collapse
Affiliation(s)
- Patricia Snarski
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Jenisha Ghimire
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, United States
| | - Suzana D Savkovic
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, United States
| |
Collapse
|
14
|
Bubak MP, Mann SN, Borowik AK, Pranay A, Batushansky A, Vieira de Sousa Neto I, Mondal SA, Doidge SM, Davidyan A, Szczygiel MM, Peelor FF, Rigsby S, Broomfield ME, Lacy CI, Rice HC, Stout MB, Miller BF. 17α-Estradiol alleviates high-fat diet-induced inflammatory and metabolic dysfunction in skeletal muscle of male and female mice. Am J Physiol Endocrinol Metab 2024; 326:E226-E244. [PMID: 38197793 PMCID: PMC11193529 DOI: 10.1152/ajpendo.00215.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/14/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
17α-estradiol (17α-E2) is a naturally occurring nonfeminizing diastereomer of 17β-estradiol that has life span-extending effects in rodent models. To date, studies of the systemic and tissue-specific benefits of 17α-E2 have largely focused on the liver, brain, and white adipose tissue with far less focus on skeletal muscle. Skeletal muscle has an important role in metabolic and age-related disease. Therefore, this study aimed to determine whether 17α-E2 treatment has positive, tissue-specific effects on skeletal muscle during a high-fat feeding. We hypothesized that male, but not female, mice, would benefit from 17α-E2 treatment during a high-fat diet (HFD) with changes in the mitochondrial proteome to support lipid oxidation and subsequent reductions in diacylglycerol (DAG) and ceramide content. To test this hypothesis, we used a multiomics approach to determine changes in lipotoxic lipid intermediates, metabolites, and proteins related to metabolic homeostasis. Unexpectedly, we found that 17α-E2 had marked, but different, beneficial effects within each sex. In male mice, we show that 17α-E2 alleviates HFD-induced metabolic detriments of skeletal muscle by reducing the accumulation of diacylglycerol (DAG), and inflammatory cytokine levels, and altered the abundance of most of the proteins related to lipolysis and β-oxidation. Similar to male mice, 17α-E2 treatment reduced fat mass while protecting muscle mass in female mice but had little muscle inflammatory cytokine levels. Although female mice were resistant to HFD-induced changes in DAGs, 17α-E2 treatment induced the upregulation of six DAG species. In female mice, 17α-E2 treatment changed the relative abundance of proteins involved in lipolysis, β-oxidation, as well as structural and contractile proteins but to a smaller extent than male mice. These data demonstrate the metabolic benefits of 17α-E2 in skeletal muscle of male and female mice and contribute to the growing literature of the use of 17α-E2 for multi tissue health span benefits.NEW & NOTEWORTHY Using a multiomics approach, we show that 17α-E2 alleviates HFD-induced metabolic detriments in skeletal muscle by altering bioactive lipid intermediates, inflammatory cytokines, and the abundance of proteins related to lipolysis and muscle contraction. The positive effects of 17α-E2 in skeletal muscle occur in both sexes but differ in their outcome.
Collapse
Affiliation(s)
- Matthew P Bubak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Shivani N Mann
- Department of Neuroscience, University of Arizona, Tucson, Arizona, United States
| | - Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Albert Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheba, Israel
| | - Ivo Vieira de Sousa Neto
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Samim A Mondal
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Stephen M Doidge
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Arik Davidyan
- Department of Biological Sciences, California State University, Sacramento, California, United States
| | - Marcelina M Szczygiel
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Sandra Rigsby
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Matle E Broomfield
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
| | - Charles I Lacy
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, United States
| | - Heather C Rice
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, United States
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma, United States
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
15
|
Khor YS, Wong PF. MicroRNAs-associated with FOXO3 in cellular senescence and other stress responses. Biogerontology 2024; 25:23-51. [PMID: 37646881 DOI: 10.1007/s10522-023-10059-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/01/2023] [Indexed: 09/01/2023]
Abstract
FOXO3 is a member of the FOXO transcription factor family and is known for regulating cellular survival in response to stress caused by various external and biological stimuli. FOXO3 decides cell fate by modulating cellular senescence, apoptosis and autophagy by transcriptional regulation of genes involved in DNA damage response and oxidative stress resistance. These cellular processes are tightly regulated physiologically, with FOXO3 acting as the hub that integrates signalling networks controlling them. The activity of FOXO3 is influenced by post-translational modifications, altering its subcellular localisation. In addition, FOXO3 can also be regulated directly or indirectly by microRNAs (miRNAs) or vice versa. This review discusses the involvement of various miRNAs in FOXO3-driven cellular responses such as senescence, apoptosis, autophagy, redox and inflammation defence. Given that these responses are linked and influence cell fate, a thorough understanding of the complex regulation by miRNAs would provide key information for developing therapeutic strategy and avoid unintended consequences caused by off-site targeting of FOXO3.
Collapse
Affiliation(s)
- Yi-Sheng Khor
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| |
Collapse
|
16
|
Kolary-Siekierska K, Niewiadomski P, Namieciński W, Miłoński J. Title Expression of FOXO3 and MAPK1 Genes in Patients with Benign Salivary Gland Tumors. J Clin Med 2023; 13:215. [PMID: 38202222 PMCID: PMC10779754 DOI: 10.3390/jcm13010215] [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: 11/21/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Pleomorphic adenomas (PAs) and Warthin tumors (WTs) are the most common benign tumors that occur in the salivary gland. PA has a tendency towards malignant transformation. Thus, searching for new methods to diagnose salivary gland tumors and treatment is important. The members of the class O forehead box transcription factor (FOXO3) and mitogen-activated protein kinase 1 (MAPK1) genes participate in the cellular processes, including in cell proliferation. The aim of this study was to analyze these genes' expression in the salivary gland tissues and in salivary gland tumors. The study group consisted of 50 patients treated for salivary gland tumors. For genetic tests, fresh samples of tissue collected during the surgery were used. The expression levels of the FOXO3 and MAPK1 genes were statistically significantly lower in PA tissue than in normal salivary gland tissue and WT tissue. This research revealed that the FOXO3 and MAPK1 genes are present in benign salivary gland tumors and also indicated a role of these genes in the development of benign salivary gland tumors. The cause of the development of pleomorphic adenomas may be apoptotic disorder and the activation of the inflammatory process. The examined genes may have potential to be new therapeutic targets for the treatment of pleomorphic adenomas.
Collapse
Affiliation(s)
- Katarzyna Kolary-Siekierska
- Department of Otolaryngology and Laryngological Oncology, Audiology and Phoniatrics, Medical University of Lodz, 90-549 Lodz, Poland
| | | | | | | |
Collapse
|
17
|
Gellhaus B, Böker KO, Schilling AF, Saul D. Therapeutic Consequences of Targeting the IGF-1/PI3K/AKT/FOXO3 Axis in Sarcopenia: A Narrative Review. Cells 2023; 12:2787. [PMID: 38132107 PMCID: PMC10741475 DOI: 10.3390/cells12242787] [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: 11/06/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
The high prevalence of sarcopenia in an aging population has an underestimated impact on quality of life by increasing the risk of falls and subsequent hospitalization. Unfortunately, the application of the major established key therapeutic-physical activity-is challenging in the immobile and injured sarcopenic patient. Consequently, novel therapeutic directions are needed. The transcription factor Forkhead-Box-Protein O3 (FOXO3) may be an option, as it and its targets have been observed to be more highly expressed in sarcopenic muscle. In such catabolic situations, Foxo3 induces the expression of two muscle specific ubiquitin ligases (Atrogin-1 and Murf-1) via the PI3K/AKT pathway. In this review, we particularly evaluate the potential of Foxo3-targeted gene therapy. Foxo3 knockdown has been shown to lead to increased muscle cross sectional area, through both the AKT-dependent and -independent pathways and the reduced impact on the two major downstream targets Atrogin-1 and Murf-1. Moreover, a Foxo3 reduction suppresses apoptosis, activates satellite cells, and initiates their differentiation into muscle cells. While this indicates a critical role in muscle regeneration, this mechanism might exhaust the stem cell pool, limiting its clinical applicability. As systemic Foxo3 knockdown has also been associated with risks of inflammation and cancer progression, a muscle-specific approach would be necessary. In this review, we summarize the current knowledge on Foxo3 and conceptualize a specific and targeted therapy that may circumvent the drawbacks of systemic Foxo3 knockdown. This approach presumably would limit the side effects and enable an activity-independent positive impact on skeletal muscle.
Collapse
Affiliation(s)
- Benjamin Gellhaus
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Kai O. Böker
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Arndt F. Schilling
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
| | - Dominik Saul
- Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August University of Goettingen, 37075 Goettingen, Germany; (B.G.); (K.O.B.); (A.F.S.)
- Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72072 Tuebingen, Germany
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
18
|
Gao Y, Zhang Y, Liu W, Zhang N, Gao Q, Shangguan J, Li N, Zhao Y, Jia Y. Danggui Buxue decoction alleviates cyclophosphamide-induced myelosuppression by regulating β-hydroxybutyric acid metabolism and suppressing oxidative stress. PHARMACEUTICAL BIOLOGY 2023; 61:710-721. [PMID: 37096658 PMCID: PMC10132245 DOI: 10.1080/13880209.2023.2201606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
CONTEXT Danggui Buxue Decoction (DBD) is an effective complementary medicine in alleviating myelosuppression after chemotherapy (MAC). However, its mechanism of action is elusive. OBJECTIVE To illustrate that regulating β-hydroxybutyric acid (β-OHB) metabolism and suppressing oxidative stress could be a potential mechanism of action for DBD in alleviating MAC. MATERIALS AND METHODS After HPLC quantification and dose testing (3, 6 and 10 g/kg, gavage) of DBD, Sprague-Dawley rats were divided into control, cyclophosphamide (CTX) (30 mg/kg CTX for 5 days, intraperitoneal administration) and CTX + DBD groups (6 g/kg DBD for 14 days, gavage). Blood cell counts, thigh bone histological examination, β-OHB levels, oxidative stress indices and HDAC1 activity were tested. The biological function of β-OHB was verified in vitro (hBMSC cells were incubated in culture mediums that contained 40 μM CTX and β-OHB in 0, 1, 2.5, 5, 10 mM) and in vivo (MAC rat model, 3 g/kg β-OHB for 14 days, gavage). RESULTS Rats in the CTX + DBD group showed upregulated blood cell counts (118-243%), β-OHB levels (495 nmol/mL in blood, 122 nmol/mg in marrow supernatant) and downregulated HDAC1 activity (59%), and oxidative stress indices (60-85%). In vitro, 5 mM β-OHB improved hBMSC cell migration (123%) and proliferation (131%). In vivo, rats treated with 3 g/kg β-OHB showed upregulated blood cell counts (121-182%) and downregulated HDAC1 activity (64%) and oxidative stress indices (65-83%). DISCUSSION AND CONCLUSIONS DBD, a traditional Chinese medicine, alleviates MAC by intervening in β-OHB metabolism and oxidative stress.
Collapse
Affiliation(s)
- Yiqiao Gao
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
- Xinxiang Key Laboratory of Clinical Psychopharmacology, Xinxiang Medical University, Xinxiang, P. R. China
- CONTACT Yiqiao Gao
| | - Yixin Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
- Xinxiang Key Laboratory of Clinical Psychopharmacology, Xinxiang Medical University, Xinxiang, P. R. China
| | - Wei Liu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
| | - Nan Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
| | - Qinghe Gao
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
| | | | - Na Li
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
- Xinxiang Key Laboratory of Clinical Psychopharmacology, Xinxiang Medical University, Xinxiang, P. R. China
| | - Ying Zhao
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
- Xinxiang Key Laboratory of Clinical Psychopharmacology, Xinxiang Medical University, Xinxiang, P. R. China
| | - Yanlong Jia
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
- Yanlong Jia School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan453003, P. R. China
| |
Collapse
|
19
|
Liu Q, Zhang X, Mao P, Wang Z, Mao Q, Wang C, Liu J, Zhu X, Wang B, Wei H. Shuangshi Tonglin capsule improves chronic prostatitis through the SIRT-1/AMPK and MAPK signalling pathways. Heliyon 2023; 9:e21745. [PMID: 38027908 PMCID: PMC10663862 DOI: 10.1016/j.heliyon.2023.e21745] [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: 05/04/2023] [Revised: 09/01/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Objectives To explore the effects of the Shuangshi Tonglin (SSTL) capsule on CP/CPPS and reveal the therapeutic mechanisms. Methods A CP/CPPS rat-model group received an intraprostatic injection of CFA. SSTL capsule were administered daily by oral gavage at doses of 1.25, 2.5, and 5.0 g/kg for 28 days. Pain threshold tests were performed, and prostate and blood samples were collected. We performed histological analysis of the prostate tissue and immunohistochemical analysis of TNF-α and COX-2. Measure the TNF-α levels, detect antioxidant levels in serum and prostate tissue, and evaluate the expression of proteins with the AMPK/SIRT-1 and MAPK signalling pathways. Results After SSTL capsule treatment, all animals exhibited an increased mechanical pain threshold in the lower abdomen, decreased inflammation in the stroma, and reduced histological structural damage. Inflammation was reduced through the observed decrease in the levels of various inflammatory factors, as well as in the increase of the levels of MDA, p-AMPK, and SIRT-1. The suppression of IKKβ, p-P38, p-ERK and p-JNK was also observed. Conclusions SSTL capsule treatment decreased inflammation in the stroma and reduced histological structural damage. It improved CP/CPPS symptoms by inhibiting oxidative stress and inflammation. Our study indicates that the SSTL capsule is an effective treatment for prostatitis.
Collapse
Affiliation(s)
- Qing Liu
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Xinyue Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Peng Mao
- Shaanxi Momentum Pharmaceutical Co., Ltd., Xianyang, 712000, Shaanxi, China
| | - Ziqiang Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Qian Mao
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Chuan Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Jiping Liu
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Xingmei Zhu
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Baoan Wang
- Shaanxi Momentum Pharmaceutical Co., Ltd., Xianyang, 712000, Shaanxi, China
| | - Hao Wei
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| |
Collapse
|
20
|
Donlon TA, Morris BJ, Chen R, Lim E, Morgen EK, Fortney K, Shah N, Masaki KH, Willcox BJ. Proteomic basis of mortality resilience mediated by FOXO3 longevity genotype. GeroScience 2023; 45:2303-2324. [PMID: 36881352 PMCID: PMC10651822 DOI: 10.1007/s11357-023-00740-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/23/2023] [Indexed: 03/08/2023] Open
Abstract
FOXO3 is a ubiquitous transcription factor expressed in response to cellular stress caused by nutrient deprivation, inflammatory cytokines, reactive oxygen species, radiation, hypoxia, and other factors. We showed previously that the association of inherited FOXO3 variants with longevity was the result of partial protection against mortality risk posed by aging-related life-long stressors, particularly cardiometabolic disease. We then referred to the longevity-associated genotypes as conferring "mortality resilience." Serum proteins whose levels change with aging and are associated with mortality risk may be considered as "stress proteins." They may serve as indirect measures of life-long stress. Our aims were to (1) identify stress proteins that increase with aging and are associated with an increased risk of mortality, and (2) to determine if FOXO3 longevity/resilience genotype dampens the expected increase in mortality risk they pose. A total of 4500 serum protein aptamers were quantified using the Somalogic SomaScan proteomics platform in the current study of 975 men aged 71-83 years. Stress proteins associated with mortality were identified. We then used age-adjusted multivariable Cox models to investigate the interaction of stress protein with FOXO3 longevity-associated rs12212067 genotypes. For all the analyses, the p values were corrected for multiple comparisons by false discovery rate. This led to the identification of 44 stress proteins influencing the association of FOXO3 genotype with reduced mortality. Biological pathways were identified for these proteins. Our results suggest that the FOXO3 resilience genotype functions by reducing mortality in pathways related to innate immunity, bone morphogenetic protein signaling, leukocyte migration, and growth factor response.
Collapse
Affiliation(s)
- Timothy A Donlon
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Brian J Morris
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA.
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA.
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia.
| | - Randi Chen
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
| | - Eunjung Lim
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Eric K Morgen
- BioAge Labs Inc., 1445A S 50th St, Richmond, California, USA
| | - Kristen Fortney
- BioAge Labs Inc., 1445A S 50th St, Richmond, California, USA
| | - Naisha Shah
- BioAge Labs Inc., 1445A S 50th St, Richmond, California, USA
| | - Kamal H Masaki
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Bradley J Willcox
- Department of Research, NIH Center of Biomedical Research Excellence for Clinical and Translational Research on Aging, Kuakini Medical Center, Honolulu, Hawaii, 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| |
Collapse
|
21
|
Cao G, Lin M, Gu W, Su Z, Duan Y, Song W, Liu H, Zhang F. The rules and regulatory mechanisms of FOXO3 on inflammation, metabolism, cell death and aging in hosts. Life Sci 2023:121877. [PMID: 37352918 DOI: 10.1016/j.lfs.2023.121877] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
The FOX family of transcription factors was originally identified in 1989, comprising the FOXA to FOXS subfamilies. FOXO3, a well-known member of the FOXO subfamily, is widely expressed in various human organs and tissues, with higher expression levels in the ovary, skeletal muscle, heart, and spleen. The biological effects of FOXO3 are mostly determined by its phosphorylation, which occurs in the nucleus or cytoplasm. Phosphorylation of FOXO3 in the nucleus can promote its translocation into the cytoplasm and inhibit its transcriptional activity. In contrast, phosphorylation of FOXO3 in the cytoplasm leads to its translocation into the nucleus and exerts regulatory effects on biological processes, such as inflammation, aerobic glycolysis, autophagy, apoptosis, oxidative stress, cell cycle arrest and DNA damage repair. Additionally, FOXO3 isoform 2 acts as an important suppressor of osteoclast differentiation. FOXO3 can also interfere with the development of various diseases, including inhibiting the proliferation and invasion of tumor cells, blocking the production of inflammatory factors in autoimmune diseases, and inhibiting β-amyloid deposition in Alzheimer's disease. Furthermore, FOXO3 slows down the aging process and exerts anti-aging effects by delaying telomere attrition, promoting cell self-renewal, and maintaining genomic stability. This review suggests that changes in the levels and post-translational modifications of FOXO3 protein can maintain organismal homeostasis and improve age-related diseases, thus counteracting aging. Moreover, this may indicate that alterations in FOXO3 protein levels are also crucial for longevity, offering new perspectives for therapeutic strategies targeting FOXO3.
Collapse
Affiliation(s)
- Guoding Cao
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Heilongjiang Key Laboratory of Immunity and Infection, Harbin 150081, China
| | - Monan Lin
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Heilongjiang Key Laboratory of Immunity and Infection, Harbin 150081, China
| | - Wei Gu
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Heilongjiang Key Laboratory of Immunity and Infection, Harbin 150081, China
| | - Zaiyu Su
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Heilongjiang Key Laboratory of Immunity and Infection, Harbin 150081, China
| | - Yagan Duan
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Heilongjiang Key Laboratory of Immunity and Infection, Harbin 150081, China
| | - Wuqi Song
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Heilongjiang Key Laboratory of Immunity and Infection, Harbin 150081, China
| | - Hailiang Liu
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Heilongjiang Key Laboratory of Immunity and Infection, Harbin 150081, China.
| | - Fengmin Zhang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Heilongjiang Key Laboratory of Immunity and Infection, Harbin 150081, China.
| |
Collapse
|
22
|
Ghimire J, Iftikhar R, Penrose HM, Snarski P, Ruiz E, Savkovic SD. FOXO3 Deficiency in Neutrophils Drives Colonic Inflammation and Tumorigenesis. Int J Mol Sci 2023; 24:ijms24119730. [PMID: 37298680 DOI: 10.3390/ijms24119730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/19/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Inflammatory bowel disease (IBD), characterized by infiltration of polymorphonuclear neutrophils (PMNs), increases the risk of colon cancer. PMN activation corresponds to the accumulation of intracellular Lipid Droplets (LDs). As increased LDs are negatively regulated by transcription factor Forkhead Box O3 (FOXO3), we aim to determine the significance of this regulatory network in PMN-mediated IBD and tumorigenesis. Affected tissue of IBD and colon cancer patients, colonic and infiltrated immune cells, have increased LDs' coat protein, PLIN2. Mouse peritoneal PMNs with stimulated LDs and FOXO3 deficiency have elevated transmigratory activity. Transcriptomic analysis of these FOXO3-deficient PMNs showed differentially expressed genes (DEGs; FDR < 0.05) involved in metabolism, inflammation, and tumorigenesis. Upstream regulators of these DEGs, similar to colonic inflammation and dysplasia in mice, were linked to IBD and human colon cancer. Additionally, a transcriptional signature representing FOXO3-deficient PMNs (PMN-FOXO3389) separated transcriptomes of affected tissue in IBD (p = 0.00018) and colon cancer (p = 0.0037) from control. Increased PMN-FOXO3389 presence predicted colon cancer invasion (lymphovascular p = 0.015; vascular p = 0.046; perineural p = 0.03) and poor survival. Validated DEGs from PMN-FOXO3389 (P2RX1, MGLL, MCAM, CDKN1A, RALBP1, CCPG1, PLA2G7) are involved in metabolism, inflammation, and tumorigenesis (p < 0.05). These findings highlight the significance of LDs and FOXO3-mediated PMN functions that promote colonic pathobiology.
Collapse
Affiliation(s)
- Jenisha Ghimire
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Rida Iftikhar
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Harrison M Penrose
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Patricia Snarski
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Emmanuelle Ruiz
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Suzana D Savkovic
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
23
|
Ahmad SM, Bhat SS, Shafi S, Dar MA, Saleem A, Haq Z, Farooq N, Nazir J, Bhat B. Identification of key transcription factors and their functional role involved in Salmonella typhimurium infection in chicken using integrated transcriptome analysis and bioinformatics approach. BMC Genomics 2023; 24:214. [PMID: 37098463 PMCID: PMC10127038 DOI: 10.1186/s12864-023-09315-3] [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: 10/21/2022] [Accepted: 04/16/2023] [Indexed: 04/27/2023] Open
Abstract
Salmonella enterica serovar typhimurium is the cause of significant morbidity and mortality worldwide that causes economic losses to poultry and is able to cause infection in humans. Indigenous chicken breeds are a potential source of animal protein and have the added advantage of being disease resistant. An indigenous chicken, Kashmir favorella and commercial broiler were selected for understanding the mechanism of disease resistance. Following infection in Kashmir favorella, three differentially expressed genes Nuclear Factor Kappa B (NF-κB1), Forkhead Box Protein O3 (FOXO3) and Paired box 5 (Pax5) were identified. FOXO3, a transcriptional activator, is the potential marker of host resistance in Salmonella infection. NF-κB1 is an inducible transcription factor which lays the foundation for studying gene network of the innate immune response of Salmonella infection in chicken. Pax5 is essential for differentiation of pre-B cells into mature B cell. The real time PCR analysis showed that in response to Salmonella Typhimurium infection a remarkable increase of NF-κB1 (P˂0.01), FOXO3 (P˂0.01) gene expression in liver and Pax5 (P˂0.01) gene expression in spleen of Kashmir favorella was observed. The protein-protein interaction (PPI) and protein-TF interaction network by STRINGDB analysis suggests that FOXO3 is a hub gene in the network and is closely related to Salmonella infection along with NF-κB1. All the three differentially expressed genes (NF-κB1, FOXO3 and PaX5) showed their influence on 12 interacting proteins and 16 TFs, where cyclic adenosine monophosphate Response Element Binding protein (CREBBP), erythroblast transformation-specific (ETSI), Tumour-protein 53(TP53I), IKKBK, lymphoid enhancer-binding factor-1 (LEF1), and interferon regulatory factor-4 (IRF4) play role in immune responses. This study shall pave the way for newer strategies for treatment and prevention of Salmonella infection and may help in increasing the innate disease resistance.
Collapse
Affiliation(s)
- Syed Mudasir Ahmad
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India.
| | - Sahar Saleem Bhat
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Shaista Shafi
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, India
| | - Mashooq Ahmad Dar
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Afnan Saleem
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Zulfqarul Haq
- Division of Livestock Production and Management, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Nida Farooq
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Junaid Nazir
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India
| | - Basharat Bhat
- Division of Animal Biotechnology, FVSc & AH, Shuhama, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190006, India.
| |
Collapse
|
24
|
Ishii K, Tamura T, Hatori K, Himi K, Nakamura T, Toyama Y, Miyata T, Takeichi O. Elevated Foxo3a and Fas-ligand expression in human periapical granulomas as a potential treatment target. Oral Dis 2023; 29:1128-1136. [PMID: 34674361 DOI: 10.1111/odi.14052] [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: 06/29/2021] [Revised: 09/12/2021] [Accepted: 10/12/2021] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Periapical granuloma is a common periodontitis type involving chronic inflammation; however, the efficacy of current therapies is limited. Its molecular pathogenesis also remains obscure. Forkhead box transcription factor class o3a (Foxo3a) and Fas-ligand (FasL) are associated with chronic inflammation. Therefore, in this study, we aimed to clarify the roles of Foxo3a and FasL in periapical granuloma pathophysiology. SUBJECTS AND METHODS Periapical lesions were obtained from patients during endodontic surgery and tooth extraction; those diagnosed with periapical granulomas using haematoxylin and eosin staining were further analysed. Immunohistochemical analysis was performed for Foxo3a and FasL, and real-time polymerase chain reaction was performed for FOXO3A, FASL and interleukin (IL)-1β. Healthy gingival tissues were also examined as controls. RESULTS Neutrophils, lymphocytes and plasma cells in the periapical granulomas, but not healthy tissues, expressed Foxo3a. Dual-colour immunofluorescence imaging revealed Foxo3a and FasL co-expression in leukocytes. FOXO3A, FASL and IL-1β mRNA levels in healthy gingival tissues were significantly lower than those in the periapical granulomas. Additionally, FOXO3A and IL-1β expressions were negatively correlated. CONCLUSIONS Phosphorylated Foxo3a may reduce IL-1β release by inhibiting apoptosis through FasL in periapical periodontitis and prevent exacerbation. Thus, Foxo3a is a potential therapeutic agent for periapical periodontitis.
Collapse
Affiliation(s)
- Kae Ishii
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan
| | - Takahito Tamura
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan
- Nihon University Graduate School of Dentistry, Dental Research Center, Tokyo, Japan
| | - Keisuke Hatori
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan
- Division of Advanced Dental Treatment, Dental Research Center, Tokyo, Japan
| | - Kazuma Himi
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan
| | - Takeshi Nakamura
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan
- Nihon University Graduate School of Dentistry, Dental Research Center, Tokyo, Japan
| | - Yurika Toyama
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan
- Nihon University Graduate School of Dentistry, Dental Research Center, Tokyo, Japan
| | - Taiki Miyata
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan
- Nihon University Graduate School of Dentistry, Dental Research Center, Tokyo, Japan
| | - Osamu Takeichi
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan
- Division of Advanced Dental Treatment, Dental Research Center, Tokyo, Japan
| |
Collapse
|
25
|
Abstract
BACKGROUND Metformin has good anti-hyperglycemic effectiveness, but does not induce hypoglycemia,is very safe, and has become the preferred drug for the treatment of type 2 diabetes. Recently, the other effects of metformin, such as being anti-inflammatory and delaying aging, have also attracted increased attention. METHODS AND RESULTS The relevant literatures on pubmed and other websites for reading, classification and sorting, and did not involve any animal experiments. CONCLUSION Metformin has anti-inflammatory effects through multiple routes, which provides potential therapeutic targets for certain inflammatory diseases, such as neuroinflammation and rheumatoid arthritis. In addition, inflammation is a key component of tumor occurrence and development ; thus, targeted inflammatory intervention is a significant benefit for both cancer prevention and treatment. Therefore, metformin may have further potential for inflammation-related disease prevention and treatmen. However, the inflammatory mechanism is complex; various molecules are connected and influence each other. For example, metformin significantly inhibits p65 nuclear translocation, but pretreatment with compound C, an AMPK inhibitor, abolishes this effect, and silencing of HMGB1 inhibits NF-κB activation . SIRT1 deacetylates FoxO, increasing its transcriptional activity . mTOR in dendritic cells regulates FoxO1 via AKT. The interactions among various molecules should be further explored to clarify their specific mechanisms and provide more direction for the treatment of inflammatory diseases, as well as cancer.
Collapse
|
26
|
Innate Immunity in Cardiovascular Diseases-Identification of Novel Molecular Players and Targets. J Clin Med 2023; 12:jcm12010335. [PMID: 36615135 PMCID: PMC9821340 DOI: 10.3390/jcm12010335] [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: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
During the past few years, unexpected developments have driven studies in the field of clinical immunology. One driver of immense impact was the outbreak of a pandemic caused by the novel virus SARS-CoV-2. Excellent recent reviews address diverse aspects of immunological re-search into cardiovascular diseases. Here, we specifically focus on selected studies taking advantage of advanced state-of-the-art molecular genetic methods ranging from genome-wide epi/transcriptome mapping and variant scanning to optogenetics and chemogenetics. First, we discuss the emerging clinical relevance of advanced diagnostics for cardiovascular diseases, including those associated with COVID-19-with a focus on the role of inflammation in cardiomyopathies and arrhythmias. Second, we consider newly identified immunological interactions at organ and system levels which affect cardiovascular pathogenesis. Thus, studies into immune influences arising from the intestinal system are moving towards therapeutic exploitation. Further, powerful new research tools have enabled novel insight into brain-immune system interactions at unprecedented resolution. This latter line of investigation emphasizes the strength of influence of emotional stress-acting through defined brain regions-upon viral and cardiovascular disorders. Several challenges need to be overcome before the full impact of these far-reaching new findings will hit the clinical arena.
Collapse
|
27
|
Dorai S, Alex Anand D. Differentially Expressed Cell Cycle Genes and STAT1/3-Driven Multiple Cancer Entanglement in Psoriasis, Coupled with Other Comorbidities. Cells 2022; 11:cells11233867. [PMID: 36497125 PMCID: PMC9740537 DOI: 10.3390/cells11233867] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Psoriasis is a persistent T-cell-supported inflammatory cutaneous disorder, which is defined by a significant expansion of basal cells in the epidermis. Cell cycle and STAT genes that control cell cycle progression and viral infection have been revealed to be comorbid with the development of certain cancers and other disorders, due to their abnormal or scanty expression. The purpose of this study is to evaluate the expression of certain cell cycle and STAT1/3 genes in psoriasis patients and to determine the types of comorbidities associated with these genes. To do so, we opted to adopt the in silico methodology, since it is a quick and easy way to discover any potential comorbidity risks that may exist in psoriasis patients. With the genes collected from early research groups, protein networks were created in this work using the NetworkAnalyst program. The crucial hub genes were identified by setting the degree parameter, and they were then used in gene ontology and pathway assessments. The transcription factors that control the hub genes were detected by exploring TRRUST, and DGIdb was probed for remedies that target transcription factors and hubs. Using the degree filter, the first protein subnetwork produced seven hub genes, including STAT3, CCNB1, STAT1, CCND1, CDC20, HSPA4, and MAD2L1. The hub genes were shown to be implicated in cell cycle pathways by the gene ontology and Reactome annotations. The former four hubs were found in signaling pathways, including prolactin, FoxO, JAK/STAT, and p53, according to the KEGG annotation. Furthermore, they enhanced several malignancies, including pancreatic cancer, Kaposi's sarcoma, non-small cell lung cancer, and acute myeloid leukemia. Viral infections, including measles, hepatitis C, Epstein-Barr virus, and HTLV-1 and viral carcinogenesis were among the other susceptible diseases. Diabetes and inflammatory bowel disease were conjointly annotated. In total, 129 medicines were discovered in DGIdb to be effective against the transcription factors BRCA1, RELA, TP53, and MYC, as opposed to 10 medications against the hubs, STAT3 and CCND1, in tandem with 8 common medicines. The study suggests that the annotated medications should be tested in suitable psoriatic cell lines and animal models to optimize the drugs used based on the kind, severity, and related comorbidities of psoriasis. Furthermore, a personalized medicine protocol must be designed for each psoriasis patient that displays different comorbidities.
Collapse
|
28
|
Zhao Y, Guo Y, Sun M, Hussion S, Zheng Y, Huang H, Huo X, Zhao Y, Zhang F, Han Y, Ning Q, Xu P, Sun J, Lu S. Selenium-sensitive histone deacetylase 2 is required for forkhead box O3A and regulates extracellular matrix metabolism in cartilage. J Bone Miner Metab 2022; 40:914-926. [PMID: 36156740 DOI: 10.1007/s00774-022-01369-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/24/2022] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Selenium (Se) as well as selenoproteins are vital for osteochondral system development. Se deficiency (SeD) has a definite impact on the expression and activity of histone deacetylases (HDACs). Abnormal expression of some HDACs affects cartilage development. This current study aims to explore the relationship between differentially expressed HDACs and cartilage development, especially extracellular matrix (ECM) homeostasis maintenance, under SeD conditions. MATERIALS AND METHODS Dark Agouti rats and C28/I2 cell line under SeD states were used to detect the differently expressed HDAC by RT-qPCR, western blotting and IHC staining. Meanwhile, the biological roles of the above HDAC in cartilage development and homeostasis maintenance were confirmed by siRNA transfection, western blotting, RNA sequence and inhibitor treatment experiments. RESULTS HDAC2 exhibited lower expression at protein level in both animals and chondrocytes during SeD condition. The results of cell-level experiments indicated that forkhead box O3A (FOXO3A), which was required to maintain metabolic homeostasis of cartilage matrix, was reduced by HDAC2 knockdown. Meanwhile, induced HDAC2 was positively associated with FOXO3A in rat SeD model. Meanwhile, knockdown of HDAC2 and FOXO3A led to an increase of intracellular ROS level, which activated NF-κB pathway. Se supplementary significantly inhibited the activation of NF-κB pathway with IL-1β treatment. CONCLUSION Our results suggested that low expression of HDAC2 under SeD condition increased ROS content by decreasing FOXO3A in chondrocytes, which led to the activation of NF-κB pathway and ECM homeostasis imbalance.
Collapse
Affiliation(s)
- Yitong Zhao
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yuanxu Guo
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Mengyao Sun
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Safdar Hussion
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Ying Zheng
- Department of Digestive Disease and Gastrointestinal Motility Research Room, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xi Wu Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Huang Huang
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Xinyu Huo
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yutong Zhao
- Department of Software Engineering, Xinjiang University School of Software, Urumqi, 830000, Xinjiang, People's Republic of China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yan Han
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Qilan Ning
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Peng Xu
- Department of Joint Surgery, Xi'an Honghui Hospital, Easter Youyi Road No. 555, Xi'an, 710054, Shaanxi, People's Republic of China
| | - Jian Sun
- Key Laboratory of Trace Elements and Endemic Diseases, Xi'an Jiaotong University School of Public Health, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China.
| | - Shemin Lu
- Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University School of Basic Medical Sciences, West Yanta Street No.76, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
29
|
Xu S, Zhang X, Ma Y, Chen Y, Xie H, Yu L, Wang J, Xu S, Pan F. FOXO3a Alleviates the Inflammation and Oxidative Stress via Regulating TGF-β and HO-1 in Ankylosing Spondylitis. Front Immunol 2022; 13:935534. [PMID: 35784335 PMCID: PMC9247177 DOI: 10.3389/fimmu.2022.935534] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
This study aimed to investigate whether Forkhead box O3a (FOXO3a) modulates inflammation and oxidative stress in ankylosing spondylitis (AS). We applied bioinformatics analysis, quantitative real-time polymerase chain reaction, immunoblotting, enzyme linked immunosorbent assay, chromatin immunoprecipitation, and dual-luciferase reporter assay. Gene overexpression and knockdown of FOXO3a were conducted via lentivirus and small interfering RNA, respectively. Downregulated FOXO3a expression was first confirmed in AS patients. Interleukin-8 (IL-8) and IL-17A were highly expressed and negatively related with FOXO3a in AS. Total antioxidant capacity (T-AOC) were markedly decreased and positively associated with FOXO3a in AS. Overexpression of FOXO3a inhibited the secretion of inflammatory cytokines and promoted the production of antioxidant enzymes in Jurkat cells. Transforming growth factor-β (TGF-β) and heme oxygenase 1 (HO-1), which had binding sites to FOXO3a based on bioinformatics analysis, were abnormally expressed and positively related with FOXO3a. Accordingly, FOXO3a obviously elevated the protein and transcription levels of TGF-β and HO-1 in Jurkat cells. The above results were verified by silencing FOXO3a. Moreover, FOXO3a directly interacted with and promoted the transcription of TGF-β and HO-1. In summary, the modulation of cellular inflammation and oxidative stress via FOXO3a-mediated TGF-β and HO-1 activation is partly involved in the pathogenesis of AS.
Collapse
Affiliation(s)
- Shanshan Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Xiaoyi Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Yubo Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Yuting Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Huimin Xie
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Lingxiang Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Jinian Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Department of Hospital Management Research, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Sheng–qian Xu
- Department of Rheumatism and Immunity, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Faming Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
- *Correspondence: Faming Pan,
| |
Collapse
|
30
|
Castaneda M, den Hollander P, Mani SA. Forkhead Box Transcription Factors: Double-Edged Swords in Cancer. Cancer Res 2022; 82:2057-2065. [PMID: 35315926 PMCID: PMC9258984 DOI: 10.1158/0008-5472.can-21-3371] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 02/12/2022] [Accepted: 03/14/2022] [Indexed: 01/07/2023]
Abstract
A plethora of treatment options exist for cancer therapeutics, but many are limited by side effects and either intrinsic or acquired resistance. The need for more effective targeted cancer treatment has led to the focus on forkhead box (FOX) transcription factors as possible drug targets. Forkhead factors such as FOXA1 and FOXM1 are involved in hormone regulation, immune system modulation, and disease progression through their regulation of the epithelial-mesenchymal transition. Forkhead factors can influence cancer development, progression, metastasis, and drug resistance. In this review, we discuss the various roles of forkhead factors in biological processes that support cancer as well as their function as pioneering factors and their potential as targetable transcription factors in the fight against cancer.
Collapse
Affiliation(s)
- Maria Castaneda
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Petra den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sendurai A. Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Corresponding Author: Sendurai A. Mani, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 2130 West Holcombe Boulevard, Suite 910, Houston, TX 77030-3304. Phone: 713-792-9638; E-mail:
| |
Collapse
|
31
|
Lu Q, Yu S, Meng X, Shi M, Huang S, Li J, Zhang J, Liang Y, Ji M, Zhao Y, Fan H. MicroRNAs: Important Regulatory Molecules in Acute Lung Injury/Acute Respiratory Distress Syndrome. Int J Mol Sci 2022; 23:5545. [PMID: 35628354 PMCID: PMC9142048 DOI: 10.3390/ijms23105545] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/07/2022] [Accepted: 05/13/2022] [Indexed: 02/06/2023] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an overactivated inflammatory response caused by direct or indirect injuries that destroy lung parenchymal cells and dramatically reduce lung function. Although some research progress has been made in recent years, the pathogenesis of ALI/ARDS remains unclear due to its heterogeneity and etiology. MicroRNAs (miRNAs), a type of small noncoding RNA, play a vital role in various diseases. In ALI/ARDS, miRNAs can regulate inflammatory and immune responses by targeting specific molecules. Regulation of miRNA expression can reduce damage and promote the recovery of ALI/ARDS. Consequently, miRNAs are considered as potential diagnostic indicators and therapeutic targets of ALI/ARDS. Given that inflammation plays an important role in the pathogenesis of ALI/ARDS, we review the miRNAs involved in the inflammatory process of ALI/ARDS to provide new ideas for the pathogenesis, clinical diagnosis, and treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Sifan Yu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Xiangyan Meng
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Mingyu Shi
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Siyu Huang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Junfeng Li
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Jianfeng Zhang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yangfan Liang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Mengjun Ji
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (S.Y.); (X.M.); (M.S.); (S.H.); (J.L.); (J.Z.); (Y.L.); (M.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| |
Collapse
|
32
|
Iftikhar R, Penrose HM, King AN, Kim Y, Ruiz E, Kandil E, Machado HL, Savkovic SD. FOXO3 Expression in Macrophages Is Lowered by a High-Fat Diet and Regulates Colonic Inflammation and Tumorigenesis. Metabolites 2022; 12:250. [PMID: 35323693 PMCID: PMC8949544 DOI: 10.3390/metabo12030250] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity, characterized by augmented inflammation and tumorigenesis, is linked to genetic predispositions, such as FOXO3 polymorphisms. As obesity is associated with aberrant macrophages infiltrating different tissues, including the colon, we aimed to identify FOXO3-dependent transcriptomic changes in macrophages that drive obesity-mediated colonic inflammation and tumorigenesis. We found that in mouse colon, high-fat-diet-(HFD)-related obesity led to diminished FOXO3 levels and increased macrophages. Transcriptomic analysis of mouse peritoneal FOXO3-deficient macrophages showed significant differentially expressed genes (DEGs; FDR < 0.05) similar to HFD obese colons. These DEG-related pathways, linked to mouse colonic inflammation and tumorigenesis, were similar to those in inflammatory bowel disease (IBD) and human colon cancer. Additionally, we identified a specific transcriptional signature for the macrophage-FOXO3 axis (MAC-FOXO382), which separated the transcriptome of affected tissue from control in both IBD (p = 5.2 × 10−8 and colon cancer (p = 1.9 × 10−11), revealing its significance in human colonic pathobiologies. Further, we identified (heatmap) and validated (qPCR) DEGs specific to FOXO3-deficient macrophages with established roles both in IBD and colon cancer (IL-1B, CXCR2, S100A8, S100A9, and TREM1) and those with unexamined roles in these colonic pathobiologies (STRA6, SERPINH1, LAMB1, NFE2L3, OLR1, DNAJC28 and VSIG10). These findings establish an important understanding of how HFD obesity and related metabolites promote colonic pathobiologies.
Collapse
Affiliation(s)
- Rida Iftikhar
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, LA 70012, USA; (R.I.); (H.M.P.); (A.N.K.); (Y.K.)
| | - Harrison M. Penrose
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, LA 70012, USA; (R.I.); (H.M.P.); (A.N.K.); (Y.K.)
| | - Angelle N. King
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, LA 70012, USA; (R.I.); (H.M.P.); (A.N.K.); (Y.K.)
| | - Yunah Kim
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, LA 70012, USA; (R.I.); (H.M.P.); (A.N.K.); (Y.K.)
| | - Emmanuelle Ruiz
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Tulane University, New Orleans, LA 70012, USA; (E.R.); (E.K.)
| | - Emad Kandil
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Tulane University, New Orleans, LA 70012, USA; (E.R.); (E.K.)
| | - Heather L. Machado
- Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, LA 70012, USA;
| | - Suzana D. Savkovic
- Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, New Orleans, LA 70012, USA; (R.I.); (H.M.P.); (A.N.K.); (Y.K.)
| |
Collapse
|
33
|
Majumder S, Shivaji UN, Kasturi R, Sigamani A, Ghosh S, Iacucci M. Inflammatory bowel disease-related colorectal cancer: Past, present and future perspectives. World J Gastrointest Oncol 2022; 14:547-567. [PMID: 35321275 PMCID: PMC8919014 DOI: 10.4251/wjgo.v14.i3.547] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/21/2021] [Accepted: 02/27/2022] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease-related colorectal cancer (IBD-CRC) is one of the most serious complications of IBD contributing to significant mortality in this cohort of patients. IBD is often associated with diet and lifestyle-related gut microbial dysbiosis, the interaction of genetic and environmental factors, leading to chronic gut inflammation. According to the “common ground hypothesis”, microbial dysbiosis and intestinal barrier impairment are at the core of the chronic inflammatory process associated with IBD-CRC. Among the many underlying factors known to increase the risk of IBD-CRC, perhaps the most important factor is chronic persistent inflammation. The persistent inflammation in the colon results in increased proliferation of cells necessary for repair but this also increases the risk of dysplastic changes due to chromosomal and microsatellite instability. Multiple pathways have been identified, regulated by many positive and negative factors involved in the development of cancer, which in this case follows the ‘inflammation-dysplasia-carcinoma’ sequence. Strategies to lower this risk are extremely important to reduce morbidity and mortality due to IBD-CRC, among which colonoscopic surveillance is the most widely accepted and implemented modality, forming part of many national and international guidelines. However, the effectiveness of surveillance in IBD has been a topic of much debate in recent years for multiple reasons — cost-benefit to health systems, resource requirements, and also because of studies showing conflicting long-term data. Our review provides a comprehensive overview of past, present, and future perspectives of IBD-CRC. We explore and analyse evidence from studies over decades and current best practices followed globally. In the future directions section, we cover emerging novel endoscopic techniques and artificial intelligence that could play an important role in managing the risk of IBD-CRC.
Collapse
Affiliation(s)
- Snehali Majumder
- Department of Clinical Research, Narayana Health, Bangalore 560099, Karnataka, India
| | - Uday Nagesh Shivaji
- National Institute for Health Research Birmingham Biomedical Research Centre, University Hospitals Birmingham, Birmingham B15 2TH, United Kingdom
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TH, United Kingdom
| | - Rangarajan Kasturi
- Department of Gastroenterology, Narayana Health, Bangalore 560099, India
| | - Alben Sigamani
- Department of Clinical Research, Narayana Health, Bangalore 560099, Karnataka, India
| | - Subrata Ghosh
- National Institute for Health Research Birmingham Biomedical Research Centre, University Hospitals Birmingham, Birmingham B15 2TH, United Kingdom
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TH, United Kingdom
| | - Marietta Iacucci
- National Institute for Health Research Birmingham Biomedical Research Centre, University Hospitals Birmingham, Birmingham B15 2TH, United Kingdom
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TH, United Kingdom
| |
Collapse
|
34
|
Foxo3a tempers excessive glutaminolysis in activated T cells to prevent fatal gut inflammation in the murine IL-10 -/- model of colitis. Cell Death Differ 2022; 29:585-599. [PMID: 34588632 PMCID: PMC8901686 DOI: 10.1038/s41418-021-00876-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023] Open
Abstract
Mutations in susceptibility alleles correlate with gut-inflammatory diseases, such as Crohn's disease; however, this does not often impact the disease progression indicating the existence of compensatory genes. We show that a reduction in Foxo3a expression in IL-10-deficient mice results in a spontaneous and aggressive Crohn's- like disease with 100% penetrance, which is rescued by deletion of myeloid cells, T cells and inhibition of mTORC1. In Foxo3a-/- IL-10-/- mice, there is poor cell death of myeloid cells in the gut, leading to increased accumulation of myeloid and T cells in the gut. Myeloid cells express high levels of inflammatory cytokines, and regulatory T cells are dysfunctional despite increased abundance. Foxo3a signaling represses the transcription of glutaminase (GLS/GLS2) to prevent over-consumption of glutamine by activated T cells and its conversion to glutamate that contributes to the TCA cycle and mTORC1 activation. Finally, we show that Foxo3a restricts the abundance of colitogenic microbiota in IL-10-deficient mice. Thus, by suppressing glutaminolysis in activated T cells Foxo3a mediates a critical checkpoint that prevents the development of fulminant gut inflammatory disease.
Collapse
|
35
|
Lv W, Wu Z, Lin Y, Jiang Y, Chen X, Zhu P, Wang S. Effect of circRNA_FOXO3 rs12196996 polymorphism and FOXO3 rs2232365 polymorphism on survival rate and severity of intensive care unit-acquired sepsis. Bioengineered 2022; 13:4821-4831. [PMID: 35156517 PMCID: PMC8974086 DOI: 10.1080/21655979.2022.2034567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The expression of circRNA_FOXO3 was found to be positively associated with the expression of Forkhead Box O3 (FOXO3), which is targeted and regulated by miR-23a. Polymorphisms in rs12196996 and rs2232365 have been reported in various diseases. In this study, we recruited intensive care unit (ICU)-acquired sepsis patients and grouped them according to their genotypes of rs12196996 and rs2232365. Quantitative real-time PCR was performed to analyze the expression of circRNA_FOXO3, FOXO3 mRNA, and miR-23a. ELISA was carried out to evaluate the abundance of cytokines and luciferase assay was used to explore the inhibitory role of miR-23a on circRNA_FOXO3 and FOXO3. Accordingly, we found that rs12196996 GG and rs2232365 AA were significantly correlated with prolonged survival of ICU-acquired sepsis patients. Rs12196996 GG and rs2232365 AA were also correlated with increased level of miR-23a, IL-10 and decreased level of TNF, IL-2, IFN, IL-6 and IL-1β in the peripheral blood cell samples of patients with ICU-acquired sepsis. The luciferase activity of wild-type (WT) circRNA_FOXO3 and FOXO3 were severely reduced by miR-23a. MiR-23a precursors could effectively suppress the expression of circRNA_FOXO3 and FOXO3 in the cells. Moreover, LPS-induced cell viability loss and dysregulation of cytokines were effectively restored by the knockdown of FOXO3 or circRNA_FOXO3 siRNA in the cells. This study revealed that the minor allele of rs12196996 polymorphism and rs2232365 polymorphism collaboratively contributed to the increased survival and suppressed severity of ICU-acquired sepsis.
Collapse
Affiliation(s)
- Wang Lv
- Department of Emergency, Wenzhou People’s Hospital, Wenzhou, Zhejiang, China
| | - Zhang Wu
- Department of Emergency, Wenzhou People’s Hospital, Wenzhou, Zhejiang, China
| | - Yue Lin
- Department of Emergency, Wenzhou People’s Hospital, Wenzhou, Zhejiang, China
| | - Yingying Jiang
- Department of Emergency, Wenzhou People’s Hospital, Wenzhou, Zhejiang, China
| | - Xinguo Chen
- Department of Emergency, Wenzhou People’s Hospital, Wenzhou, Zhejiang, China
| | - Peng Zhu
- Department of Emergency, Wenzhou People’s Hospital, Wenzhou, Zhejiang, China
| | - Shengnan Wang
- Department of Rheumatology and Immunology, Wenzhou People’s Hospital, Wenzhou, Zhejiang, China
| |
Collapse
|
36
|
Xu S, Zhang X, Ma Y, Xu S, Pan F. The Expression Level of FOXO3a in Patients With Autoimmune Diseases: A Meta-analysis. J Clin Rheumatol 2022; 28:e228-e233. [PMID: 33938500 DOI: 10.1097/rhu.0000000000001675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
METHODS PubMed, Web of Science, and China National Knowledge Infrastructure were used to retrieve relevant articles. The pooled standard mean difference with 95% confidence interval was calculated. RESULTS Totally, 10 studies from 7 publications were included. The levels of FOXO3a were significantly decreased in patients with autoimmune diseases compared with healthy controls (standard mean difference, -1.045; 95% confidence interval, -1.892 to -0.197). When stratified by disease, FOXO3a levels were significantly decreased in rheumatoid arthritis (RA) and inflammatory bowel disease (IBD), but were significantly increased in systemic lupus erythematosus. FOXO3a levels of specific tissues or cells in patients with autoimmune diseases were significantly decreased, but no significant difference was observed in the subgroup of peripheral blood mononuclear cells. In the subgroup analysis combining disease and sample, significant differences of FOXO3a were observed in non-PMBCs of RA and IBD patients. CONCLUSIONS Our study indicated that FOXO3a were significantly decreased in patients with autoimmune diseases. FOXO3a levels was a potential therapeutic target of autoimmune diseases.
Collapse
Affiliation(s)
| | - Xiaoyi Zhang
- Department of Health Toxicology, School of Public Health, Anhui Medical University
| | | | - Shengqian Xu
- Department of Rheumatism and Immunity, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | | |
Collapse
|
37
|
Cameron B, Zaheer SA, Dominguez-Villar M. Control of CD4+ T Cell Differentiation and Function by PI3K Isoforms. Curr Top Microbiol Immunol 2022; 436:197-216. [DOI: 10.1007/978-3-031-06566-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
38
|
Jalgaonkar MP, Parmar UM, Kulkarni YA, Oza MJ. SIRT1-FOXOs activity regulates diabetic complications. Pharmacol Res 2021; 175:106014. [PMID: 34856334 DOI: 10.1016/j.phrs.2021.106014] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 02/07/2023]
Abstract
The prevalence of diabetes is continuously increasing in the recent decades. Persistent hyperglycemia, hyperinsulinemia and the subsequent oxidative stress result in diabetic complications, primarily categorized as microvascular (nephropathy, retinopathy and neuropathy) and macrovascular (cardiomyopathy) complications. The complications are prevalent in both type 1 and type 2 diabetic patients. Polyol pathway, elevated AGE production, PKC activation and hexosamine pathway are indeed the critical pathways involved in the progression of diabetic complications. Silent information regulator 2 or SIR2 or more commonly known as sirtuins are NAD+ dependent histone deacetylase. SIRT1, a member of the sirtuin family has been extensively studied for its role in lifespan extension and needs to be explored for its beneficial effects in diabetic complications. Moreover, it is also known to regulate the activity of other proteins and transcription factors. One such substrate of SIRT1 is FOXOs transcription factor which has gained much attention as the mediator of various cellular processes such as cell cycle arrest and proliferation, DNA repair and metabolism. It has been reported that SIRT1 regulates the activity of FOXOs, whereas few recent advances also suggest a role FOXOs in governing the activity of SIRT1, which permits for a crosstalk between SIRT1 and FOXOs. Therefore, the focus of the present review is to describe and explore the interaction between SIRT1 and FOXOs, predominantly FOXO1 and FOXO3 and to understand the underlying mechanism of SIRT1-FOXOs in controlling and alleviating diabetic complications. Thus, this crosstalk suggests that SIRT1 and FOXOs may serve as potential therapeutic targets in treating diabetic complications.
Collapse
Affiliation(s)
- Manjiri P Jalgaonkar
- SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai 400056, India
| | - Urvi M Parmar
- SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai 400056, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai 400056, India
| | - Manisha J Oza
- SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai 400056, India.
| |
Collapse
|
39
|
Han RH, Huang HM, Han H, Chen H, Zeng F, Xie X, Liu DY, Cai Y, Zhang LQ, Liu X, Xia ZY, Tang J. Propofol postconditioning ameliorates hypoxia/reoxygenation induced H9c2 cell apoptosis and autophagy via upregulating forkhead transcription factors under hyperglycemia. Mil Med Res 2021; 8:58. [PMID: 34753510 PMCID: PMC8579603 DOI: 10.1186/s40779-021-00353-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 10/26/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Administration of propofol, an intravenous anesthetic with antioxidant property, immediately at the onset of post-ischemic reperfusion (propofol postconditioning, P-PostC) has been shown to confer cardioprotection against ischemia-reperfusion injury, while the underlying mechanism remains incompletely understood. The FoxO transcription factors are reported to play critical roles in activating cardiomyocyte survival signaling throughout the process of cellular injuries induced by oxidative stress and are also involved in hypoxic postconditioning mediated neuroprotection, however, the role of FoxO in postconditioning mediated protection in the heart and in particular in high glucose condition is unknown. METHODS Rat heart-derived H9c2 cells were exposed to high glucose (HG) for 48 h (h), then subjected to hypoxia/reoxygenation (H/R, composed of 8 h of hypoxia followed by 12 h of reoxygenation) in the absence or presence of postconditioning with various concentrations of propofol (P-PostC) at the onset of reoxygenation. After having identified the optical concentration of propofol, H9c2 cells were subjected to H/R and P-PostC in the absence or presence of FoxO1 or FoxO3a gene silencing to explore their roles in P-PostC mediated protection against apoptotic and autophagic cell deaths under hyperglycemia. RESULTS The results showed that HG with or without H/R decreased cell viability, increased lactate dehydrogenase (LDH) leakage and the production of reactive oxygen species (ROS) in H9c2 cells, all of which were significantly reversed by propofol (P-PostC), especially at the concentration of 25 µmol/L (P25) (all P < 0.05, NC vs. HG; HG vs. HG + HR; HG + HR + P12.5 or HG + HR + P25 or HG + HR + P50 vs. HG + HR). Moreover, we found that propofol (P25) decreased H9c2 cells apoptosis and autophagy that were concomitant with increased FoxO1 and FoxO3a expression (all P < 0.05, HG + HR + P25 vs. HG + HR). The protective effects of propofol (P25) against H/R injury were reversed by silencing FoxO1 or FoxO3a (all P < 0.05, HG + HR + P25 vs. HG + HR + P25 + siRNA-1 or HG + HR + P25 + siRNA-5). CONCLUSION It is concluded that propofol postconditioning attenuated H9c2 cardiac cells apoptosis and autophagy induced by H/R injury through upregulating FoxO1 and FoxO3a under hyperglycemia.
Collapse
Affiliation(s)
- Rong-Hui Han
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - He-Meng Huang
- Department of Emergency, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Hong Han
- Department of Anesthesiology, the Eighth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 518000, China
| | - Hao Chen
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - Fei Zeng
- Department of Anesthesiology, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, 510000, China
| | - Xiang Xie
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Dan-Yong Liu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - Yin Cai
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China.,Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, China
| | - Liang-Qing Zhang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - Xin Liu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - Zheng-Yuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China. .,State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Pok Fu Lam, 999077, Hong Kong SAR, China.
| | - Jing Tang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue Xiashan District, Zhanjiang City, 524000, Guandong Province, China.
| |
Collapse
|
40
|
Bernabò N, Di Berardino C, Capacchietti G, Peserico A, Buoncuore G, Tosi U, Crociati M, Monaci M, Barboni B. In Vitro Folliculogenesis in Mammalian Models: A Computational Biology Study. Front Mol Biosci 2021; 8:737912. [PMID: 34859047 PMCID: PMC8630647 DOI: 10.3389/fmolb.2021.737912] [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: 07/07/2021] [Accepted: 10/04/2021] [Indexed: 11/27/2022] Open
Abstract
In vitro folliculogenesis (ivF) has been proposed as an emerging technology to support follicle growth and oocyte development. It holds a great deal of attraction from preserving human fertility to improving animal reproductive biotechnology. Despite the mice model, where live offspring have been achieved,in medium-sized mammals, ivF has not been validated yet. Thus, the employment of a network theory approach has been proposed for interpreting the large amount of ivF information collected to date in different mammalian models in order to identify the controllers of the in vitro system. The WoS-derived data generated a scale-free network, easily navigable including 641 nodes and 2089 links. A limited number of controllers (7.2%) are responsible for network robustness by preserving it against random damage. The network nodes were stratified in a coherent biological manner on three layers: the input was composed of systemic hormones and somatic-oocyte paracrine factors; the intermediate one recognized mainly key signaling molecules such as PI3K, KL, JAK-STAT, SMAD4, and cAMP; and the output layer molecules were related to functional ivF endpoints such as the FSH receptor and steroidogenesis. Notably, the phenotypes of knock-out mice previously developed for hub.BN indirectly corroborate their biological relevance in early folliculogenesis. Finally, taking advantage of the STRING analysis approach, further controllers belonging to the metabolic axis backbone were identified, such as mTOR/FOXO, FOXO3/SIRT1, and VEGF, which have been poorly considered in ivF to date. Overall, this in silico study identifies new metabolic sensor molecules controlling ivF serving as a basis for designing innovative diagnostic and treatment methods to preserve female fertility.
Collapse
Affiliation(s)
- Nicola Bernabò
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
- National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | | | | | - Alessia Peserico
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| | - Giorgia Buoncuore
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| | - Umberto Tosi
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| | - Martina Crociati
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
- Centre for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Maurizio Monaci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
- Centre for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Barbara Barboni
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| |
Collapse
|
41
|
Du S, Zheng H. Role of FoxO transcription factors in aging and age-related metabolic and neurodegenerative diseases. Cell Biosci 2021; 11:188. [PMID: 34727995 PMCID: PMC8561869 DOI: 10.1186/s13578-021-00700-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 10/20/2021] [Indexed: 12/18/2022] Open
Abstract
Aging happens to all of us as we live. Thanks to the improved living standard and discovery of life-saving medicines, our life expectancy has increased substantially across the world in the past century. However, the rise in lifespan leads to unprecedented increases in both the number and the percentage of individuals 65 years and older, accompanied by the increased incidences of age-related diseases such as type 2 diabetes mellitus and Alzheimer's disease. FoxO transcription factors are evolutionarily conserved molecules that play critical roles in diverse biological processes, in particular aging and metabolism. Their dysfunction is often found in the pathogenesis of many age-related diseases. Here, we summarize the signaling pathways and cellular functions of FoxO proteins. We also review the complex role of FoxO in aging and age-related diseases, with focus on type 2 diabetes and Alzheimer's disease and discuss the possibility of FoxO as a molecular link between aging and disease risks.
Collapse
Affiliation(s)
- Shuqi Du
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
42
|
Li L, Shi X, Shi Y, Wang Z. The Signaling Pathways Involved in Ovarian Follicle Development. Front Physiol 2021; 12:730196. [PMID: 34646156 PMCID: PMC8504451 DOI: 10.3389/fphys.2021.730196] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/31/2021] [Indexed: 01/13/2023] Open
Abstract
The follicle is the functional unit of the ovary, which is composed of three types of cells: oocytes, granulosa cells, and theca cells. Ovarian follicle development and the subsequent ovulation process are coordinated by highly complex interplay between endocrine, paracrine, and autocrine signals, which coordinate steroidogenesis and gametogenesis. Follicle development is regulated mainly by three organs, the hypothalamus, anterior pituitary, and gonad, which make up the hypothalamic-pituitary-gonadal axis. Steroid hormones and their receptors play pivotal roles in follicle development and participate in a series of classical signaling pathways. In this review, we summarize and compare the role of classical signaling pathways, such as the WNT, insulin, Notch, and Hedgehog pathways, in ovarian follicle development and the underlying regulatory mechanism. We have also found that these four signaling pathways all interact with FOXO3, a transcription factor that is widely known to be under control of the PI3K/AKT signaling pathway and has been implicated as a major signaling pathway in the regulation of dormancy and initial follicular activation in the ovary. Although some of these interactions with FOXO3 have not been verified in ovarian follicle cells, there is a high possibility that FOXO3 plays a core role in follicular development and is regulated by classical signaling pathways. In this review, we present these signaling pathways from a comprehensive perspective to obtain a better understanding of the follicular development process.
Collapse
Affiliation(s)
- Liyuan Li
- Protein Science Key Laboratory of the Ministry of Education, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xiaojin Shi
- Protein Science Key Laboratory of the Ministry of Education, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Yun Shi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhao Wang
- Protein Science Key Laboratory of the Ministry of Education, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
43
|
Kim DH, Bang E, Ha S, Jung HJ, Choi YJ, Yu BP, Chung HY. Organ-differential Roles of Akt/FoxOs Axis as a Key Metabolic Modulator during Aging. Aging Dis 2021; 12:1713-1728. [PMID: 34631216 PMCID: PMC8460295 DOI: 10.14336/ad.2021.0225] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
FoxOs and their post-translational modification by phosphorylation, acetylation, and methylation can affect epigenetic modifications and promote the expression of downstream target genes. Therefore, they ultimately affect cellular and biological functions during aging or occurrence of age-related diseases including cancer, diabetes, and kidney diseases. As known for its key role in aging, FoxOs play various biological roles in the aging process by regulating reactive oxygen species, lipid accumulation, and inflammation. FoxOs regulated by PI3K/Akt pathway modulate the expression of various target genes encoding MnSOD, catalases, PPARγ, and IL-1β during aging, which are associated with age-related diseases. This review highlights the age-dependent differential regulatory mechanism of Akt/FoxOs axis in metabolic and non-metabolic organs. We demonstrated that age-dependent suppression of Akt increases the activity of FoxOs (Akt/FoxOs axis upregulation) in metabolic organs such as liver and muscle. This Akt/FoxOs axis could be modulated and reversed by antiaging paradigm calorie restriction (CR). In contrast, hyperinsulinemia-mediated PI3K/Akt activation inhibited FoxOs activity (Akt/FoxOs axis downregulation) leading to decrease of antioxidant genes expression in non-metabolic organs such as kidneys and lungs during aging. These phenomena are reversed by CR. The results of studies on the process of aging and CR indicate that the Akt/FoxOs axis plays a critical role in regulating metabolic homeostasis, redox stress, and inflammation in various organs during aging process. The benefical actions of CR on the Akt/FoxOs axis in metabolic and non-metabolic organs provide further insights into the molecular mechanisms of organ-differential roles of Akt/FoxOs axis during aging.
Collapse
Affiliation(s)
- Dae Hyun Kim
- 1Department of Pharmacy, College of Pharmacy, Pusan National University, Gumjung-gu, Busan 46241, Korea
| | - EunJin Bang
- 1Department of Pharmacy, College of Pharmacy, Pusan National University, Gumjung-gu, Busan 46241, Korea
| | - Sugyeong Ha
- 1Department of Pharmacy, College of Pharmacy, Pusan National University, Gumjung-gu, Busan 46241, Korea
| | - Hee Jin Jung
- 1Department of Pharmacy, College of Pharmacy, Pusan National University, Gumjung-gu, Busan 46241, Korea
| | - Yeon Ja Choi
- 2Department of Biopharmaceutical Engineering, Division of Chemistry and Biotechnology, Dongguk University, Gyeongju 38066, Korea
| | - Byung Pal Yu
- 3Department of Physiology, The University of Texas Health Science Center at San Antonio, TX 78229, USA
| | - Hae Young Chung
- 1Department of Pharmacy, College of Pharmacy, Pusan National University, Gumjung-gu, Busan 46241, Korea
| |
Collapse
|
44
|
Hussain S, Yadav SS, Banerjee M, Usman K, Khattri S. Evaluation of the Effect of FOXO3 rs13217795 Genotype and Minor Allele (C) on Clinical Chemistry and Genetic Risk of Diabetes Among the Elderly Individuals from Northern India. Mol Syndromol 2021; 13:99-107. [DOI: 10.1159/000518636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/21/2021] [Indexed: 11/19/2022] Open
Abstract
The forkhead box O family (FOXO) is expressed ubiquitously in a spatio-temporal manner and plays a key role in cellular metabolism, senescence, and aging. Genetic mutations in FOXO lead to metabolic diseases and cancer,and affect the longevity of individuals. Our study investigated how the genetic risk of type 2 diabetes mellitus (T2DM) altered due to an intronic variant rs13217795 of the longevity-associated <i>FOXO3</i> gene in the geriatric population of North India. Genotypic characteristics of rs13217795 were determined among 347 age sex-matched (177 diabetic cases, 170 healthy controls) elderly individuals by TaqMan SNP assays after clinical assessment. Clinical chemistry and circulating cytokines level were assessed by biochemical and immunoassays. Genotype frequencies were not significantly (<i>p</i> = 0.526) different between cases and controls. The minor allele (C) frequency in diabetic cases and controls was 0.47 and 0.49, respectively (OR = 0.94, 95% CI = 0.69–1.26, <i>p</i> > 0.05). The minor allele was associated with lower fasting plasma glucose (FPG), fasting insulin, HOMA-IR, CRP, TNF-α, and IL-6 (<i>p</i> < 0.05). The homozygous minor allele carriers showed significantly lower levels of FPG, HOMA-IR, and TNF-α in T2DM patients. The minor allele (C) of intronic polymorphism in <i>FOXO3</i> (rs13217795: T/C) confers the protective role characterized by its association with a decrease in glycemic and insulin resistance and proinflammatory markers.
Collapse
|
45
|
Role of Nrf2 in Synaptic Plasticity and Memory in Alzheimer's Disease. Cells 2021; 10:cells10081884. [PMID: 34440653 PMCID: PMC8391447 DOI: 10.3390/cells10081884] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important transcription factor that reduces oxidative stress. When reactive oxygen species (ROS) or reactive nitrogen species (RNS) are detected, Nrf2 translocates from the cytoplasm into the nucleus and binds to the antioxidant response element (ARE), which regulates the expression of antioxidant and anti-inflammatory genes. Nrf2 impairments are observed in the majority of neurodegenerative disorders, including Alzheimer’s disease (AD). The classic hallmarks of AD include β-amyloid (Aβ) plaques, and neurofibrillary tangles (NFTs). Oxidative stress is observed early in AD and is a novel therapeutic target for the treatment of AD. The nuclear translocation of Nrf2 is impaired in AD compared to controls. Increased oxidative stress is associated with impaired memory and synaptic plasticity. The administration of Nrf2 activators reverses memory and synaptic plasticity impairments in rodent models of AD. Therefore, Nrf2 activators are a potential novel therapeutic for neurodegenerative disorders including AD.
Collapse
|
46
|
Xu S, Pan Z, Huang L, Chen Y, Xie H, Wang F, Zhou T, Yu L, Kong J, Xu S, Pan F. Association of FOXO3a gene polymorphisms and ankylosing spondylitis susceptibility in Eastern Chinese Han population. Gene 2021; 800:145832. [PMID: 34274476 DOI: 10.1016/j.gene.2021.145832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/26/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate the association of FOXO3a polymorphisms and ankylosing spondylitis (AS) susceptibility in Eastern Chinese Han population. METHODS FOXO3a polymorphisms rs12206094, rs12212067, rs2253310, rs3800232, and rs4946933 were genotyped in 650 AS patients and 646 controls by the improved Multiple Ligase Detection Reaction. RESULTS The distribution of genotype in rs12212067 polymorphism was significantly different between AS patients and controls (P = 0.020), especially in male population (P = 0.009). There was significant difference of the genotype frequency distribution at rs3800232 between patients and controls in male population. The results of binary regression analysis showed that the rs12212067 GG genotype and rs3800232 TT genotype were obviously correlated with elevated AS risk, and the associations were still significant after being adjusted by age and gender (all P < 0.05). Interestingly, rs12212067 and rs3800232 genotypes were associated with disease activity of patients. Additionally, haplotype block rs12212067G- rs3800232T (OR = 1.403, 95%CI = 1.011-1.949) was further shown to confer promoting effect on developing AS. CONCLUSION Among Eastern Chinese Han population, FOXO3a polymorphism rs12212067 and rs3800232 may contribute to increased risk of developing AS, but well-designed multicenter studies are needed to further confirm these preliminary findings in the future.
Collapse
Affiliation(s)
- Shanshan Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Zhipeng Pan
- Department of Medical Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Li Huang
- Anhui Medical College, 387 Wuhu Road, Hefei, Anhui 230032, China
| | - Yuting Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Huimin Xie
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Feier Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Tingting Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Lingxiang Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Jiangpiang Kong
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Shengqian Xu
- Department of Rheumatism and Immunity, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Faming Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| |
Collapse
|
47
|
Klinpudtan N, Allsopp RC, Kabayama M, Godai K, Gondo Y, Masui Y, Akagi Y, Srithumsuk W, Sugimoto K, Akasaka H, Takami Y, Takeya Y, Yamamoto K, Ikebe K, Yasumoto S, Ogawa M, Ishizaki T, Arai Y, Rakugi H, Chen R, Willcox BJ, Willcox DC, Kamide K. The association between longevity associated FOXO3 allele and heart disease in Septuagenarians and Octogenarians: The SONIC study. J Gerontol A Biol Sci Med Sci 2021; 77:1542-1548. [PMID: 34254639 PMCID: PMC9373940 DOI: 10.1093/gerona/glab204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Indexed: 01/22/2023] Open
Abstract
The G allele of FOXO3 gene (single-nucleotide polymorphism; rs2802292) is strongly associated with human longevity. However, knowledge of the effect of FOXO3 in older populations, men or women, with heart disease is limited. This cross-sectional study in Japan included 1836 older adults in the 70- and 80-year-old groups. DNA samples isolated from buffy coat samples of peripheral blood were used to genotype FOXO3 (rs2802292). Self-reports were used to obtain heart disease data according to physician diagnosis. Multiple logistic regression was used to test the association by adjusting for the traditional risk factor of heart disease. The prevalence of heart disease in women FOXO3 G-allele carriers was higher than noncarriers (16.7% vs 11.6%, p = .022). The prevalence of coronary heart disease was lower for FOXO3 G carriers in the 70-year-old group for both sexes (men: 9.3% vs 4.3%, p = .042 and women: 10% vs 9%, p = .079, respectively). The G allele was negatively associated with heart disease after adjusting for diabetes, hypertension, dyslipidemia, and smoking in men (odds ratio [OR] = 0.70, 95% confidence intervals [CIs], 0.49–0.99, p = .046), although the association was weaker after full adjustment. In contrast, women carriers of the FOXO3 G allele showed a positive association with heart disease after total adjustment (OR = 1.49, 95% CI, 1.00–2.21, p = .049). In conclusion, the longevity-associated G allele of FOXO3 was observed to have contrasting associations with heart disease prevalence according to sex in older Japanese. To further confirm this association, a longitudinal study and a large sample size will be required.
Collapse
Affiliation(s)
- Nonglak Klinpudtan
- Department of Health Promotion System Sciences, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Richard C Allsopp
- Institute for Biogenesis Research, Department of Anatomy Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Mai Kabayama
- Department of Health Promotion System Sciences, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kayo Godai
- Department of Health Promotion System Sciences, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yasuyuki Gondo
- Department of Clinical Thanatology and Geriatric Behavioral Science, Graduate School of Human Sciences, Osaka University, Osaka, Japan
| | - Yukie Masui
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Yuya Akagi
- Department of Health Promotion System Sciences, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Werayuth Srithumsuk
- Department of Health Promotion System Sciences, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ken Sugimoto
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroshi Akasaka
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoichi Takami
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yasushi Takeya
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazunori Ikebe
- Department of Prosthodontics, Gerodontology and Oral Rehabilitation, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Saori Yasumoto
- Department of Clinical Thanatology and Geriatric Behavioral Science, Graduate School of Human Sciences, Osaka University, Osaka, Japan
| | - Madoka Ogawa
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Tatsuro Ishizaki
- Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Yasumichi Arai
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Randi Chen
- Kuakini Medical Center, Honolulu, Hawaii, USA
| | - Bradley J Willcox
- Kuakini Medical Center, Honolulu, Hawaii, USA.,Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii Honolulu, HI, USA
| | - D Craig Willcox
- Kuakini Medical Center, Honolulu, Hawaii, USA.,Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii Honolulu, HI, USA.,Okinawa International University, Okinawa, Japan
| | - Kei Kamide
- Department of Health Promotion System Sciences, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
48
|
Yi D, Yu H, Lu K, Ruan C, Ding C, Tong L, Zhao X, Chen D. AMPK Signaling in Energy Control, Cartilage Biology, and Osteoarthritis. Front Cell Dev Biol 2021; 9:696602. [PMID: 34239878 PMCID: PMC8258395 DOI: 10.3389/fcell.2021.696602] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022] Open
Abstract
The adenosine monophosphate (AMP)-activated protein kinase (AMPK) was initially identified as an enzyme acting as an "energy sensor" in maintaining energy homeostasis via serine/threonine phosphorylation when low cellular adenosine triphosphate (ATP) level was sensed. AMPK participates in catabolic and anabolic processes at the molecular and cellular levels and is involved in appetite-regulating circuit in the hypothalamus. AMPK signaling also modulates energy metabolism in organs such as adipose tissue, brain, muscle, and heart, which are highly dependent on energy consumption via adjusting the AMP/ADP:ATP ratio. In clinics, biguanides and thiazolidinediones are prescribed to patients with metabolic disorders through activating AMPK signaling and inhibiting complex I in the mitochondria, leading to a reduction in mitochondrial respiration and elevated ATP production. The role of AMPK in mediating skeletal development and related diseases remains obscure. In this review, in addition to discuss the emerging advances of AMPK studies in energy control, we will also illustrate current discoveries of AMPK in chondrocyte homeostasis, osteoarthritis (OA) development, and the signaling interaction of AMPK with other pathways, such as mTOR (mechanistic target of rapamycin), Wnt, and NF-κB (nuclear factor κB) under OA condition.
Collapse
Affiliation(s)
- Dan Yi
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huan Yu
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ke Lu
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Changshun Ruan
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Liping Tong
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaoli Zhao
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Di Chen
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
49
|
Cheema PS, Nandi D, Nag A. Exploring the therapeutic potential of forkhead box O for outfoxing COVID-19. Open Biol 2021; 11:210069. [PMID: 34102081 PMCID: PMC8187014 DOI: 10.1098/rsob.210069] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/27/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has wreaked unprecedented societal havoc worldwide. The infected individuals may present mild to severe symptoms, with nearly 20% of the confirmed patients impaired with significant complications, including multi-organ failure. Acute respiratory distress imposed by SARS-CoV-2 largely results from an aggravated cytokine storm and deregulated immune response. The forkhead box O (FoxO) transcription factors are reported to play a significant role in maintaining normal cell physiology by regulating survival, apoptosis, oxidative stress, development and maturation of T and B lymphocytes, secretion of inflammatory cytokines, etc. We propose a potent anti-inflammatory approach based on activation of the FoxO as an attractive strategy against the novel coronavirus. This regime will be focused on restoring redox and inflammatory homeostasis along with repair of the damaged tissue, activation of lymphocyte effector and memory cells. Repurposing FoxO activators as a means to alleviate the inflammatory burst following SARS-CoV-2 infection can prove immensely valuable in the ongoing pandemic and provide a reliable groundwork for enriching our repertoire of antiviral modalities for any such complication in the future. Altogether, our review highlights the possible efficacy of FoxO activation as a novel arsenal for clinical management of COVID-19.
Collapse
Affiliation(s)
- Pradeep Singh Cheema
- Department of Biochemistry, University of Delhi, South Campus, Biotech Building, 2nd Floor, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| | - Deeptashree Nandi
- Department of Biochemistry, University of Delhi, South Campus, Biotech Building, 2nd Floor, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi, South Campus, Biotech Building, 2nd Floor, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| |
Collapse
|
50
|
Forkhead Transcription Factors in Health and Disease. Trends Genet 2021; 37:460-475. [DOI: 10.1016/j.tig.2020.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022]
|