1
|
Wang Y, Zhao N, Yang D, Zhao J, Di T, Meng Y, Li L, Chen J, Zhu H, Zhang X, Ma H, Qu B, Ma Y, Li P, Wang Y. Metabolic reprogramming and AMPK activation: Key players in the therapeutic effects of Cooling Blood and Detoxicating Formular on psoriasis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118825. [PMID: 39278294 DOI: 10.1016/j.jep.2024.118825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cooling Blood and Detoxicating Formular (CBDF) based on the theory of cooling blood and dosing detoxification, is a useful traditional Chinese medicine (TCM) medication for psoriasis with blood-heat syndrome. AIM OF THE STUDY Investigate the active constituents and mechanisms of the CBDF for the treatment of psoriasis. MATERIALS AND METHODS UPLC-Q-Orbitrap-HRMS technique was used to analyse the ingredients of CBDF absorbed into plasma and skin tissue. The therapeutic efficacy of CBDF was evaluated in treating an imiquimod (IMQ)-induced mouse model was assessed. Transcriptome analysis and gene enrichment analysis were used to explore the changes in gene expression and pathways following treatment with the CBDF. Validation was performed using western blotting, quantitative RT-PCR, flow cytometry, gene knockout and molecular docking in vitro and in vivo. RESULTS 26 compounds were identified in the plasma of IMQ-induced psoriasis-like mouse with CBDF treatment, and higher levels of cimifugin in the lesion. CBDF improved the pathological changes of psoriasis, with inhibition of TNF-α, IL-23, and IL-17A and upregulation of IL-10. Gene enrichment analysis showed that the therapeutic effect of CBDF was related to AMPK pathway. In psoriasis lesions, the AMPK and fatty acid oxidation were suppressed, and glycolysis was enhanced. The Prkaa2, encoding AMPKα2 was down-regulated in psoriasis patients. CBDF inhibited glycolysis while stimulating fatty acid oxidation by the activating AMPK, thereby exerting an inhibitory effect on inflammation. CBDF inhibited MHCII, CD80, and CD86 on dendritic cells of skin drainage lymph node. In vitro, CBDF inhibited bone marrow-derived DCs secrete IL-23, TNF-α, and lactate, while enhanced fatty acid oxidation and AMPK activity. However, the therapeutic effect was weakened in AMPKα2 deletion. Additionally, psoriasis lesions and dendritic cells activation were significantly aggravated after AMPKα2 knockout. The key ingredients of the CBDF, cimifugin, rutin, astilbin, quercetin, and prim-O-glucosylcimifugin, all exhibit a notable affinity towards AMPKα2 binding. CONCLUSIONS CBDF ameliorates psoriasis symptoms and inhibit dendritic cells maturation by regulating metabolic reprogramming in an AMPK-dependent mechanism.
Collapse
Affiliation(s)
- Yazhuo Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Ning Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Danyang Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Tingting Di
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Yujiao Meng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Lin Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Jia Chen
- Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Haoyue Zhu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Xiawei Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Huike Ma
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Baoquan Qu
- Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Yueyue Ma
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China
| | - Yan Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis, Beijing, 100010, China.
| |
Collapse
|
2
|
Wang G, Liu D, Zhang K, Wang Y, Xu Z. Metrnl/IL-41 Alleviates Lipopolysaccharide-Induced Acute Lung Injury by Suppressing Inflammatory Responses through the AMPK/SIRT1 Signaling Pathway. Int Arch Allergy Immunol 2024:1-12. [PMID: 39581185 DOI: 10.1159/000542112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/10/2024] [Indexed: 11/26/2024] Open
Abstract
INTRODUCTION Interleukin-41 (IL-41), also known as Metrnl, is a multifunctional adipokine recognized for its neurotrophic and anti-inflammatory properties that play a significant role in diseases such as sepsis and chronic obstructive pulmonary disease. Despite its crucial biological functions, the mechanisms by which IL-41 mitigates lipopolysaccharide (LPS)-induced acute lung injury (ALI) are not well understood. This study aimed to elucidate the protective effects of IL-41 against LPS-induced inflammation and apoptosis in a murine model and in vitro using bronchial epithelial cells (Beas-2b). METHODS We administered recombinant IL-41 to mice subjected to LPS-induced ALI and observed changes in lung histopathology, inflammation, and apoptosis. Concurrently, Beas-2b cells were transfected with IL-41 constructs, and the role of the AMPK/SIRT1 pathway was investigated using specific inhibitors and agonists. RESULTS Our results demonstrated that LPS-induced ALI is characterized by increased inflammatory cell chemotaxis in lung lavage fluid, enhanced phosphorylation of NFκB p65, and elevated Bax protein expression, coupled with a decrease in IL-41 protein levels. Treatment with recombinant IL-41 effectively mitigated these pathological changes by upregulating AMPK phosphorylation and SIRT1 expression and inhibiting IκB/NFκB p65 phosphorylation. In cellular assays, overexpression of IL-41 reversed LPS-induced oxidative stress, apoptosis, and the secretion of inflammatory cytokines, whereas suppression of IL-41 or inhibition of AMPK reversed these protective effects. CONCLUSIONS In conclusion, IL-41 exerts significant protective effects against ALI by activating the AMPK/SIRT1 signaling pathway and reducing excessive inflammation and apoptosis. These findings suggest that IL-41 holds promise as a therapeutic target for ALI, potentially allowing for personalized treatments based on serum IL-41 levels.
Collapse
Affiliation(s)
- Guannan Wang
- Blood Purification Center, Ningbo Medical Center Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| | - Danqin Liu
- Department of Neurocritical Care Medicine, Ningbo Medical Center Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| | - Kejing Zhang
- Department of Neurocritical Care Medicine, Ningbo Medical Center Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| | - Yan Wang
- Department of Neurocritical Care Medicine, Ningbo Medical Center Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| | - Zhiwei Xu
- Department of Neurocritical Care Medicine, Ningbo Medical Center Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
3
|
Elmorsy EA, Youssef ME, Abdel-Hamed MR, Amer MM, Elghandour SR, Alkhamiss AS, Mohamed NB, Khodeir MM, Elsisi HA, Alsaeed TS, Kamal MM, Ellethy AT, Elesawy BH, Saber S. Activation of AMPK/SIRT1/FOXO3a signaling by BMS-477118 (saxagliptin) mitigates chronic colitis in rats: uncovering new anti-inflammatory and antifibrotic roles. Front Pharmacol 2024; 15:1456058. [PMID: 39359253 PMCID: PMC11445602 DOI: 10.3389/fphar.2024.1456058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
Ulcerative colitis (UC) is a debilitating chronic disease marked by persistent inflammation and intestinal fibrosis. Despite the availability of various treatments, many patients fail to achieve long-term remission, underscoring a significant unmet therapeutic need. BMS-477118, a reversible inhibitor of dipeptidyl peptidase 4 (DPP4), has demonstrated anti-inflammatory properties in preclinical and clinical studies with minimal adverse effects compared to other antidiabetic agents. However, the potential benefits of BMS-477118 in chronic UC have not yet been explored. In this study, we aimed to investigate the effects of BMS-477118 in rats subjected to chronic dextran sodium sulfate (DSS) administration. Our findings indicate that BMS-477118 activates the interconnected positive feedback loop involving AMPK, SIRT1, and FOXO3a, improving histological appearance in injured rat colons. BMS-477118 also reduced fibrotic changes associated with the chronic nature of the animal model, alleviated macroscopic damage and disease severity, and improved the colon weight-to-length ratio. Additionally, BMS-477118 prevented DSS-induced weight loss and enhanced tight junction proteins. These effects, in conjunction with reduced oxidative stress and its potential anti-inflammatory, antiapoptotic, and autophagy-inducing properties, fostered prolonged survival in rats with chronic UC. To conclude, BMS-477118 has the potential to activate the AMPK/SIRT1/FOXO3a signaling pathway in inflamed colons. These results suggest that the AMPK/SIRT1/FOXO3a pathway could be a new therapeutic target for UC. Further research is mandatory to explore the therapeutic possibilities of this pathway. Additionally, continued studies on the therapeutic potential of BMS-477118 and other DPP4 inhibitors are promising for creating new treatments for various conditions, including UC in diabetic patients.
Collapse
Affiliation(s)
- Elsayed A. Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Mahmoud E. Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Mohamed R. Abdel-Hamed
- Department of Anatomy, College of Medicine, Qassim University, Buraidah, Saudi Arabia
- Department of Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maha M. Amer
- Department of Anatomy, College of Medicine, Qassim University, Buraidah, Saudi Arabia
- Department of Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sahar R. Elghandour
- Department of Anatomy and Histology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Abdullah S. Alkhamiss
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Nahla B. Mohamed
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Mostafa M. Khodeir
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
- Department of Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hossam A. Elsisi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah, Saudi Arabia
- Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Thamir Saad Alsaeed
- Department of Biology and Immunology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Manal M. Kamal
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Physiology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Abousree T. Ellethy
- Department of Oral and Medical Basic Sciences, Biochemistry Division, College of Dentistry, Qassim University, Buraidah, Saudi Arabia
| | - Basem H. Elesawy
- Department of Pathology, College of Medicine, Taif University, Taif, Saudi Arabia
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
4
|
Huang Q, Ren Y, Yuan P, Huang M, Liu G, Shi Y, Jia G, Chen M. Targeting the AMPK/Nrf2 Pathway: A Novel Therapeutic Approach for Acute Lung Injury. J Inflamm Res 2024; 17:4683-4700. [PMID: 39051049 PMCID: PMC11268519 DOI: 10.2147/jir.s467882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
ALI(acute lung injury) is a severe respiratory dysfunction caused by various intrapulmonary and extrapulmonary factors. It is primarily characterized by oxidative stress and affects the integrity of the pulmonary barrier. In severe cases, ALI can progress to ARDS(acute respiratory distress syndrome), a condition that poses a serious threat to the lives of affected patients. To date, the etiological mechanisms underlying ALI remain elusive, and available therapeutic options are quite limited. AMPK(AMP-activated protein kinase), an essential serine/threonine protein kinase, performs a pivotal function in the regulation of cellular energy levels and cellular regulatory mechanisms, including the detection of redox signals and mitigating oxidative stress. Meanwhile, Nrf2(nuclear factor erythroid 2-related factor 2), a critical transcription factor, alleviates inflammation and oxidative responses by interacting with multiple signaling pathways and contributing to the modulation of oxidative enzymes associated with inflammation and programmed cell death. Indeed, AMPK induces the dissociation of Nrf2 from Keap1(kelch-like ECH-associated protein-1) and facilitates its translocation into the nucleus to trigger the transcription of downstream antioxidant genes, ultimately suppressing the expression of inflammatory cells in the lungs. Given their roles, AMPK and Nrf2 hold promise as novel treatment targets for ALI. This study aimed to summarise the current status of research on the AMPK/Nrf2 signaling pathway in ALI, encompassing recently reported natural compounds and drugs that can activate the AMPK/Nrf2 signaling pathway to alleviate lung injury, and provide a theoretical reference for early intervention in lung injury and future research on lung protection.
Collapse
Affiliation(s)
- Qianxia Huang
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi City, Gui Zhou, People’s Republic of China
| | - Yingcong Ren
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi City, Gui Zhou, People’s Republic of China
| | - Ping Yuan
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi City, Gui Zhou, People’s Republic of China
| | - Ma Huang
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi City, Gui Zhou, People’s Republic of China
| | - Guoyue Liu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi City, Gui Zhou, People’s Republic of China
| | - Yuanzhi Shi
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi City, Gui Zhou, People’s Republic of China
| | - Guiyang Jia
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi City, Gui Zhou, People’s Republic of China
| | - Miao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi City, Gui Zhou, People’s Republic of China
| |
Collapse
|
5
|
Zhang M, Niu Z, Huang Q, Han L, Du J, Liang J, Cheng Y, Cao R, Yawalkar N, Zhang Z, Yan K. Identification of an exosomal miRNA-mRNA regulatory network contributing to methotrexate efficacy. Int Immunopharmacol 2024; 135:112280. [PMID: 38776848 DOI: 10.1016/j.intimp.2024.112280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 05/05/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Methotrexate (MTX) is an economic and effective medicine treatment for psoriasis. Extracellular vesicle (EV) miRNA biomarkers related to its efficiency have been identified in various diseases. Whether certain miRNA profiles are associated with psoriasis treatment is unknown. In order to determine specific miRNA biomarkers for MTX effectiveness prediction and the severity of psoriasis, our study looked at the variations in circulating EV miRNA profiles before and after MTX therapy. METHODS Plasma EV isolation and next-generation sequencing were performed to identify differentially expressed EV miRNAs between GRs (n = 14) and NRs (n = 6). Univariate and multiple linear regression analyses were performed to evaluate the correlation between PASI scores and miRNA expression levels. RESULTS 15 miRNAs out of a total profile of 443 miRNAs were substantially different between GRs and NRs at baseline, 4 of them (miR-199a-5p, miR-195-5p, miR-196a-5p, and miR-1246) have the potential to distinguish between GRs and NRs [area under the curve (AUC) ≥ 0.70, all P < 0.05]. KEGG pathway analyses revealed differentially expressed miRNAs to potentially target immune-related pathways. SIRT1 was discovered to be a target of miR-199a-5p and involved in MAPK signaling pathway. MiR-191-5p and miR-21-5p expression levels have been discovered to positively correlate with PASI scores[P < 0.05]. CONCLUSION This pilot investigation found that miR-199a-5p, miR-195-5p, miR-196a-5p, and miR-1246 might be prospective biomarkers to predict the efficacy of MTX, and that miR-191-5p and miR-21-5p were correlated with psoriasis severity. Five of them previously reported to be involved in MAPK signaling pathway, indicating a potential role of MTX in delaying the progression of psoriatic inflammation.
Collapse
Affiliation(s)
- Mengmeng Zhang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhenmin Niu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai and Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Qiong Huang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ling Han
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Juan Du
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun Liang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanwen Cheng
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruoshui Cao
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Nikhil Yawalkar
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Zhenghua Zhang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Kexiang Yan
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Xiao Y, Jing D, Zhou G, Tang Z, Peng C, Kuang Y, Zhu W, Chen X, Liu H, Shen M. Adenosine 5'monophosphate-activated protein kinase activation reduces the risks of psoriasis and its comorbidities: a Mendelian randomization study in the UK Biobank. Rheumatology (Oxford) 2024; 63:1664-1671. [PMID: 37672020 DOI: 10.1093/rheumatology/kead462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/25/2023] [Accepted: 08/08/2023] [Indexed: 09/07/2023] Open
Abstract
OBJECTIVE Whether metformin and its adenosine 5'monophosphate-activated protein kinase (AMPK) activation protect from psoriasis risk is unconcluded. We investigated the effect of AMPK, a pharmacological target of metformin, on the risk of psoriasis and its comorbidities and mortality among participants in the UK Biobank (UKB). METHODS To avoid immortal time biases in pharmacoepidemiologic studies, Mendelian randomization was used to infer the AMPK pathway-dependent effects. The cut-off age for distinguishing early-onset/late-onset psoriasis (EOP/LOP) was set at 60 years, based on the incident psoriasis peak in UKB. A genetic instrument comprising 44 single-nucleotide polymorphisms associated with glycated haemoglobin (HbA1c), serving as a proxy for AMPK genetic risk score (negatively associated with AMPK activation), was employed as previously reported in the literature. Log-binomial models were used to estimate the effect size of AMPK regarding relative risk (RR) and 95% CI. RESULTS A total of 407 159 participants were analysed, including 9126 EOP and 3324 LOP. The AMPK genetic risk score was associated with a 12.4% increase in the risk of LOP in men (RR = 1.124, 95% CI: 1.022-1.236). This association was not significant for EOP or women. AMPK genetic risk score exhibited an elevated risk of ischemic heart disease (RR = 1.217, 95% CI 1.062-1.395) in male psoriasis patients. CONCLUSIONS AMPK activation may protect against LOPs and associated ischemic heart disease in men. A sex-specific, comorbidity-targeted intervention for psoriasis is needed.
Collapse
Affiliation(s)
- Yi Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Danrong Jing
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Guowei Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenwei Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Yehong Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Wu Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
| | - Minxue Shen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Furong Laboratory, Changsha, China
- Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
7
|
Garcia BREV, Makiyama EN, Sampaio GR, Soares-Freitas RAM, Bonvini A, Amaral AG, Bordin S, Fock RA, Rogero MM. Effects of Branched-Chain Amino Acids on the Inflammatory Response Induced by LPS in Caco-2 Cells. Metabolites 2024; 14:76. [PMID: 38276311 PMCID: PMC10821323 DOI: 10.3390/metabo14010076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/09/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024] Open
Abstract
Branched-chain amino acids (BCAA) are essential for maintaining intestinal mucosal integrity. However, only a few studies have explored the role of BCAA in the modulation of intestinal inflammation. In this study, we investigated in vitro effects of BCAA on the inflammatory response induced by lipopolysaccharide (LPS) (1 µg/mL) in Caco-2 cells. Caco-2 cells were assigned to six groups: control without BCAA (CTL0), normal BCAA (CTL; 0.8 mM leucine, 0.8 mM isoleucine, and 0.8 mM valine); leucine (LEU; 2 mM leucine), isoleucine (ISO; 2 mM isoleucine), valine (VAL; 2 mM valine), and high BCAA (LIV; 2 mM leucine, 2 mM isoleucine, and 2 mM valine). BCAA was added to the culture medium 24 h before LPS stimulation. Our results indicated that BCAA supplementation did not impair cell viability. The amino acids leucine and isoleucine attenuated the synthesis of IL-8 and JNK and NF-kB phosphorylation induced by LPS. Furthermore, neither BCAA supplementation nor LPS treatment modulated the activity of glutathione peroxidase or the intracellular reduced glutathione/oxidized glutathione ratio. Therefore, leucine and isoleucine exert anti-inflammatory effects in Caco-2 cells exposed to LPS by modulating JNK and NF-kB phosphorylation and IL-8 production. Further in vivo studies are required to validate these findings and gather valuable information for potential therapeutic or dietary interventions.
Collapse
Affiliation(s)
- Bruna Ruschel Ewald Vega Garcia
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil; (B.R.E.V.G.); (G.R.S.); (R.A.M.S.-F.)
| | - Edson Naoto Makiyama
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (E.N.M.); (R.A.F.)
| | - Geni Rodrigues Sampaio
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil; (B.R.E.V.G.); (G.R.S.); (R.A.M.S.-F.)
| | | | - Andrea Bonvini
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of Sao Paulo, São Paulo 05508-000, Brazil;
| | - Andressa Godoy Amaral
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508-000, Brazil; (A.G.A.); (S.B.)
| | - Silvana Bordin
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508-000, Brazil; (A.G.A.); (S.B.)
| | - Ricardo Ambrósio Fock
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (E.N.M.); (R.A.F.)
| | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil; (B.R.E.V.G.); (G.R.S.); (R.A.M.S.-F.)
- Food Research Center (FoRC), CEPID-FAPESP (Research Innovation and Dissemination Centers São Paulo Research Foundation), São Paulo 05508-000, Brazil
| |
Collapse
|
8
|
Yang H, Deng M, Jia H, Zhang K, Liu Y, Cheng M, Xiao W. A review of structural modification and biological activities of oleanolic acid. Chin J Nat Med 2024; 22:15-30. [PMID: 38278556 DOI: 10.1016/s1875-5364(24)60559-5] [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: 07/17/2023] [Indexed: 01/28/2024]
Abstract
Oleanolic acid (OA), a pentacyclic triterpenoid, exhibits a broad spectrum of biological activities, including antitumor, antiviral, antibacterial, anti-inflammatory, hepatoprotective, hypoglycemic, and hypolipidemic effects. Since its initial isolation and identification, numerous studies have reported on the structural modifications and pharmacological activities of OA and its derivatives. Despite this, there has been a dearth of comprehensive reviews in the past two decades, leading to challenges in subsequent research on OA. Based on the main biological activities of OA, this paper comprehensively summarized the modification strategies and structure-activity relationships (SARs) of OA and its derivatives to provide valuable reference for future investigations into OA.
Collapse
Affiliation(s)
- Huali Yang
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang 222001, China; Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Minghui Deng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hongwei Jia
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Kaicheng Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yang Liu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang 222001, China.
| |
Collapse
|
9
|
Cui Y, Chen J, Zhang Z, Shi H, Sun W, Yi Q. The role of AMPK in macrophage metabolism, function and polarisation. J Transl Med 2023; 21:892. [PMID: 38066566 PMCID: PMC10709986 DOI: 10.1186/s12967-023-04772-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is a ubiquitous sensor of energy and nutritional status in eukaryotic cells. It plays a key role in regulating cellular energy homeostasis and multiple aspects of cell metabolism. During macrophage polarisation, AMPK not only guides the metabolic programming of macrophages, but also counter-regulates the inflammatory function of macrophages and promotes their polarisation toward the anti-inflammatory phenotype. AMPK is located at the intersection of macrophage metabolism and inflammation. The metabolic characteristics of macrophages are closely related to immune-related diseases, infectious diseases, cancer progression and immunotherapy. This review discusses the structure of AMPK and its role in the metabolism, function and polarisation of macrophages. In addition, it summarises the important role of the AMPK pathway and AMPK activators in the development of macrophage-related diseases.
Collapse
Affiliation(s)
- Yinxing Cui
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Junhua Chen
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Zhao Zhang
- Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Houyin Shi
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Weichao Sun
- Department of Bone Joint and Bone Oncology, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong, China.
- The Central Laboratory, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong, China.
| | - Qian Yi
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
10
|
Zhu MZ, Xu HM, Liang YJ, Xu J, Yue NN, Zhang Y, Tian CM, Yao J, Wang LS, Nie YQ, Li DF. Edible exosome-like nanoparticles from portulaca oleracea L mitigate DSS-induced colitis via facilitating double-positive CD4 +CD8 +T cells expansion. J Nanobiotechnology 2023; 21:309. [PMID: 37653406 PMCID: PMC10469825 DOI: 10.1186/s12951-023-02065-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023] Open
Abstract
Plant-derived exosome-like nanoparticles (PDENs) have been paid great attention in the treatment of ulcerative colitis (UC). As a proof of concept, we isolated and identified Portulaca oleracea L-derived exosome-like nanoparticles (PELNs) from edible Portulaca oleracea L, which exhibited desirable nano-size (~ 160 nm) and a negative zeta potential value (-31.4 mV). Oral administration of PELNs effectively suppressed the expressions of pro-inflammatory cytokines (TNF-α, IL-6, IL-12, and IL-1β) and myeloperoxidase (MPO), increased levels of the anti-inflammatory cytokine (IL-10), and alleviated acute colitis in dextran sulfate sodium (DSS)-induced C57 mice and IL-10-/- mice. Notably, PELNs exhibited excellent stability and safety within the gastrointestinal tract and displayed specific targeting to inflamed sites in the colons of mice. Mechanistically, oral administration of PELNs played a crucial role in maintaining the diversity and balance of gut microbiota. Furthermore, PELNs treatment enhanced Lactobacillus reuteri growth and elevated indole derivative levels, which might activate the aryl-hydrocarbon receptor (AhR) in conventional CD4+ T cells. This activation downregulated Zbtb7b expression, leading to the reprogramming of conventional CD4+ T cells into double-positive CD4+CD8+T cells (DP CD4+CD8+ T cells). In conclusion, our findings highlighted the potential of orally administered PELNs as a novel, natural, and colon-targeted agent, offering a promising therapeutic approach for managing UC. Schematic illustration of therapeutic effects of oral Portulaca oleracea L -derived natural exosome-like nanoparticles (PELNs) on UC. PELNs treatment enhanced Lactobacillus reuteri growth and elevated indole derivative levels, which activate the aryl-hydrocarbon receptor (AhR) in conventional CD4+ T cells leading to downregulate the expression of Zbtb7b, reprogram of conventional CD4+ T cells into double-positive CD4+CD8+T cells (DP CD4+CD8+ T cells), and decrease the levels of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Yu-Jie Liang
- School of Rehabilitation Medicine, Jining Medical University, Jining, 272029, Shandong, China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Ning-Ning Yue
- Department of Gastroenterology, Shenzhen People's Hospital, the Second Clinical Medical College, Jinan University, Shenzhen, 518020, Guangdong, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, 516000, Guangdong, China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People's Hospital, the Second Clinical Medical College, the First Affiliated Hospital, Jinan University, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital, the Second Clinical Medical College, the First Affiliated Hospital, Jinan University, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China.
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital, the Second Clinical Medical College, the First Affiliated Hospital, Jinan University, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China.
| | - Yu-Qiang Nie
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital, the Second Clinical Medical College, the First Affiliated Hospital, Jinan University, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, China.
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China.
| |
Collapse
|
11
|
Li DF, Tang Q, Yang MF, Xu HM, Zhu MZ, Zhang Y, Tian CM, Nie YQ, Wang JY, Liang YJ, Wang LS, Yao J. Plant-derived exosomal nanoparticles: potential therapeutic for inflammatory bowel disease. NANOSCALE ADVANCES 2023; 5:3575-3588. [PMID: 37441251 PMCID: PMC10334410 DOI: 10.1039/d3na00093a] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023]
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease and ulcerative colitis, is a chronic autoimmune disorder characterized by inflammation. However, currently available disease-modifying anti-IBD drugs exhibit limited efficacy in IBD therapy. Furthermore, existing therapeutic approaches provide only partial relief from IBD symptoms and are associated with certain side effects. In recent years, a novel category of nanoscale membrane vesicles, known as plant-derived exosome-like nanoparticles (PDENs), has been identified in edible plants. These PDENs are abundant in bioactive lipids, proteins, microRNAs, and other pharmacologically active compounds. Notably, PDENs possess immunomodulatory, antitumor, regenerative, and anti-inflammatory properties, making them particularly promising for the treatment of intestinal diseases. Moreover, PDENs can be engineered as targeted delivery systems for the efficient transport of chemical or nucleic acid drugs to the site of intestinal inflammation. In the present study, we provided an overview of PDENs, including their biogenesis, extraction, purification, and construction strategies, and elucidated their physiological functions and therapeutic effects on IBD. Additionally, we summarized the applications and potential of PDENs in IBD treatment while highlighting the future directions and challenges in the field of emerging nanotherapeutics for IBD therapy.
Collapse
Affiliation(s)
- De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology) Shenzhen 518020 Guangdong China
| | - Qi Tang
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology) Shenzhen 518020 Guangdong China
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People's Hospital Shenzhen 518020 Guangdong China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology Guangzhou 510030 China
| | - Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology Guangzhou 510030 China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention Huizhou 516000 Guangdong China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology) Shenzhen 518020 Guangdong China
| | - Yu-Qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology Guangzhou 510030 China
| | - Jian-Yao Wang
- Department of General Surgery, Shenzhen Children's Hospital Shenzhen 518026 Guangdong China
| | - Yu-Jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital Shenzhen 518020 Guangdong China
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology) Shenzhen 518020 Guangdong China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology) Shenzhen 518020 Guangdong China
| |
Collapse
|
12
|
Alzahrani FA, Khan MI, Kameli N, Alsahafi E, Riza YM. Plant-Derived Extracellular Vesicles and Their Exciting Potential as the Future of Next-Generation Drug Delivery. Biomolecules 2023; 13:biom13050839. [PMID: 37238708 DOI: 10.3390/biom13050839] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Plant cells release tiny membranous vesicles called extracellular vesicles (EVs), which are rich in lipids, proteins, nucleic acids, and pharmacologically active compounds. These plant-derived EVs (PDEVs) are safe and easily extractable and have been shown to have therapeutic effects against inflammation, cancer, bacteria, and aging. They have shown promise in preventing or treating colitis, cancer, alcoholic liver disease, and even COVID-19. PDEVs can also be used as natural carriers for small-molecule drugs and nucleic acids through various administration routes such as oral, transdermal, or injection. The unique advantages of PDEVs make them highly competitive in clinical applications and preventive healthcare products in the future. This review covers the latest methods for isolating and characterizing PDEVs, their applications in disease prevention and treatment, and their potential as a new drug carrier, with special attention to their commercial viability and toxicological profile, as the future of nanomedicine therapeutics. This review champions the formation of a new task force specializing in PDEVs to address a global need for rigor and standardization in PDEV research.
Collapse
Affiliation(s)
- Faisal A Alzahrani
- Department of Biochemistry, Faculty of science, Embryonic Stem Cell Unit, King Fahad Center for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre of Artificial Intelligence for Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammad Imran Khan
- Centre of Artificial Intelligence for Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nader Kameli
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan 82621, Saudi Arabia
- Medical Research Center, Jazan University, Jazan 45142, Saudi Arabia
| | - Elham Alsahafi
- Department of Basic and Clinical Sciences, Faculty of Dentistry, Umm AlQura University, P.O. Box 715, Mecca 21955, Saudi Arabia
| | - Yasir Mohamed Riza
- Department of Biochemistry, Faculty of science, Embryonic Stem Cell Unit, King Fahad Center for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Centre of Artificial Intelligence for Precision Medicines, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
13
|
Mangoni AA, Sotgia S, Zinellu A, Carru C, Pintus G, Damiani G, Erre GL, Tommasi S. Methotrexate and cardiovascular prevention: an appraisal of the current evidence. Ther Adv Cardiovasc Dis 2023; 17:17539447231215213. [PMID: 38115784 PMCID: PMC10732001 DOI: 10.1177/17539447231215213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/02/2023] [Indexed: 12/21/2023] Open
Abstract
New evidence continues to accumulate regarding a significant association between excessive inflammation and dysregulated immunity (local and systemic) and the risk of cardiovascular events in different patient cohorts. Whilst research has sought to identify novel atheroprotective therapies targeting inflammation and immunity, several marketed drugs for rheumatological conditions may serve a similar purpose. One such drug, methotrexate, has been used since 1948 for treating cancer and, more recently, for a wide range of dysimmune conditions. Over the last 30 years, epidemiological and experimental studies have shown that methotrexate is independently associated with a reduced risk of cardiovascular disease, particularly in rheumatological patients, and exerts several beneficial effects on vascular homeostasis and blood pressure control. This review article discusses the current challenges with managing cardiovascular risk and the new frontiers offered by drug discovery and drug repurposing targeting inflammation and immunity with a focus on methotrexate. Specifically, the article critically appraises the results of observational, cross-sectional and intervention studies investigating the effects of methotrexate on overall cardiovascular risk and individual risk factors. It also discusses the putative molecular mechanisms underpinning the atheroprotective effects of methotrexate and the practical advantages of using methotrexate in cardiovascular prevention, and highlights future research directions in this area.
Collapse
Affiliation(s)
- Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, SA 5042, Australia
| | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy; Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Giovanni Damiani
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Italian Centre of Precision Medicine and Chronic Inflammation, Milan, Italy
| | - Gian Luca Erre
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital (AOUSS) and University of Sassari, Sassari, Italy
| | - Sara Tommasi
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, SA, Australia
| |
Collapse
|
14
|
Tian Y, Wang J, Qin X, Li S, Lan C, Sun X. Narciclasine ameliorated T cell mediated acute liver injury through activating AMPK pathway. Cell Immunol 2022; 382:104631. [PMID: 36272268 DOI: 10.1016/j.cellimm.2022.104631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/25/2022] [Accepted: 10/08/2022] [Indexed: 01/13/2023]
Abstract
Hepatitis is closely related to cirrhosis and liver cancer, and it is vital that we develop new drugs and identify new drug targets. Traditional Chinese medicine has demonstrated excellent curative effects on liver diseases. The ingredients from Chinese herbals are important source for drug development in the treatment of hepatitis. Here, we found that narciclasine (NCS), a major component extracted from narcissus bulbs, showed hepatoprotective effect against concanavalin A (Con A) induced hepatitis. NCS treatment significantly reduced hepatocyte death, hepatic inflammatory cells infiltration, and serum cytokine levels in Con A challenged mice. We further observed that NCS directly inhibited Con A induced splenocytes proliferation and cytokine production in vitro. RNA-seq results showed that genes related to immune response were upregulated in Con A treated CD4+ T cells, which were down-regulated in the presence of NCS. Moreover, the AMPK pathway had been found activated in response to NCS treatment, suggesting a potential target for NCS targets. In conclusion, our results reveal that NCS is a powerful immunosuppressor against T cell activation, thus leading to protection against Con A induced liver injury in mice. These findings provide new insights into the use of natural products in the treatment of autoimmune hepatitis.
Collapse
Affiliation(s)
- Yuanyuan Tian
- Department of Gastroenterology, Hainan General Hospital/ Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, China
| | - Jiao Wang
- Department of Infectious Diseases, Hainan General Hospital/ Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, China
| | - Xiaori Qin
- Department of Gastroenterology, Hainan General Hospital/ Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, China
| | - Shibing Li
- Department of Pediatric Surgery, Hainan General Hospital/ Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, China
| | - Cheng Lan
- Department of Gastroenterology, Hainan General Hospital/ Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, China.
| | - Xiaoning Sun
- Clinical College, Hainan Medical University, Haikou 570100, China.
| |
Collapse
|
15
|
Abdul Khaliq H, Alhouayek M, Quetin-Leclercq J, Muccioli GG. 5'AMP-activated protein kinase: an emerging target of phytochemicals to treat chronic inflammatory diseases. Crit Rev Food Sci Nutr 2022; 64:4763-4788. [PMID: 36450301 DOI: 10.1080/10408398.2022.2145264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Inflammation is a defensive response of the organism to traumatic, infectious, toxic, ischemic, and autoimmune injury. Inflammatory mediators are released to effectively eliminate the inflammatory trigger and restore homeostasis. However, failure of these processes can lead to chronic inflammatory conditions and diseases such as inflammatory bowel diseases, rheumatoid arthritis, inflammatory lung diseases, atherosclerosis, and neurodegenerative diseases. The cure of chronic inflammatory diseases remains challenging as current therapies have various limitations, such as pronounced side effects, progressive loss of efficacy, and high cost especially for biologics. In this context, phytochemicals (such as alkaloids, flavonoids, lignans, phenolic acids, saponins, terpenoids, and other classes) are considered as an interesting alternative approach. Among the numerous targets of phytochemicals, AMP-activated protein kinase (AMPK) can be considered as an interesting target in the context of inflammation. AMPK regulates inflammatory response by inhibiting inflammatory pathways (NF-κB, JAK/STAT, and MAPK) and regulating several other processes of the inflammatory response (oxidative stress, autophagy, and apoptosis). In this review, we summarize and discuss the studies focusing on phytochemicals that showed beneficial effects by blocking different inflammatory pathways implicating AMPK activation in chronic inflammatory disease models. We also highlight elements to consider when investigating AMPK in the context of phytochemicals.
Collapse
Affiliation(s)
- Hafiz Abdul Khaliq
- Pharmacognosy Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
- Department of Pharmacognosy, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Mireille Alhouayek
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Joëlle Quetin-Leclercq
- Pharmacognosy Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| |
Collapse
|
16
|
Focusing on Future Applications and Current Challenges of Plant Derived Extracellular Vesicles. Pharmaceuticals (Basel) 2022; 15:ph15060708. [PMID: 35745626 PMCID: PMC9229679 DOI: 10.3390/ph15060708] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 11/22/2022] Open
Abstract
Plant derived extracellular vesicles (EVs) are nano-sized membranous vesicles released by plant cells, which contain lipids, proteins, nucleic acids and specific pharmacologically active substances. They are safe, widely available and expediently extractive. They have gratifyingly biological activity against inflammation, cancer, bacteria and oxidative aging, especially for the prevention or treatment of colitis, cancer, alcoholic liver, and COVID-19. In addition, as natural drug carriers, plant derived EVs have the potential to target the delivery of small molecule drugs and nucleic acid through oral, transdermal, injection. With the above advantages, plant derived EVs are expected to have excellent strong competitiveness in clinical application or preventive health care products in the future. We comprehensively reviewed the latest separation methods and physical characterization techniques of plant derived EVs, summarized the application of them in disease prevention or treatment and as a new drug carrier, and analyzed the clinical application prospect of plant derived EVs as a new drug carrier in the future. Finally, the problems hindering the development of plant derived EVs at present and consideration of the standardized application of them are discussed.
Collapse
|
17
|
Pawar A, Russo M, Rani I, Goswami K, Russo GL, Pal A. A critical evaluation of risk to reward ratio of quercetin supplementation for COVID-19 and associated comorbid conditions. Phytother Res 2022; 36:2394-2415. [PMID: 35393674 PMCID: PMC9111035 DOI: 10.1002/ptr.7461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 03/19/2022] [Accepted: 03/26/2022] [Indexed: 01/08/2023]
Abstract
The interim results of the large, multinational trials on coronavirus disease 2019 (COVID‐19) using a combination of antiviral drugs appear to have little to no effect on the 28‐day mortality or the in‐hospital course. Therefore, there is a still vivid interest in finding alternate re‐purposed drugs and nutrition supplements, which can halt or slow the disease severity. We review here the multiple preclinical studies, partially supported by clinical evidence showing the quercetin's possible therapeutic/prophylaxis efficacy against severe acute respiratory syndrome coronavirus (SARS‐CoV) as well as comorbidities like chronic obstructive pulmonary disease (COPD), diabetes mellitus, obesity, coagulopathy, and hypertension. Currently, 14 interventional clinical trials are underway assessing the efficacy of quercetin along with other antiviral drugs/nutritional supplements as prophylaxis/treatment option against COVID‐19. The present review is tempting to suggest that, based on circumstantial scientific evidence and preliminary clinical data, the flavonoid quercetin can ameliorate COVID‐19 infection and symptoms acting in concert on two parallel and independent paths: inhibiting key factors responsible for SARS‐CoV‐2 infections and mitigating the clinical manifestations of the disease in patients with comorbid conditions. Despite the broad therapeutic properties of quercetin, further high power randomized clinical trials are needed to firmly establish its clinical efficacy against COVID‐19.
Collapse
Affiliation(s)
- Anil Pawar
- Department of Zoology, DAV University, Jalandhar, India
| | - Maria Russo
- National Research Council, Institute of Food Sciences, Avellino, Italy
| | - Isha Rani
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research (MMIMSR), Maharishi Markandeshwar University (MMU), Ambala, India
| | | | - Gian Luigi Russo
- National Research Council, Institute of Food Sciences, Avellino, Italy
| | - Amit Pal
- Department of Biochemistry, AIIMS, Kalyani, India
| |
Collapse
|
18
|
Hypoglycemic Effects of Plant Flavonoids: A Review. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2057333. [PMID: 34925525 PMCID: PMC8674047 DOI: 10.1155/2021/2057333] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022]
Abstract
Diabetes mellitus is a metabolic disorder with chronic high blood glucose levels, and it is associated with defects in insulin secretion, insulin resistance, or both. It is also a major public issue, affecting the world's population. This disease contributes to long-term health complications such as dysfunction and failure of multiple organs, including nerves, heart, blood vessels, kidneys, and eyes. Flavonoids are phenolic compounds found in nature and usually present as secondary metabolites in plants, vegetables, and fungi. Flavonoids possess many health benefits such as anti-inflammatory and antioxidant activities, and naturally occurring flavonoids contribute to antidiabetic effects.Many studies conducted in vivo and in vitro have proven the hypoglycemic effect of plant flavonoids. A large number of studies showed that flavonoids hold positive results in controlling the blood glucose level in streptozotocin (STZ)-induced diabetic rats and further prevent the complications of diabetes. The future development of flavonoid-based drugs is believed to provide significant effects on diabetes mellitus and diabetes complication diseases. This review aims at summarizing the various types of flavonoids that function as hyperglycemia regulators such as inhibitors of α-glucosidase and glucose cotransporters in the body. This review article discusses the hypoglycemic effects of selected plant flavonoids namely quercetin, kaempferol, rutin, naringenin, fisetin, and morin. Four search engines, PubMed, Google Scholar, Scopus, and SciFinder, are used to collect the data.
Collapse
|
19
|
Koutouroushis C, Sarkar O. Role of Autophagy in Cardiovascular Disease and Aging. Cureus 2021; 13:e20042. [PMID: 34873555 PMCID: PMC8631374 DOI: 10.7759/cureus.20042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2021] [Indexed: 11/15/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and is expected to further increase as people continue to live even longer. Although the life span of the general population is increasing, the con of such a prolonged life span is that aging has certain detrimental effects on the molecular, structural, and functional elements of the cardiovascular system. This review will discuss various molecular pathways linked to longevity, most notably autophagy and its associated mechanisms, and how these pathways can be targeted to promote cardiovascular health through the process of aging. It is to be noted that the process of autophagy decreases with aging; hence, this review concludes that the promotion of autophagy, through implementation of caloric restriction, intermittent fasting, and pharmacologic agents, has proven to be an efficacious means of stimulating cardiovascular health. Therefore, autophagy is an important target for prevention and procrastination of cardiovascular pathologies in the geriatric population.
Collapse
Affiliation(s)
| | - Oiendrila Sarkar
- General Internal Medicine, St. Mary's Hospital, Isle of Wight NHS Trust, Newport, GBR
| |
Collapse
|
20
|
Antonioli L, Pellegrini C, Fornai M, Benvenuti L, D’Antongiovanni V, Colucci R, Bertani L, Di Salvo C, Semeghini G, La Motta C, Giusti L, Zallocco L, Ronci M, Quattrini L, Angelucci F, Coviello V, Oh WK, Ha QTK, Németh ZH, Haskó G, Blandizzi C. Preclinical Development of FA5, a Novel AMP-Activated Protein Kinase (AMPK) Activator as an Innovative Drug for the Management of Bowel Inflammation. Int J Mol Sci 2021; 22:6325. [PMID: 34199160 PMCID: PMC8231528 DOI: 10.3390/ijms22126325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
Acadesine (ACA), a pharmacological activator of AMP-activated protein kinase (AMPK), showed a promising beneficial effect in a mouse model of colitis, indicating this drug as an alternative tool to manage IBDs. However, ACA displays some pharmacodynamic limitations precluding its therapeutical applications. Our study was aimed at evaluating the in vitro and in vivo effects of FA-5 (a novel direct AMPK activator synthesized in our laboratories) in an experimental model of colitis in rats. A set of experiments evaluated the ability of FA5 to activate AMPK and to compare the efficacy of FA5 with ACA in an experimental model of colitis. The effects of FA-5, ACA, or dexamethasone were tested in rats with 2,4-dinitrobenzenesulfonic acid (DNBS)-induced colitis to assess systemic and tissue inflammatory parameters. In in vitro experiments, FA5 induced phosphorylation, and thus the activation, of AMPK, contextually to the activation of SIRT-1. In vivo, FA5 counteracted the increase in spleen weight, improved the colon length, ameliorated macroscopic damage score, and reduced TNF and MDA tissue levels in DNBS-treated rats. Of note, FA-5 displayed an increased anti-inflammatory efficacy as compared with ACA. The novel AMPK activator FA-5 displays an improved anti-inflammatory efficacy representing a promising pharmacological tool against bowel inflammation.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Carolina Pellegrini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Vanessa D’Antongiovanni
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy;
| | - Lorenzo Bertani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Clelia Di Salvo
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Giorgia Semeghini
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Concettina La Motta
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Laura Giusti
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | - Lorenzo Zallocco
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Maurizio Ronci
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy;
| | - Luca Quattrini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Francesco Angelucci
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Vito Coviello
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Won-Keun Oh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Korea; (W.-K.O.); (Q.T.K.H.)
| | - Quy Thi Kim Ha
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Korea; (W.-K.O.); (Q.T.K.H.)
| | - Zoltan H. Németh
- Department of Anesthesiology, Columbia University, New York City, NY 10027, USA; (Z.H.N.); (G.H.)
- Department of Surgery, Morristown Medical Center, Morristown, NJ 07960, USA
| | - Gyorgy Haskó
- Department of Anesthesiology, Columbia University, New York City, NY 10027, USA; (Z.H.N.); (G.H.)
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| |
Collapse
|
21
|
Marsilio I, Caputi V, Latorre E, Cerantola S, Paquola A, Alcalde AI, Mesonero JE, O'Mahony SM, Bertazzo A, Giaroni C, Giron MC. Oxidized phospholipids affect small intestine neuromuscular transmission and serotonergic pathways in juvenile mice. Neurogastroenterol Motil 2021; 33:e14036. [PMID: 33222337 DOI: 10.1111/nmo.14036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/14/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Oxidized phospholipid derivatives (OxPAPCs) act as bacterial lipopolysaccharide (LPS)-like damage-associated molecular patterns. OxPAPCs dose-dependently exert pro- or anti-inflammatory effects by interacting with several cellular receptors, mainly Toll-like receptors 2 and 4. It is currently unknown whether OxPAPCs may affect enteric nervous system (ENS) functional and structural integrity. METHODS Juvenile (3 weeks old) male C57Bl/6 mice were treated intraperitoneally with OxPAPCs, twice daily for 3 days. Changes in small intestinal contractility were evaluated by isometric neuromuscular responses to receptor and non-receptor-mediated stimuli. Alterations in ENS integrity and serotonergic pathways were assessed by real-time PCR and confocal immunofluorescence microscopy in longitudinal muscle-myenteric plexus whole-mount preparations (LMMPs). Tissue levels of serotonin (5-HT), tryptophan, and kynurenine were measured by HPLC coupled to UV/fluorescent detection. KEY RESULTS OxPAPC treatment induced enteric gliosis, loss of myenteric plexus neurons, and excitatory hypercontractility, and reduced nitrergic neurotransmission with no changes in nNOS+ neurons. Interestingly, these changes were associated with a higher functional response to 5-HT, altered immunoreactivity of 5-HT receptors and serotonin transporter (SERT) together with a marked decrease in 5-HT levels, shifting tryptophan metabolism toward kynurenine production. CONCLUSIONS AND INFERENCES OxPAPC treatment disrupted structural and functional integrity of the ENS, affecting serotoninergic tone and 5-HT tissue levels toward a higher kynurenine content during adolescence, suggesting that changes in intestinal lipid metabolism toward oxidation can affect serotoninergic pathways, potentially increasing the risk of developing functional gastrointestinal disorders during critical stages of development.
Collapse
Affiliation(s)
- Ilaria Marsilio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Valentina Caputi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.,Department of Anatomy and Neuroscience and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eva Latorre
- Departamento Farmacología y Fisiología, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain.,Instituto Agroalimentario de Aragón - IA2-(Universidad de Zaragoza - CITA), Zaragoza, Spain
| | - Silvia Cerantola
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.,San Camillo Hospital, Treviso, Italy
| | - Andrea Paquola
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Ana I Alcalde
- Departamento Farmacología y Fisiología, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain.,Instituto Agroalimentario de Aragón - IA2-(Universidad de Zaragoza - CITA), Zaragoza, Spain
| | - José E Mesonero
- Departamento Farmacología y Fisiología, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain.,Instituto Agroalimentario de Aragón - IA2-(Universidad de Zaragoza - CITA), Zaragoza, Spain
| | - Siobhain M O'Mahony
- Department of Anatomy and Neuroscience and APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Antonella Bertazzo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
22
|
Pan Y, Liu L, Zhang Q, Shi W, Feng W, Wang J, Wang Q, Li S, Li M. Activation of AMPK suppresses S1P-induced airway smooth muscle cells proliferation and its potential mechanisms. Mol Immunol 2020; 128:106-115. [PMID: 33126079 DOI: 10.1016/j.molimm.2020.09.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 01/13/2023]
Abstract
The aims of the present study were to investigate the signaling mechanisms for sphingosine-1-phosphate (S1P)-induced airway smooth muscle cells (ASMCs) proliferation and to explore the effect of activation of adenosine monophosphate-activated protein kinase (AMPK) on S1P-induced ASMCs proliferation and its underlying mechanisms. S1P phosphorylated signal transducer and activator of transcription 3 (STAT3) through binding to S1PR2/3, and this further sequentially up-regulated polo-like kinase 1 (PLK1) and inhibitor of differentiation 2 (ID2) protein expression. Pretreatment of cells with S1PR2 antagonist JTE-013, S1PR3 antagonist CAY-10444, knockdown of STAT3, PLK1 and ID2 attenuated S1P-triggered ASMCs proliferation. In addition, activation of AMPK by metformin inhibited S1P-induced ASMCs proliferation by suppressing STAT3 phosphorylation and therefore suppression of PLK1 and ID2 protein expression. Our study suggests that S1P promotes ASMCs proliferation by stimulating S1PR2/3/STAT3/PLK1/ID2 axis, and activation of AMPK suppresses ASMCs proliferation by targeting on STAT3 signaling pathway. Activation of AMPK might benefit asthma by inhibiting airway remodeling.
Collapse
Affiliation(s)
- Yilin Pan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Pulmonary and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wei Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
23
|
Synthesis and anti-inflammatory activity of saponin derivatives of δ-oleanolic acid. Eur J Med Chem 2020; 209:112932. [PMID: 33131725 DOI: 10.1016/j.ejmech.2020.112932] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023]
Abstract
Pentacyclic triterpenes (PTs) are the active ingredients of many medicinal herbs and pharmaceutical formulations, and are well-known for their anti-inflammatory activity. On the other hand, anti-inflammatory effects of AMP-activated protein kinase (AMPK) have recently drawn much attention. In this study, we found that a variety of naturally occurring PTs sapogenins and saponins could stimulate the phosphorylation of AMPK, and identified δ-oleanolic acid (10) as a potent AMPK activator. Based on these findings, 23 saponin derivatives of δ-oleanolic acid were synthesized in order to find more potent anti-inflammatory agents with improved pharmacokinetic properties. The results of cellular assays showed that saponin 29 significantly inhibited LPS-induced secretion of pro-inflammatory factors TNF-α and IL-6 in THP1-derived macrophages. Preliminary mechanistic studies showed that 29 stimulated the phosphorylation of AMPK and acetyl-CoA carboxylase (ACC). The bioavailability of 29 was significantly improved in comparison with its aglycon. More importantly, 29 showed significant anti-inflammatory and liver-protective effects in LPS/D-GalN-induced fulminant hepatic failure mice. Taken together, PTs saponins hold promise as therapeutic agents for inflammatory diseases.
Collapse
|
24
|
Salvatore T, Pafundi PC, Galiero R, Gjeloshi K, Masini F, Acierno C, Di Martino A, Albanese G, Alfano M, Rinaldi L, Sasso FC. Metformin: A Potential Therapeutic Tool for Rheumatologists. Pharmaceuticals (Basel) 2020; 13:ph13090234. [PMID: 32899806 PMCID: PMC7560003 DOI: 10.3390/ph13090234] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Metformin is an oral antihyperglycemic drug widely used to treat type 2 diabetes, acting via indirect activation of 5′ Adenosine Monophosphate-activated Protein Kinase (AMPK). Actually, evidence has accumulated of an intriguing anti-inflammatory activity, mainly mediated by AMPK through a variety of mechanisms such as the inhibition of cytokine-stimulated Nuclear Factor-κB (NF-κB) and the downregulation of the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) signaling pathways. Moreover, AMPK plays an important role in the modulation of T lymphocytes and other pivotal cells of the innate immune system. The current understanding of these AMPK effects provides a strong rationale for metformin repurposing in the management of autoimmune and inflammatory conditions. Several studies demonstrated metformin’s beneficial effects on both animal and human rheumatologic diseases, especially on rheumatoid arthritis. Unfortunately, even though data are large and remarkable, they almost exclusively come from experimental investigations with only a few from clinical trials. The lack of support from prospective placebo-controlled trials does not allow metformin to enter the therapeutic repertoire of rheumatologists. However, a large proportion of rheumatologic patients can currently benefit from metformin, such as those with concomitant obesity and type 2 diabetes, two conditions strongly associated with rheumatoid arthritis, osteoarthritis, and gout, as well as those with diabetes secondary to steroid therapy.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via de Crecchio, 7, I-80138 Naples, Italy;
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia, 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (K.G.); (F.M.); (C.A.); (A.D.M.); (G.A.); (M.A.); (L.R.)
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia, 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (K.G.); (F.M.); (C.A.); (A.D.M.); (G.A.); (M.A.); (L.R.)
| | - Klodian Gjeloshi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia, 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (K.G.); (F.M.); (C.A.); (A.D.M.); (G.A.); (M.A.); (L.R.)
| | - Francesco Masini
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia, 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (K.G.); (F.M.); (C.A.); (A.D.M.); (G.A.); (M.A.); (L.R.)
| | - Carlo Acierno
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia, 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (K.G.); (F.M.); (C.A.); (A.D.M.); (G.A.); (M.A.); (L.R.)
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia, 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (K.G.); (F.M.); (C.A.); (A.D.M.); (G.A.); (M.A.); (L.R.)
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia, 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (K.G.); (F.M.); (C.A.); (A.D.M.); (G.A.); (M.A.); (L.R.)
| | - Maria Alfano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia, 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (K.G.); (F.M.); (C.A.); (A.D.M.); (G.A.); (M.A.); (L.R.)
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia, 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (K.G.); (F.M.); (C.A.); (A.D.M.); (G.A.); (M.A.); (L.R.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Piazza L. Miraglia, 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (K.G.); (F.M.); (C.A.); (A.D.M.); (G.A.); (M.A.); (L.R.)
- Correspondence: ; Tel.: +39-081-566-5010
| |
Collapse
|
25
|
Shakya AK, Naik RR, Almasri IM, Kaur A. Role and Function of Adenosine and its Receptors in Inflammation, Neuroinflammation, IBS, Autoimmune Inflammatory Disorders, Rheumatoid Arthritis and Psoriasis. Curr Pharm Des 2020; 25:2875-2891. [PMID: 31333103 DOI: 10.2174/1381612825666190716145206] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
The physiological effects of endogenous adenosine on various organ systems are very complex and numerous which are elicited upon activation of any of the four G-protein-coupled receptors (GPCRs) denoted as A1, A2A, A2B and A3 adenosine receptors (ARs). Several fused heterocyclic and non-xanthine derivatives are reported as a possible target for these receptors due to physiological problems and lack of selectivity of xanthine derivatives. In the present review, we have discussed the development of various new chemical entities as a target for these receptors. In addition, compounds acting on adenosine receptors can be utilized in treating diseases like inflammation, neuroinflammation, autoimmune and related diseases.
Collapse
Affiliation(s)
- Ashok K Shakya
- Medicinal Chemistry, Drug Design and Drug Metabolism, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al- Ahliyya Amman University, PO Box 263, Amman 19328, Jordan
| | - Rajashri R Naik
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Ihab M Almasri
- Medicinal Chemistry and Drug Design, Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Al Azhar University Gaza, Gaza Strip, Palestinian Territory, Occupied
| | - Avneet Kaur
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Mehrauli-Badarpur Road, Pushp Vihar, Sector-3, New Delhi-110017, India
| |
Collapse
|
26
|
Abdelkader NF, Eitah HE, Maklad YA, Gamaleldin AA, Badawi MA, Kenawy SA. New combination therapy of gliclazide and quercetin for protection against STZ-induced diabetic rats. Life Sci 2020; 247:117458. [PMID: 32092333 DOI: 10.1016/j.lfs.2020.117458] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022]
Abstract
AIMS The use of natural agents with anti-diabetic effect in combination therapy adds further positive clinical implications in the management of diabetes mellitus. Interestingly, quercetin is one of the most potent naturally occurring antioxidant which possesses various pharmacological actions including anti-diabetic effect. Thus, this research was conducted to assess the efficiency of a new combination from gliclazide and quercetin on glycemic control as well as pancreatic islets and beta cells in STZ-experimental model of diabetes. MAIN METHODS Diabetes has been induced by a single intraperitoneal injection of streptozotocin (STZ; 45 mg/kg) in adult male Wistar rats. For 3 consecutive weeks, diabetic rats were given orally either gliclazide (10 mg/kg), quercetin (50 mg/kg), or their combination. At the end of the experiment, histological, immunohistochemical and morphometrical examination of pancreatic tissues was performed. Furthermore, the changes in glucose metabolism, lipid profile, oxidative and inflammatory status were evaluated. KEY FINDINGS Treatment with gliclazide alone decreased serum glucose, total cholesterol, triglycerides, malondialdehyde, tumor necrosis factor-alpha and nuclear factor kappa-Beta while increased serum C-peptide, superoxide dismutase, reduced glutathione and adiponectin levels. Combined administration of quercetin with gliclazide markedly augmented serum superoxide dismutase and reduced glutathione more than gliclazide alone and normalized all the above-mentioned parameters. Besides, this combination therapy restored immunostaining intensity, number of pancreatic islets and beta cells along with its area and perimeter. SIGNIFICANCE Based on the aforementioned results, this combination could be considered a promising one in diabetes mellitus management.
Collapse
Affiliation(s)
- Noha F Abdelkader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Hebatollah E Eitah
- Department of Medicinal and Pharmaceutical Chemistry, Pharmacology Group, National Research Centre, Cairo, Egypt
| | - Yousreya A Maklad
- Department of Medicinal and Pharmaceutical Chemistry, Pharmacology Group, National Research Centre, Cairo, Egypt
| | | | - Manal A Badawi
- Department of Pathology, National Research Centre, Cairo, Egypt
| | - Sanaa A Kenawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
27
|
Vamecq J, Papegay B, Nuyens V, Boogaerts J, Leo O, Kruys V. Mitochondrial dysfunction, AMPK activation and peroxisomal metabolism: A coherent scenario for non-canonical 3-methylglutaconic acidurias. Biochimie 2019; 168:53-82. [PMID: 31626852 DOI: 10.1016/j.biochi.2019.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022]
Abstract
The occurrence of 3-methylglutaconic aciduria (3-MGA) is a well understood phenomenon in leucine oxidation and ketogenesis disorders (primary 3-MGAs). In contrast, its genesis in non-canonical (secondary) 3-MGAs, a growing-up group of disorders encompassing more than a dozen of inherited metabolic diseases, is a mystery still remaining unresolved for three decades. To puzzle out this anthologic problem of metabolism, three clues were considered: (i) the variety of disorders suggests a common cellular target at the cross-road of metabolic and signaling pathways, (ii) the response to leucine loading test only discriminative for primary but not secondary 3-MGAs suggests these latter are disorders of extramitochondrial HMG-CoA metabolism as also attested by their failure to increase 3-hydroxyisovalerate, a mitochondrial metabolite accumulating only in primary 3-MGAs, (iii) the peroxisome is an extramitochondrial site possessing its own pool and displaying metabolism of HMG-CoA, suggesting its possible involvement in producing extramitochondrial 3-methylglutaconate (3-MG). Following these clues provides a unifying common basis to non-canonical 3-MGAs: constitutive mitochondrial dysfunction induces AMPK activation which, by inhibiting early steps in cholesterol and fatty acid syntheses, pipelines cytoplasmic acetyl-CoA to peroxisomes where a rise in HMG-CoA followed by local dehydration and hydrolysis may lead to 3-MGA yield. Additional contributors are considered, notably for 3-MGAs associated with hyperammonemia, and to a lesser extent in CLPB deficiency. Metabolic and signaling itineraries followed by the proposed scenario are essentially sketched, being provided with compelling evidence from the literature coming in their support.
Collapse
Affiliation(s)
- Joseph Vamecq
- Inserm, CHU Lille, Univ Lille, Department of Biochemistry and Molecular Biology, Laboratory of Hormonology, Metabolism-Nutrition & Oncology (HMNO), Center of Biology and Pathology (CBP) Pierre-Marie Degand, CHRU Lille, EA 7364 RADEME, University of North France, Lille, France.
| | - Bérengère Papegay
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Vincent Nuyens
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Jean Boogaerts
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Oberdan Leo
- Laboratory of Immunobiology, Department of Molecular Biology, ULB Immunology Research Center (UIRC), Free University of Brussels (ULB), Gosselies, Belgium
| | - Véronique Kruys
- Laboratory of Molecular Biology of the Gene, Department of Molecular Biology, ULB Immunology Research Center (UIRC), Free University of Brussels (ULB), Gosselies, Belgium
| |
Collapse
|
28
|
Bule M, Abdurahman A, Nikfar S, Abdollahi M, Amini M. Antidiabetic effect of quercetin: A systematic review and meta-analysis of animal studies. Food Chem Toxicol 2019; 125:494-502. [DOI: 10.1016/j.fct.2019.01.037] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/27/2019] [Accepted: 01/30/2019] [Indexed: 12/22/2022]
|
29
|
Mangoni AA, Tommasi S, Zinellu A, Sotgia S, Carru C, Piga M, Erre GL. Repurposing existing drugs for cardiovascular risk management: a focus on methotrexate. Drugs Context 2018; 7:212557. [PMID: 30459819 PMCID: PMC6239018 DOI: 10.7573/dic.212557] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
About 20% of patients with a history of atherosclerotic cardiovascular disease will experience further cardiovascular events despite maximal pharmacological treatment with cardioprotective drugs. This highlights the presence of residual cardiovascular risk in a significant proportion of patients and the need for novel, more effective therapies. These therapies should ideally target different pathophysiological pathways involved in the onset and the progression of atherosclerosis, particularly the inflammatory and immune pathways. Methotrexate is a first-line disease-modifying antirheumatic drug that is widely used for the management of autoimmune and chronic inflammatory disorders. There is some in vitro and in vivo evidence that methotrexate might exert a unique combination of anti-inflammatory, blood pressure lowering, and vasculoprotective effects. Pending the results of large prospective studies investigating surrogate end-points as well as morbidity and mortality, repurposing methotrexate for cardiovascular risk management might represent a cost-effective strategy with immediate public health benefits. This review discusses the current challenges in the management of cardiovascular disease; the available evidence on the effects of methotrexate on inflammation, blood pressure, and surrogate markers of arterial function; suggestions for future research directions; and practical considerations with the use of methotrexate in this context.
Collapse
Affiliation(s)
- Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Sara Tommasi
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Quality Control Unit, University Hospital (AOUSS), Sassari, Italy
| | - Matteo Piga
- Rheumatology Unit, University Clinic and AOU of Cagliari, Italy
| | - Gian Luca Erre
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital (AOUSS) and University of Sassari, Sassari, Italy
| |
Collapse
|
30
|
The ecto-enzymes CD73 and adenosine deaminase modulate 5'-AMP-derived adenosine in myofibroblasts of the rat small intestine. Purinergic Signal 2018; 14:409-421. [PMID: 30269308 DOI: 10.1007/s11302-018-9623-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 08/21/2018] [Indexed: 12/12/2022] Open
Abstract
Adenosine is a versatile signaling molecule recognized to physiologically influence gut motor functions. Both the duration and magnitude of adenosine signaling in enteric neuromuscular function depend on its availability, which is regulated by the ecto-enzymes ecto-5'-nucleotidase (CD73), alkaline phosphatase (AP), and ecto-adenosine deaminase (ADA) and by dipyridamole-sensitive equilibrative transporters (ENTs). Our purpose was to assess the involvement of CD73, APs, ecto-ADA in the formation of AMP-derived adenosine in primary cultures of ileal myofibroblasts (IMFs). IMFs were isolated from rat ileum longitudinal muscle segments by means of primary explant technique and identified by immunofluorescence staining for vimentin and α-smooth muscle actin. IMFs confluent monolayers were exposed to exogenous 5'-AMP in the presence or absence of CD73, APs, ecto-ADA, or ENTs inhibitors. The formation of adenosine and its metabolites in the IMFs medium was monitored by high-performance liquid chromatography. The distribution of CD73 and ADA in IMFs was detected by confocal immunocytochemistry and qRT-PCR. Exogenous 5'-AMP was rapidly cleared being almost undetectable after 60-min incubation, while adenosine levels significantly increased. Treatment of IMFs with CD73 inhibitors markedly reduced 5'-AMP clearance whereas ADA blockade or inhibition of both ADA and ENTs prevented adenosine catabolism. By contrast, inhibition of APs did not affect 5'-AMP metabolism. Immunofluorescence staining and qRT-PCR analysis confirmed the expression of CD73 and ADA in IMFs. Overall, our data show that in IMFs an extracellular AMP-adenosine pathway is functionally active and among the different enzymatic pathways regulating extracellular adenosine levels, CD73 and ecto-ADA represent the critical catabolic pathway.
Collapse
|
31
|
Bagatini MD, dos Santos AA, Cardoso AM, Mânica A, Reschke CR, Carvalho FB. The Impact of Purinergic System Enzymes on Noncommunicable, Neurological, and Degenerative Diseases. J Immunol Res 2018; 2018:4892473. [PMID: 30159340 PMCID: PMC6109496 DOI: 10.1155/2018/4892473] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 07/03/2018] [Accepted: 07/22/2018] [Indexed: 12/11/2022] Open
Abstract
Evidences show that purinergic signaling is involved in processes associated with health and disease, including noncommunicable, neurological, and degenerative diseases. These diseases strike from children to elderly and are generally characterized by progressive deterioration of cells, eventually leading to tissue or organ degeneration. These pathological conditions can be associated with disturbance in the signaling mediated by nucleotides and nucleosides of adenine, in expression or activity of extracellular ectonucleotidases and in activation of P2X and P2Y receptors. Among the best known of these diseases are atherosclerosis, hypertension, cancer, epilepsy, Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). The currently available treatments present limited effectiveness and are mostly palliative. This review aims to present the role of purinergic signaling highlighting the ectonucleotidases E-NTPDase, E-NPP, E-5'-nucleotidase, and adenosine deaminase in noncommunicable, neurological, and degenerative diseases associated with the cardiovascular and central nervous systems and cancer. In conclusion, changes in the activity of ectonucleotidases were verified in all reviewed diseases. Although the role of ectonucleotidases still remains to be further investigated, evidences reviewed here can contribute to a better understanding of the molecular mechanisms of highly complex diseases, which majorly impact on patients' quality of life.
Collapse
Affiliation(s)
- Margarete Dulce Bagatini
- Coordenação Acadêmica, Universidade Federal da Fronteira Sul, Campus Chapecó, Chapecó, SC, Brazil
- Programa de Pós-graduação em Ciências Biológicas-Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | | - Andréia Machado Cardoso
- Coordenação Acadêmica, Universidade Federal da Fronteira Sul, Campus Chapecó, Chapecó, SC, Brazil
- Programa de Pós-graduação em Ciências Biológicas-Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Aline Mânica
- Programa de Pós-graduação em Ciências Biológicas-Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Cristina Ruedell Reschke
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Fabiano Barbosa Carvalho
- Programa de Pós-graduação em Ciências Biológicas-Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
- Laboratório de Pesquisa em Patologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
32
|
Fan K, Lin L, Ai Q, Wan J, Dai J, Liu G, Tang L, Yang Y, Ge P, Jiang R, Zhang L. Lipopolysaccharide-Induced Dephosphorylation of AMPK-Activated Protein Kinase Potentiates Inflammatory Injury via Repression of ULK1-Dependent Autophagy. Front Immunol 2018; 9:1464. [PMID: 29988556 PMCID: PMC6026648 DOI: 10.3389/fimmu.2018.01464] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 06/12/2018] [Indexed: 12/14/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a crucial metabolic regulator with profound modulatory activities on inflammation. Although the anti-inflammatory benefits of AMPK activators were well documented in experimental studies, the pathological significance of endogenous AMPK in inflammatory disorders largely remains unknown. This study investigated the phosphorylation status of endogenous AMPK and the potential roles of AMPK in mice with lipopolysaccharide (LPS)-induced lethal inflammation. The results indicated that LPS dose-dependently decreased the phosphorylation level of AMPK and its target protein acetyl-CoA carboxylase (ACC). Reactivation of AMPK with the AMPK activator A-769662 suppressed LPS-induced elevation of interleukin 6, alleviated histological abnormalities in lung and improved the survival of LPS-challenged mice. Treatment with A-769662 restored LPS-induced suppression of autophagy, inhibition of autophagy by 3-MA reversed the beneficial effects of A-769662. Treatment with A-769662 suppressed LPS-induced activation of mammalian target of rapamycin (mTOR), co-administration of mTOR activator abolished the beneficial effects of A-769662, and the suppressive effects of A-769662 on uncoordinated-51-like kinase 1 (ULK1) phosphorylation. Inhibition of ULK1 removed the beneficial effects of A-769662. These data indicated that LPS-induced dephosphorylation of AMPK could result in weakened inhibition of mTOR and repression of ULK1-dependent autophagy, which might potentiate the development of LPS-induced inflammatory injury. These data suggest that pharmacological restoration of AMPK activation might be a beneficial approach for the intervention of inflammatory disorders.
Collapse
Affiliation(s)
- Kerui Fan
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Ling Lin
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Qing Ai
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Jingyuan Wan
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Jie Dai
- Hospital of Chongqing University of Arts and Sciences, Chongqing, China
| | - Gang Liu
- Department of Emergency, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Li Tang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yongqiang Yang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Pu Ge
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
33
|
Fagone P, Mazzon E, Cavalli E, Bramanti A, Petralia MC, Mangano K, Al-Abed Y, Bramati P, Nicoletti F. Contribution of the macrophage migration inhibitory factor superfamily of cytokines in the pathogenesis of preclinical and human multiple sclerosis: In silico and in vivo evidences. J Neuroimmunol 2018; 322:46-56. [PMID: 29935880 DOI: 10.1016/j.jneuroim.2018.06.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/30/2018] [Accepted: 06/12/2018] [Indexed: 01/05/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine with pleiotropic actions involved in the pathogenesis of autoimmune disorders, including Multiple Sclerosis (MS). We have first evaluated in silico the involvement of MIF, its homologue D-DT, and the receptors CD74, CD44, CXCR2 and CXCR4 in encephalitogenic T cells from a mouse model of MS, the Experimental Allergic Encephalomyelitis (EAE), as well as in circulating T helper cells from MS patients. We show an upregulation of the receptors involved in MIF signaling both in the animal model and in patients. Also, a significant increase in MIF receptors is found in the CNS lesions associated to MS. Finally, the specific inhibitor of MIF, ISO-1, improved both ex vivo and in vivo the features of EAE. Overall, our data indicate that there is a significant involvement of the MIF pathway in MS ethiopathogenesis and that interventions specifically blocking MIF receptors may represent useful therapeutic approaches in the clinical setting.
Collapse
Affiliation(s)
- Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Eugenio Cavalli
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Maria Cristina Petralia
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Department of Formative Processes, University of Catania, Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, Manhasset, New York, United States
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
34
|
Antonioli L, Fornai M, Blandizzi C, Pacher P, Haskó G. Adenosine signaling and the immune system: When a lot could be too much. Immunol Lett 2018; 205:9-15. [PMID: 29702147 DOI: 10.1016/j.imlet.2018.04.006] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 04/23/2018] [Indexed: 02/07/2023]
Abstract
Adenosine is increasingly recognized as a key mediator of the immune response. Signals delivered by extracellular adenosine are detected and transduced by G-protein-coupled cell-surface receptors, classified into four subtypes: A1, A2A, A2B and A3. These receptors, expressed virtually on all immune cells, modulate all aspects of immune/inflammatory responses. These immunoregulatory effects, which are mostly anti-inflammatory, contribute to the general tissue protective effects of adenosine and its receptors. In some instances, however, the effect of adenosine on the immune system is deleterious, as prolonged adenosine signaling can hinder anti-tumor and antibacterial immunity, thereby promoting cancer development and progression and sepsis, respectively.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy; Department of Anesthesiology, Columbia University, New York, NY, 10032, USA.
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy
| | - Pál Pacher
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
35
|
Abstract
In addition to the well-characterized role of AMPK in the regulation of nutrient metabolism, it is increasingly clear that AMPK activation has multiple actions on inflammatory signalling. Here we describe methods to identify effects of AMPK activity on pro-inflammatory signalling, specifically (1) the nuclear localization of the key inflammatory mediators nuclear factor-κB (NFκB) and phosphorylated c-Jun N-terminal kinase (JNK), (2) preparation of conditioned medium to analyze the secretion of cytokines/chemokines, and (3) the pro-inflammatory adhesion of leukocytes to cultured cells.
Collapse
Affiliation(s)
- Sarah J Mancini
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Ian P Salt
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
36
|
Wang Y, Zhou L, Li Y, Guo L, Zhou Z, Xie H, Hou Y, Wang B. The Effects of Berberine on Concanavalin A-Induced Autoimmune Hepatitis (AIH) in Mice and the Adenosine 5'-Monophosphate (AMP)-Activated Protein Kinase (AMPK) Pathway. Med Sci Monit 2017; 23:6150-6161. [PMID: 29283990 PMCID: PMC5753750 DOI: 10.12659/msm.907377] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Background Berberine, a herbal extract, has been reported to protect against inflammatory disorders. The adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK) signaling pathway can be activated by berberine and inhibited by the synthetic, reversible AMP-competitive inhibitor, Compound C. The aim of this study was to investigate the effects of berberine on concanavalin A (Con A)-induced autoimmune hepatitis (AIH) in mice via the AMPK pathway. Material/Methods BALB/c mice were treated with berberine, with or without Compound C, followed by treatment with Con A. Serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were measured. Liver tissue histology was performed to evaluate hepatic injury and AIH. Cytokine levels in serum and hepatic tissue were measured by enzyme-linked immunoassay (ELISA) and used quantitative polymerase chain reaction (qPCR). Levels of phosphorylated acetyl coenzyme-A carboxylase (ACC), representing AMPK activation, were detected by Western blotting. Results Serum ALT and AST levels were significantly reduced by berberine (100 and 200 mg/kg/day) in mice with Con A-induced hepatitis. Berberine also reduced Con A-induced hepatocyte swelling, cell death, and infiltration of leukocytes. Serum levels of tumor necrosis factor (TNF)-alpha, interferon (IF)-gamma, interleukin (IL)-2, and IL-1beta were reduced by berberine pre-treatment; levels of serum IL-10, an anti-inflammatory cytokine, was elevated. These protective effects of berberine on Con-A-induced AIH were reversed by treatment with Compound C. Conclusions In a murine model of Con A-induced AIH, berberine treatment reduced hepatic injury via activation of the AMPK pathway. Further studies are recommended to determine the potential therapeutic role for berberine in AIH.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Department of Gastroenterology, Affiliated Hospital of Hebei University, Baoding, Hebei, China (mainland)
| | - Lu Zhou
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Yanni Li
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Liping Guo
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Zhe Zhou
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Haoran Xie
- College of Medicine, Hebei University, Baoding, Hebei, China (mainland)
| | - Yingjian Hou
- College of Medicine, Hebei University, Baoding, Hebei, China (mainland)
| | - Bangmao Wang
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| |
Collapse
|
37
|
Protective Effects of Methotrexate against Proatherosclerotic Cytokines: A Review of the Evidence. Mediators Inflamm 2017; 2017:9632846. [PMID: 29430085 PMCID: PMC5753000 DOI: 10.1155/2017/9632846] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/02/2017] [Accepted: 11/26/2017] [Indexed: 12/16/2022] Open
Abstract
There is good epidemiological evidence that patients with autoimmune rheumatic disease states, particularly rheumatoid arthritis, have an increased risk of cardiovascular morbidity and mortality when compared to the general population. The presence of a chronic systemic proinflammatory state in this patient group disrupts the structural and functional integrity of the endothelium and the arterial wall, favouring the onset and progression of atherosclerosis. A significant role in the detrimental effects of inflammation on endothelial function and vascular homeostasis is played by specific proatherosclerotic cytokines such as tumour necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), and interleukin-6 (IL-6). Recent systematic reviews and meta-analyses have shown that treatment with methotrexate, a first-line disease-modifying antirheumatic drug (DMARD), is associated with a significant reduction in atherosclerosis-mediated cardiovascular events, such as myocardial infarction and stroke, and mortality, when compared to other DMARDs. This suggests that methotrexate might exert specific protective effects against vascular inflammation and atherosclerosis in the context of autoimmune rheumatic disease. This review discusses the available evidence regarding the potential antiatherosclerotic effects of methotrexate through the inhibition of TNF-α, IL-1, and IL-6 and provides suggestions for future experimental and human studies addressing this issue.
Collapse
|
38
|
Deng Z, Rong Y, Teng Y, Mu J, Zhuang X, Tseng M, Samykutty A, Zhang L, Yan J, Miller D, Suttles J, Zhang HG. Broccoli-Derived Nanoparticle Inhibits Mouse Colitis by Activating Dendritic Cell AMP-Activated Protein Kinase. Mol Ther 2017; 25:1641-1654. [PMID: 28274798 DOI: 10.1016/j.ymthe.2017.01.025] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 12/19/2022] Open
Abstract
The intestinal immune system is continuously exposed to massive amounts of nanoparticles derived from food. Whether nanoparticles from plants we eat daily have a role in maintaining intestinal immune homeostasis is poorly defined. Here, we present evidence supporting our hypothesis that edible nanoparticles regulate intestinal immune homeostasis by targeting dendritic cells (DCs). Using three mouse colitis models, our data show that orally given nanoparticles isolated from broccoli extracts protect mice against colitis. Broccoli-derived nanoparticle (BDN)-mediated activation of adenosine monophosphate-activated protein kinase (AMPK) in DCs plays a role in not only prevention of DC activation but also induction of tolerant DCs. Adoptively transferring DCs pre-pulsed with total BDN lipids, but not sulforaphane (SFN)-depleted BDN lipids, prevented DSS-induced colitis in C57BL/6 (B6) mice, supporting the role of BDN SFN in the induction of DC tolerance. Adoptively transferring AMPK+/+, but not AMPK-/-, DCs pre-pulsed with SFN prevented DSS-induced colitis in B6 mice, further supporting the DC AMPK role in SFN-mediated prevention of DSS-induced colitis. This finding could open new preventive or therapeutic avenues to address intestinal-related inflammatory diseases via activating AMPK.
Collapse
Affiliation(s)
- Zhongbin Deng
- Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Yuan Rong
- Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Yun Teng
- Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Jingyao Mu
- Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Xiaoying Zhuang
- Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Michael Tseng
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY 40202, USA
| | - Abhilash Samykutty
- Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Lifeng Zhang
- Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Jun Yan
- Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Donald Miller
- Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Jill Suttles
- Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Huang-Ge Zhang
- Robley Rex VA Medical Center, Louisville, KY 40206, USA; Department of Microbiology & Immunology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|