1
|
Raisinghani AB, Luis SA. Treatment options to break the cycle of recurrent pericarditis. Curr Opin Cardiol 2025; 40:107-114. [PMID: 39819644 DOI: 10.1097/hco.0000000000001201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
PURPOSE OF REVIEW This review provides a contemporary, evidence-based update on the pathophysiological mechanisms and rapidly evolving therapeutic options for recurrent pericarditis. RECENT FINDINGS Recent studies have elucidated the pathogenesis of recurrent pericarditis, identifying autoinflammation as a key mechanism and interleukin-1 (IL-1) as a central modulator of the inflammatory cascade. Multiple clinical trials have investigated novel therapeutic approaches, particularly focusing on IL-1 inhibition. The recent FDA approval of IL-1 pathway blockade for recurrent pericarditis has revolutionized treatment, offering patients significantly improved quality of life and symptom management. SUMMARY The enhanced understanding of the autoinflammatory nature of recurrent pericarditis, coupled with groundbreaking advances in targeted therapies, has transformed the treatment landscape for affected patients. The emergence of IL-1 inhibitors as an effective therapeutic option promises substantial improvements in clinical outcomes and patient well being. Clinicians must familiarize themselves with these new treatments, their efficacy, and potential limitations to optimize patient care and guide therapeutic decision-making in this challenging condition.
Collapse
Affiliation(s)
| | - Sushil Allen Luis
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
2
|
Xu G, Yu J, Lyu J, Zhan M, Xu J, Huang M, Zhao R, Li Y, Zhu J, Feng J, Tan S, Ran P, Su Z, Liu X, Zhao J, Zhang H, Xu C, Chang J, Hou Y, Ding C. Proteogenomic Landscape of Breast Ductal Carcinoma Reveals Tumor Progression Characteristics and Therapeutic Targets. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401041. [PMID: 39418072 PMCID: PMC11633542 DOI: 10.1002/advs.202401041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Multi-omics studies of breast ductal carcinoma (BRDC) have advanced the understanding of the disease's biology and accelerated targeted therapies. However, the temporal order of a series of biological events in the progression of BRDC is still poorly understood. A comprehensive proteogenomic analysis of 224 samples from 168 patients with malignant and benign breast diseases is carried out. Proteogenomic analysis reveals the characteristics of linear multi-step progression of BRDC, such as tumor protein P53 (TP53) mutation-associated estrogen receptor 1 (ESR1) overexpression is involved in the transition from ductal hyperplasia (DH) to ductal carcinoma in situ (DCIS). 6q21 amplification-associated nuclear receptor subfamily 3 group C member 1 (NR3C1) overexpression helps DCIS_Pure (pure DCIS, no histologic evidence of invasion) cells avoid immune destruction. The T-cell lymphoma invasion and metastasis 1, androgen receptor, and aldo-keto reductase family 1 member C1 (TIAM1-AR-AKR1C1) axis promotes cell invasion and migration in DCIS_adjIDC (DCIS regions of invasive cancers). In addition, AKR1C1 is identified as a potential therapeutic target and demonstrated the inhibitory effect of aspirin and dydrogesterone as its inhibitors on tumor cells. The integrative multi-omics analysis helps to understand the progression of BRDC and provides an opportunity to treat BRDC in different stages.
Collapse
Affiliation(s)
- Ganfei Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Juan Yu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jiacheng Lyu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Mengna Zhan
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jie Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Minjing Huang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Rui Zhao
- Institute for Developmental and Regenerative Cardiovascular MedicineMOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
| | - Yan Li
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jiajun Zhu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jinwen Feng
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Subei Tan
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Peng Ran
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Zhenghua Su
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Xinhua Liu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jianyuan Zhao
- Institute for Developmental and Regenerative Cardiovascular MedicineMOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
| | - Hongwei Zhang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Chen Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jun Chang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Yingyong Hou
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Chen Ding
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
- Departments of Cancer Research InstituteAffiliated Cancer Hospital of Xinjiang Medical UniversityXinjiang Key Laboratory of Translational Biomedical EngineeringUrumqi830000P. R. China
| |
Collapse
|
3
|
Ning S, Zhang Z, Zhou C, Wang B, Liu Z, Feng B. Cross-talk between macrophages and gut microbiota in inflammatory bowel disease: a dynamic interplay influencing pathogenesis and therapy. Front Med (Lausanne) 2024; 11:1457218. [PMID: 39355844 PMCID: PMC11443506 DOI: 10.3389/fmed.2024.1457218] [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: 06/30/2024] [Accepted: 08/28/2024] [Indexed: 10/03/2024] Open
Abstract
Inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD), is a group of chronic immune-mediated gastrointestinal disorders. The etiology of IBD is multifactorial, involving genetic susceptibility, environmental factors, and a complex interplay between the gut microbiota and the host's immune system. Intestinal resident macrophages play an important role in the pathogenesis and progress of IBD, as well as in maintaining intestinal homeostasis and facilitating tissue repair. This review delves into the intricate relationship between intestinal macrophages and gut microbiota, highlighting their pivotal roles in IBD pathogenesis. We discuss the impact of macrophage dysregulation and the consequent polarization of different phenotypes on intestinal inflammation. Furthermore, we explore the compositional and functional alterations in gut microbiota associated with IBD, including the emerging significance of fungal and viral components. This review also examines the effects of current therapeutic strategies, such as 5-aminosalicylic acid (5-ASA), antibiotics, steroids, immunomodulators, and biologics, on gut microbiota and macrophage function. We underscore the potential of fecal microbiota transplantation (FMT) and probiotics as innovative approaches to modulate the gut microbiome in IBD. The aim is to provide insights into the development of novel therapies targeting the gut microbiota and macrophages to improve IBD management.
Collapse
Affiliation(s)
- Shiyang Ning
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhe Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chuan Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Binbin Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhanju Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Shanghai Tenth People’s Hospital, Shanghai, China
| | - Baisui Feng
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Guo J, Li L, Cai Y, Kang Y. The development of probiotics and prebiotics therapy to ulcerative colitis: a therapy that has gained considerable momentum. Cell Commun Signal 2024; 22:268. [PMID: 38745207 PMCID: PMC11094941 DOI: 10.1186/s12964-024-01611-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 04/10/2024] [Indexed: 05/16/2024] Open
Abstract
Ulcerative colitis (UC) is increasingly common, and it is gradually become a kind of global epidemic. UC is a type of inflammatory bowel disease (IBD), and it is a lifetime recurrent disease. UC as a common disease has become a financial burden for many people and has the potential to develop into cancer if not prevented or treated. There are multiple factors such as genetic factors, host immune system disorders, and environmental factors to cause UC. A growing body of research have suggested that intestinal microbiota as an environmental factor play an important role in the occurrence and development of UC. Meanwhile, evidence to date suggests that manipulating the gut microbiome may represent effective treatment for the prevention or management of UC. In addition, the main clinical drugs to treat UC are amino salicylate and corticosteroid. These clinical drugs always have some side effects and low success rate when treating patients with UC. Therefore, there is an urgent need for safe and efficient methods to treat UC. Based on this, probiotics and prebiotics may be a valuable treatment for UC. In order to promote the wide clinical application of probiotics and prebiotics in the treatment of UC. This review aims to summarize the recent literature as an aid to better understanding how the probiotics and prebiotics contributes to UC while evaluating and prospecting the therapeutic effect of the probiotics and prebiotics in the treatment of UC based on previous publications.
Collapse
Affiliation(s)
- Jing Guo
- Department of microbiology and immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Liping Li
- Department of microbiology and immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yue Cai
- Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yongbo Kang
- Department of microbiology and immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
5
|
Jia B, Zhang B, Li J, Qin J, Huang Y, Huang M, Ming Y, Jiang J, Chen R, Xiao Y, Du J. Emerging polymeric materials for treatment of oral diseases: design strategy towards a unique oral environment. Chem Soc Rev 2024; 53:3273-3301. [PMID: 38507263 DOI: 10.1039/d3cs01039b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Oral diseases are prevalent but challenging diseases owing to the highly movable and wet, microbial and inflammatory environment. Polymeric materials are regarded as one of the most promising biomaterials due to their good compatibility, facile preparation, and flexible design to obtain multifunctionality. Therefore, a variety of strategies have been employed to develop materials with improved therapeutic efficacy by overcoming physicobiological barriers in oral diseases. In this review, we summarize the design strategies of polymeric biomaterials for the treatment of oral diseases. First, we present the unique oral environment including highly movable and wet, microbial and inflammatory environment, which hinders the effective treatment of oral diseases. Second, a series of strategies for designing polymeric materials towards such a unique oral environment are highlighted. For example, multifunctional polymeric materials are armed with wet-adhesive, antimicrobial, and anti-inflammatory functions through advanced chemistry and nanotechnology to effectively treat oral diseases. These are achieved by designing wet-adhesive polymers modified with hydroxy, amine, quinone, and aldehyde groups to provide strong wet-adhesion through hydrogen and covalent bonding, and electrostatic and hydrophobic interactions, by developing antimicrobial polymers including cationic polymers, antimicrobial peptides, and antibiotic-conjugated polymers, and by synthesizing anti-inflammatory polymers with phenolic hydroxy and cysteine groups that function as immunomodulators and electron donors to reactive oxygen species to reduce inflammation. Third, various delivery systems with strong wet-adhesion and enhanced mucosa and biofilm penetration capabilities, such as nanoparticles, hydrogels, patches, and microneedles, are constructed for delivery of antibiotics, immunomodulators, and antioxidants to achieve therapeutic efficacy. Finally, we provide insights into challenges and future development of polymeric materials for oral diseases with promise for clinical translation.
Collapse
Affiliation(s)
- Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Beibei Zhang
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jianhua Li
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jinlong Qin
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Yisheng Huang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Mingshu Huang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Yue Ming
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Jingjing Jiang
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Ran Chen
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Yufen Xiao
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| |
Collapse
|
6
|
Kim YG, Lee Y, Lee N, Soh M, Kim D, Hyeon T. Ceria-Based Therapeutic Antioxidants for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2210819. [PMID: 36793245 DOI: 10.1002/adma.202210819] [Citation(s) in RCA: 56] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/20/2023] [Indexed: 06/18/2023]
Abstract
The growing interest in nanomedicine over the last 20 years has carved out a research field called "nanocatalytic therapy," where catalytic reactions mediated by nanomaterials are employed to intervene in disease-critical biomolecular processes. Among many kinds of catalytic/enzyme-mimetic nanomaterials investigated thus far, ceria nanoparticles stand out from others owing to their unique scavenging properties against biologically noxious free radicals, including reactive oxygen species (ROS) and reactive nitrogen species (RNS), by exerting enzyme mimicry and nonenzymatic activities. Much effort has been made to utilize ceria nanoparticles as self-regenerating antioxidative and anti-inflammatory agents for various kinds of diseases, given the detrimental effects of ROS and RNS therein that need alleviation. In this context, this review is intended to provide an overview as to what makes ceria nanoparticles merit attention in disease therapy. The introductory part describes the characteristics of ceria nanoparticles as an oxygen-deficient metal oxide. The pathophysiological roles of ROS and RNS are then presented, as well as their scavenging mechanisms by ceria nanoparticles. Representative examples of recent ceria-nanoparticle-based therapeutics are summarized by categorization into organ and disease types, followed by the discussion on the remaining challenges and future research directions.
Collapse
Affiliation(s)
- Young Geon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yunjung Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nohyun Lee
- School of Advanced Materials Engineering, Kookmin University, Seoul, 02707, Republic of Korea
| | - Min Soh
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Center for Advanced Pharmaceutical Technology, HyeonTechNBio, Inc., Seoul, 08826, Republic of Korea
| | - Dokyoon Kim
- Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan, 15588, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
7
|
Fan G, Huang L, Wang M, Kuang H, Li Y, Yang X. GPAT3 deficiency attenuates corticosterone-caused hepatic steatosis and oxidative stress through GSK3β/Nrf2 signals. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167007. [PMID: 38185063 DOI: 10.1016/j.bbadis.2023.167007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/13/2023] [Accepted: 12/26/2023] [Indexed: 01/09/2024]
Abstract
The development of nonalcoholic fatty liver disease (NAFLD) may worsen due to chronic stress or prolonged use of glucocorticoids. Glycerol-3-phosphate acyltransferase 3 (GPAT3), has a function in obesity and serves as a key rate-limiting enzyme that regulates triglyceride synthesis. However, the precise impact of GPAT3 on corticosterone (CORT)-induced NAFLD and its underlying molecular mechanism remain unclear. For our in vivo experiments, we utilized male and female mice that were GPAT3-/- and wild type (WT) and treated them with CORT for a duration of 4 weeks. In our in vitro experiments, we transfected AML12 cells with GPAT3 siRNA and subsequently treated them with CORT. Under CORT-treated conditions, the absence of GPAT3 greatly improved obesity and hepatic steatosis while enhancing the expression of genes involved in fatty acid oxidation, as evidenced by our findings. In addition, the deletion of GPAT3 significantly inhibited the production of reactive oxygen species (ROS), increased the expression of antioxidant genes, and recovered the mitochondrial membrane potential in AML12 cells treated with CORT. In terms of mechanism, the absence of GPAT3 encouraged the activation of the glycogen synthase kinase 3β (GSK3β)/nuclear factor-erythroid 2 related factor 2 (Nrf2) pathway, which served as a defense mechanism against liver fat accumulation and oxidative stress. Furthermore, GPAT3 expression was directly controlled at the transcriptional level by the glucocorticoid receptor (GR). Collectively, our findings suggest that GPAT3 deletion significantly alleviated hepatic steatosis and oxidative stress through promoting GSK3β/Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Guoqiang Fan
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Lingling Huang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Mengxuan Wang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Haoran Kuang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yanfei Li
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xiaojing Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
8
|
Zhou FW, Liu C, Li DZ, Zhang Y, Zhou FC. Efficacy and safety of corticosteroid therapy in patients with cardiac arrest: A meta-analysis of randomized controlled trials. Am J Emerg Med 2024; 75:111-118. [PMID: 37939521 DOI: 10.1016/j.ajem.2023.10.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 10/05/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND The clinical benefits of steroid therapy during cardiac arrest (CA) are unclear. Several recent clinical trials have shown that administering corticosteroid therapy during CA may improve patient outcomes. The purpose of the present study was to determine whether providing corticosteroids improves outcomes for patients following CA. METHODS We searched the PubMed, Embase, Cochrane Library, Web of Science and CNKI databases for randomized controlled trials comparing corticosteroid therapy to placebo during CA. RESULTS Eleven relevant studies involving a total of 2273 patients were included in the meta-analysis. The statistical analysis showed that corticosteroid treatment during CA was significantly associated with an increased rate of sustained return of spontaneous circulation (ROSC) (OR: 2.05, 95% CI: 1.24 to 3.37, P < 0.01). Corticosteroid treatment during CA did not show a significant benefit in favorable neurological outcomes (OR: 1.13, 95% CI: 0.81 to 1.58, P = 0.49) or overall survival rate at hospital discharge (OR: 1.29, 95% CI: 0.74 to 2.26, P = 0.38). However, in the subgroup analysis, we found that patients had a significantly increased survival rate and ROSC if the dose of corticosteroid therapy above 100 mg methylprednisolone. The statistical analysis revealed no significant differences in adverse events. CONCLUSION High-dose corticosteroid treatment (above 100 mg methylprednisolone) is associated with better overall survival rate at hospital discharge and ROSC outcomes. However, there is uncertainty regarding whether this treatment results in a benefit or harm to the favorable neurological outcomes at hospital discharge.
Collapse
Affiliation(s)
- Fa-Wei Zhou
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Emergency, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Chang Liu
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - De-Zhong Li
- Department of Emergency, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Yong Zhang
- Department of Nephrology, Jianli People's Hospital, Jianli, China.
| | - Fa-Chun Zhou
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
9
|
Milara J, Morell A, Roger I, Montero P, Cortijo J. Mechanisms underlying corticosteroid resistance in patients with asthma: a review of current knowledge. Expert Rev Respir Med 2023; 17:701-715. [PMID: 37658478 DOI: 10.1080/17476348.2023.2255124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/25/2023] [Accepted: 08/31/2023] [Indexed: 09/03/2023]
Abstract
INTRODUCTION Corticosteroids are the most cost-effective anti-inflammatory drugs available for the treatment of asthma. Despite their effectiveness, several asthmatic patients have corticosteroid resistance or insensitivity and exhibit a poor response. Corticosteroid insensitivity implies a poor prognosis due to challenges in finding alternative therapeutic options for asthma. AREAS COVERED In this review, we describe asthma phenotypes and endotypes, as well as their differential responsiveness to corticosteroids. In addition, we describe the mechanism of action of corticosteroids underlying their regulation of the expression of glucocorticoid receptors (GRs) and their anti-inflammatory effects. Furthermore, we summarize the mechanistic evidence underlying corticosteroid-insensitive asthma, which is mainly related to changes in GR gene expression, structure, and post-transcriptional modifications. Finally, various pharmacological strategies designed to reverse corticosteroid insensitivity are discussed. EXPERT OPINION Corticosteroid insensitivity is influenced by the asthma phenotype, endotype, and severity, and serves as an indication for biological therapy. The molecular mechanisms underlying corticosteroid-insensitive asthma have been used to develop targeted therapeutic strategies. However, the lack of clinical trials prevents the clinical application of these treatments.
Collapse
Affiliation(s)
- Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy department, University General Hospital of Valencia, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
| | - Anselm Morell
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Inés Roger
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
| | - Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy department, University General Hospital of Valencia, Valencia, Spain
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
| |
Collapse
|
10
|
Cortisol Interaction with Aquaporin-2 Modulates Its Water Permeability: Perspectives for Non-Genomic Effects of Corticosteroids. Int J Mol Sci 2023; 24:ijms24021499. [PMID: 36675012 PMCID: PMC9862916 DOI: 10.3390/ijms24021499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Aquaporins (AQPs) are water channels widely distributed in living organisms and involved in many pathophysiologies as well as in cell volume regulations (CVR). In the present study, based on the structural homology existing between mineralocorticoid receptors (MRs), glucocorticoid receptors (GRs), cholesterol consensus motif (CCM) and the extra-cellular vestibules of AQPs, we investigated the binding of corticosteroids on the AQP family through in silico molecular dynamics simulations of AQP2 interactions with cortisol. We propose, for the first time, a putative AQPs corticosteroid binding site (ACBS) and discussed its conservation through structural alignment. Corticosteroids can mediate non-genomic effects; nonetheless, the transduction pathways involved are still misunderstood. Moreover, a growing body of evidence is pointing toward the existence of a novel membrane receptor mediating part of these rapid corticosteroids' effects. Our results suggest that the naturally produced glucocorticoid cortisol inhibits channel water permeability. Based on these results, we propose a detailed description of a putative underlying molecular mechanism. In this process, we also bring new insights on the regulatory function of AQPs extra-cellular loops and on the role of ions in tuning the water permeability. Altogether, this work brings new insights into the non-genomic effects of corticosteroids through the proposition of AQPs as the membrane receptor of this family of regulatory molecules. This original result is the starting point for future investigations to define more in-depth and in vivo the validity of this functional model.
Collapse
|
11
|
Minchenko DO, Khita OO, Viletska YM, Sliusar MY, Rudnytska OV, Kozynkevych HE, Bezrodnyi BH, Khikhlo YP, Minchenko OH. Cortisol controls endoplasmic reticulum stress and hypoxia dependent regulation of insulin receptor and related genes expression in HEK293 cells. Endocr Regul 2023; 58:1-10. [PMID: 38345493 DOI: 10.2478/enr-2024-0001] [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] [Indexed: 02/15/2024] Open
Abstract
Objective. Glucocorticoids are important stress-responsive regulators of insulin-dependent metabolic processes realized through specific changes in genome function. The aim of this study was to investigate the impact of cortisol on insulin receptor and related genes expression in HEK293 cells upon induction the endoplasmic reticulum (ER) stress by tunicamycin and hypoxia. Methods. The human embryonic kidney cell line HEK293 was used. Cells were exposed to cortisol (10 µM) as well as inducers of hypoxia (dimethyloxalylglycine, DMOG; 0.5 mM) and ER stress (tunicamycin; 0.2 µg/ml) for 4 h. The RNA from these cells was extracted and reverse transcribed. The expression level of INSR, IRS2, and INSIG2 and some ER stress responsive genes encoding XBP1n, non-spliced variant, XBP1s, alternatively spliced variant of XBP1, and DNAJB9 proteins, was measured by quantitative polymerase chain reaction and normalized to ACTB. Results. We showed that exposure of HEK293 cells to cortisol elicited up-regulation in the expression of INSR and DNAJB9 genes and down-regulation of XBP1s, XBP1n, IRS2, and INSIG2 mRNA levels. At the same time, induction of hypoxia by DMOG led to an up-regulation of the expression level of most studied mRNAs: XBP1s and XBP1n, IRS2 and INSIG2, but did not change significantly INSR and DNAJB9 gene expression. We also showed that combined impact of cortisol and hypoxia introduced the up-regulation of INSR and suppressed XBP1n mRNA expression levels. Furthermore, the exposure of HEK293 cells to tunicamycin affected the expression of IRS2 gene and increased the level of XBP1n mRNA. At the same time, the combined treatment of these cells with cortisol and inductor of ER stress had much stronger impact on the expression of all the tested genes: strongly increased the mRNA level of ER stress dependent factors XBP1s and DNAJB9 as well as INSR and INSIG2, but down-regulated IRS2 and XBP1n. Conclusion. Taken together, the present study indicates that cortisol may interact with ER stress and hypoxia in the regulation of ER stress dependent XBP1 and DNAJB9 mRNA expression as well as INSR and its signaling and that this corticosteroid hormone modified the impact of hypoxia and especially tunicamycin on the expression of most studied genes in HEK293 cells. These data demonstrate molecular mechanisms of glucocorticoids interaction with ER stress and insulin signaling at the cellular level.
Collapse
Affiliation(s)
- Dmytro O Minchenko
- Department of Molecular Biology, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kiev 01030, Ukraine
- Departments of Pediatrics No.1 and Surgery, National Bohomolets Medical University, Kyiv, Ukraine
| | - Olena O Khita
- Department of Molecular Biology, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kiev 01030, Ukraine
| | - Yuliia M Viletska
- Department of Molecular Biology, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kiev 01030, Ukraine
| | - Myroslava Y Sliusar
- Department of Molecular Biology, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kiev 01030, Ukraine
| | - Olha V Rudnytska
- Department of Molecular Biology, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kiev 01030, Ukraine
| | - Halyna E Kozynkevych
- Departments of Pediatrics No.1 and Surgery, National Bohomolets Medical University, Kyiv, Ukraine
| | - Borys H Bezrodnyi
- Departments of Pediatrics No.1 and Surgery, National Bohomolets Medical University, Kyiv, Ukraine
| | - Yevgen P Khikhlo
- Department of Molecular Biology, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kiev 01030, Ukraine
| | - Oleksandr H Minchenko
- Department of Molecular Biology, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kiev 01030, Ukraine
| |
Collapse
|
12
|
Galeev SR, Gautier SV. Risks and ways of preventing kidney dysfunction in drug-induced immunosuppression in solid organ recipients. RUSSIAN JOURNAL OF TRANSPLANTOLOGY AND ARTIFICIAL ORGANS 2022. [DOI: 10.15825/1995-1191-2022-4-24-38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunosuppressive therapy (IMT) is the cornerstone of treatment after transplantation. The goal of immunosuppression is to prevent acute and chronic rejection while maximizing patient survival and long-term graft function. However, the expected effects of IMT must be balanced against the major adverse effects of these drugs and their toxicity. The purpose of this review is to summarize world experience on current immunosuppressive strategies and to assess their effects on renal function.
Collapse
Affiliation(s)
- Sh. R. Galeev
- Shumakov National Medical Research Center of Transplantology and Artificial Organs
| | - S. V. Gautier
- Shumakov National Medical Research Center of Transplantology and Artificial Organs; Sechenov University
| |
Collapse
|
13
|
Winkler MS, Osuchowski MF, Payen D, Torres A, Dickel S, Skirecki T. Renaissance of glucocorticoids in critical care in the era of COVID-19: ten urging questions. Crit Care 2022; 26:308. [PMID: 36209188 PMCID: PMC9547674 DOI: 10.1186/s13054-022-04185-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022] Open
Abstract
The 40-year-old experience with glucocorticosteroids (GCs) in the context of severe infections is complex and troublesome. Recently, however, a clear indication for GCs in severe COVID-19 has been established. This may constitute a harbinger of a wider use of GCs in critical illnesses. A fundamental prerequisite of such an action is a better understanding of the heterogeneity of critical illness and GCs operationalization within the precision medicine approach. In this perspective, we formulate ten major questions regarding the use of GCs in critical illness. Answering them will likely facilitate a new era of effective and personalized GCs use in modern critical care.
Collapse
Affiliation(s)
- Martin S. Winkler
- grid.7450.60000 0001 2364 4210Department of Anaesthesiology and Intensive Care Medicine, University Medical Center, Georg-August University of Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Marcin F. Osuchowski
- grid.420022.60000 0001 0723 5126Ludwig Boltzmann Institute for Traumatology Ludwig Boltzmann Institute for Trauma in Cooperation with the AUVA, Vienna, Austria
| | - Didier Payen
- grid.508487.60000 0004 7885 7602Emeritus Professor of Anesthesiology and Critical Care, University of Paris 7, Cité, Sorbonne, Paris, France
| | - Antoni Torres
- grid.413448.e0000 0000 9314 1427Servei de Pneumologia, Hospital Clinic IDIBAPS, Universitat de Barcelona, Centro de Investigación Biomedica En Red-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Steffen Dickel
- grid.7450.60000 0001 2364 4210Department of Anaesthesiology and Intensive Care Medicine, University Medical Center, Georg-August University of Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Tomasz Skirecki
- grid.414852.e0000 0001 2205 7719Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| |
Collapse
|
14
|
Arai Y, Kinoshita Y, Kobayashi T, Takahashi Y, Ohyama T, Yokota N, Sugai Y, Takano S, Hamasaki Y, Kaneko U, Kanada S. A rare case of eosinophilic gastrointestinal disorders with short bowel syndrome after strangulated bowel obstruction. Surg Case Rep 2022; 8:168. [PMID: 36103004 PMCID: PMC9474956 DOI: 10.1186/s40792-022-01527-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/10/2022] [Indexed: 11/10/2022] Open
Abstract
Background Short bowel syndrome (SBS) is a rare yet costly disease with an incidence rate of 3 per million people. Herein, we report a rare case of eosinophilic gastrointestinal disorders (EGIDs) with SBS after strangulated bowel obstruction. Case presentation A 5-year-old male had a necrotic intestine of 340 cm resected due to strangulated bowel obstruction caused by an intestinal mesenteric hiatal hernia. The length of the residual intestine was 51 cm. Bloody stools appeared 19 days postoperatively. Colonoscopy showed diffuse redness of the colonic mucosa, and pathological findings showed moderate chronic inflammatory cellular infiltration. On blood examination, the eosinophil count was > 30%. EGIDs with short bowel syndrome (SBS) were suspected. Because his symptoms did not improve with initial nutrition therapy, he was transferred to our hospital 5 months after the operation. Prednisolone was administrated at an initial dose of 1.4 mg/kg/day, 6 days after his transfer. Bloody stools disappeared after prednisolone administration. Seven months after discharge, he had no bloody stool recurrence. Conclusion The risk of developing secondary EGIDs in children with SBS should be considered, and postoperative management should include attention to abdominal symptoms and elevated eosinophil counts on blood examination.
Collapse
|
15
|
Zhou Q, Li T, Wang K, Zhang Q, Geng Z, Deng S, Cheng C, Wang Y. Current status of xenotransplantation research and the strategies for preventing xenograft rejection. Front Immunol 2022; 13:928173. [PMID: 35967435 PMCID: PMC9367636 DOI: 10.3389/fimmu.2022.928173] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/07/2022] [Indexed: 12/13/2022] Open
Abstract
Transplantation is often the last resort for end-stage organ failures, e.g., kidney, liver, heart, lung, and pancreas. The shortage of donor organs is the main limiting factor for successful transplantation in humans. Except living donations, other alternatives are needed, e.g., xenotransplantation of pig organs. However, immune rejection remains the major challenge to overcome in xenotransplantation. There are three different xenogeneic types of rejections, based on the responses and mechanisms involved. It includes hyperacute rejection (HAR), delayed xenograft rejection (DXR) and chronic rejection. DXR, sometimes involves acute humoral xenograft rejection (AHR) and cellular xenograft rejection (CXR), which cannot be strictly distinguished from each other in pathological process. In this review, we comprehensively discussed the mechanism of these immunological rejections and summarized the strategies for preventing them, such as generation of gene knock out donors by different genome editing tools and the use of immunosuppressive regimens. We also addressed organ-specific barriers and challenges needed to pave the way for clinical xenotransplantation. Taken together, this information will benefit the current immunological research in the field of xenotransplantation.
Collapse
Affiliation(s)
- Qiao Zhou
- Department of Rheumatology and Immunology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Ting Li
- Department of Rheumatology, Wenjiang District People’s Hospital, Chengdu, China
| | - Kaiwen Wang
- School of Medicine, Faculty of Medicine and Health, The University of Leeds, Leeds, United Kingdom
| | - Qi Zhang
- School of Medicine, University of Electronics and Technology of China, Chengdu, China
| | - Zhuowen Geng
- School of Medicine, Faculty of Medicine and Health, The University of Leeds, Leeds, United Kingdom
| | - Shaoping Deng
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
- Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, China
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at The Ohio State University, Columbus, OH, United States
- *Correspondence: Chunming Cheng, ; Yi Wang,
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, China
- *Correspondence: Chunming Cheng, ; Yi Wang,
| |
Collapse
|
16
|
Tholen S, Patel R, Agas A, Kovary KM, Rabiee A, Nicholls HT, Bielczyk-Maczyńska E, Yang W, Kraemer FB, Teruel MN. Flattening of circadian glucocorticoid oscillations drives acute hyperinsulinemia and adipocyte hypertrophy. Cell Rep 2022; 39:111018. [PMID: 35767959 PMCID: PMC9391061 DOI: 10.1016/j.celrep.2022.111018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 04/14/2022] [Accepted: 06/08/2022] [Indexed: 11/03/2022] Open
Abstract
Disruption of circadian glucocorticoid oscillations in Cushing's disease and chronic stress results in obesity and adipocyte hypertrophy, which is believed to be a main source of the harmful effects of obesity. Here, we recapitulate stress due to jet lag or work-life imbalances by flattening glucocorticoid oscillations in mice. Within 3 days, mice achieve a metabolic state with persistently high insulin, but surprisingly low glucose and fatty acids in the bloodstream, that precedes a more than 2-fold increase in brown and white adipose tissue mass within 3 weeks. Transcriptomic and Cd36-knockout mouse analyses show that hyperinsulinemia-mediated de novo fatty acid synthesis and Cd36-mediated fatty acid uptake drive fat mass increases. Intriguingly, this mechanism by which glucocorticoid flattening causes acute hyperinsulinemia and adipocyte hypertrophy is unexpectedly beneficial in preventing high levels of circulating fatty acids and glucose for weeks, thus serving as a protective response to preserve metabolic health during chronic stress.
Collapse
Affiliation(s)
- Stefan Tholen
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Roma Patel
- Department of Biochemistry and the Gale and Ira Drukier Institute of Children's Health, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Agnieszka Agas
- Department of Biochemistry and the Gale and Ira Drukier Institute of Children's Health, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Kyle M Kovary
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Atefeh Rabiee
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Hayley T Nicholls
- Weill Center for Metabolic Health, Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Ewa Bielczyk-Maczyńska
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Wenting Yang
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Fredric B Kraemer
- Department of Medicine, Division of Endocrinology, Stanford University, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA 94305, USA
| | - Mary N Teruel
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Biochemistry and the Gale and Ira Drukier Institute of Children's Health, Weill Cornell Medical College of Cornell University, New York, NY, USA; Weill Center for Metabolic Health, Division of Endocrinology, Diabetes and Metabolism, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA.
| |
Collapse
|
17
|
Ammirati E, Bizzi E, Veronese G, Groh M, Van de Heyning CM, Lehtonen J, Pineton de Chambrun M, Cereda A, Picchi C, Trotta L, Moslehi JJ, Brucato A. Immunomodulating Therapies in Acute Myocarditis and Recurrent/Acute Pericarditis. Front Med (Lausanne) 2022; 9:838564. [PMID: 35350578 PMCID: PMC8958011 DOI: 10.3389/fmed.2022.838564] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/28/2022] [Indexed: 12/15/2022] Open
Abstract
The field of inflammatory disease of the heart or "cardio-immunology" is rapidly evolving due to the wider use of non-invasive diagnostic tools able to detect and monitor myocardial inflammation. In acute myocarditis, recent data on the use of immunomodulating therapies have been reported both in the setting of systemic autoimmune disorders and in the setting of isolated forms, especially in patients with specific histology (e.g., eosinophilic myocarditis) or with an arrhythmicburden. A role for immunosuppressive therapies has been also shown in severe cases of coronavirus disease 2019 (COVID-19), a condition that can be associated with cardiac injury and acute myocarditis. Furthermore, ongoing clinical trials are assessing the role of high dosage methylprednisolone in the context of acute myocarditis complicated by heart failure or fulminant presentation or the role of anakinra to treat patients with acute myocarditis excluding patients with hemodynamically unstable conditions. In addition, the explosion of immune-mediated therapies in oncology has introduced new pathophysiological entities, such as immune-checkpoint inhibitor-associated myocarditis and new basic research models to understand the interaction between the cardiac and immune systems. Here we provide a broad overview of evolving areas in cardio-immunology. We summarize the use of new imaging tools in combination with endomyocardial biopsy and laboratory parameters such as high sensitivity troponin to monitor the response to immunomodulating therapies based on recent evidence and clinical experience. Concerning pericarditis, the normal composition of pericardial fluid has been recently elucidated, allowing to assess the actual presence of inflammation; indeed, normal pericardial fluid is rich in nucleated cells, protein, albumin, LDH, at levels consistent with inflammatory exudates in other biological fluids. Importantly, recent findings showed how innate immunity plays a pivotal role in the pathogenesis of recurrent pericarditis with raised C-reactive protein, with inflammasome and IL-1 overproduction as drivers for systemic inflammatory response. In the era of tailored medicine, anti-IL-1 agents such as anakinra and rilonacept have been demonstrated highly effective in patients with recurrent pericarditis associated with an inflammatory phenotype.
Collapse
Affiliation(s)
- Enrico Ammirati
- De Gasperis Cardio Center and Transplant Center, Niguarda Hospital, Milano, Italy
| | - Emanuele Bizzi
- Internal Medicine, Fatebenefratelli Hospital, Milano, Italy
| | - Giacomo Veronese
- Department of Health Sciences, University of Milano-Bicocca, Monza, Italy
| | - Matthieu Groh
- National Reference Center for Hypereosinophilic Syndromes, CEREO, Suresnes, France
- Department of Internal Medicine, Hôpital Foch, Suresnes, France
| | - Caroline M. Van de Heyning
- Department of Cardiology, Antwerp University Hospital, and GENCOR Research Group, Antwerp University, Antwerp, Belgium
| | - Jukka Lehtonen
- Department of Cardiology, Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Marc Pineton de Chambrun
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (APHP), Hôpital La Pitié-Salpêtrière, Service de Médecine Intensive-Réanimation, Paris, France
- Sorbonne Université, APHP, Hôpital de la Pitié-Salpêtrière, Service de Médecine Interne 2, Centre de Référence National Lupus et SAPL et Autres Maladies Auto-immunes et Systémiques Rares, Paris, France
- Sorbonne Université, INSERM, UMRS_1166-ICAN, ICAN, Paris, France
| | - Alberto Cereda
- Cardiovascular Department, Association Socio Sanitary Territorial Santi Paolo e Carlo, Milano, Italy
| | - Chiara Picchi
- Internal Medicine, Fatebenefratelli Hospital, Milano, Italy
| | - Lucia Trotta
- Internal Medicine, Fatebenefratelli Hospital, Milano, Italy
| | - Javid J. Moslehi
- Section of Cardio-Oncology and Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Antonio Brucato
- Internal Medicine, Fatebenefratelli Hospital, Milano, Italy
- Department of Biomedical and Clinical Sciences “Luigi Sacco, ” Fatebenefratelli Hospital, University of Milano, Milano, Italy
| |
Collapse
|
18
|
Ul-Abideen Z, Ahmad HI, Nadeem M, Khan AA, Imran M, Majeed T, Jiang S, Elokil A. The therapeutic effect of bromocriptine as mesylate and estradiol valerate on serum and blood biochemistry of common quails. Poult Sci 2022; 101:101552. [PMID: 34942520 PMCID: PMC8704486 DOI: 10.1016/j.psj.2021.101552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/25/2021] [Accepted: 10/10/2021] [Indexed: 10/31/2022] Open
Abstract
Hematology and serum biochemistry study may provide antique knowledge about the physical status of individuals, making them a valuable tool to differentiate healthy animals from affected animals. We aimed to investigate Steroid safety levels in birds through ex-situ studies at regular therapeutic doses. A total of 100 birds were used for hematology and serum biochemistry. This study was designed into 2 trials over the summer and winter, each comprised 5, 10, 15, and 20 d. Each study group was based on 5 control group birds and 20 experimental group birds. A sum of 2 groups representing 2 different steroids trial groups was treated with therapeutic doses to the stretch of 5, 10, 15, and 20 d each season. A therapeutic dose of each of the steroids was given at the rate of 3 drops 2 times a day to each bird. Analysis of data reveals that steroids had severe effects on bird's (Coturnix coturnix) hematological parameters. In most trials, the hematological effects of bromocriptine as mesylate showed an increase in red blood cell count and white blood cell count. On the other hand, steroid estradiol valerate showed a decrease in these parameters. Effect of steroids on serum biochemistry profile indicate acute damage to vital organs, especially to liver and kidney, indicating an increase in cholesterol, total protein, albumin, urea, and uric acid. The overall effect of steroids on the bird's serum and biochemistry of quails were nearly similar but different only in their intensity.
Collapse
Affiliation(s)
| | - Hafiz Ishfaq Ahmad
- Department of Animal Breeding and Genetics, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | | | - Aleem Ahmad Khan
- Department of Zoology, Bahauddin Zakariya University, Multan, Pakistan
| | - Muhammad Imran
- Department of Veterinary Surgery and Pet Science, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Tanveer Majeed
- Department of Biotechnology, Kinnaird College for Women, Lahore, Pakistan
| | - Shouqun Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P. R. China.
| | - Abdelmotaleb Elokil
- Animal Production Department, Faculty of Agriculture, Benha University, Moshtohor 13736, Egypt
| |
Collapse
|
19
|
Bizzi E, Picchi C, Mastrangelo G, Imazio M, Brucato A. Recent advances in pericarditis. Eur J Intern Med 2022; 95:24-31. [PMID: 34556390 DOI: 10.1016/j.ejim.2021.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/27/2021] [Accepted: 09/04/2021] [Indexed: 12/15/2022]
Abstract
Pericardial diseases are an heterogeneous group of entities, ranging from acute pericarditis to asymptomatic pericardial effusions. New advances in understanding the processes underlying them have been made. In 2020 a prospective study defined the reference intervals of the component of normal pericardial fluid, that was found to be rich in nucleated cells, proteins, albumin and LDH, at levels compatible with the inflammatory exudates of other biological fluids such as pleural or peritoneal fluid; Light's criteria should not be used to evaluate it. Recently we also analyzed systematically large chronic idiopathic non-inflammatory pericardial effusions, observing that a non-invasive wait-and-see approach may be the best choice in clinical practice in oligosymptomatic cases. Concerning acute recurrent pericarditis (RP), an innovative interaction between cardiologists, internists and pediatric rheumatologists led to the intuition of a pivotal role of IL-1 in recurrent pericarditis characterized by an evident inflammatory recurrent phenotype, and recent data have shown the striking efficacy of anakinra and rilonacept in these patients. The proper selection of the patient is important; the ideal candidate for anti-IL-1 therapy is the patient with RP with high levels of serum C-reactive protein, high fever, neutrophil leukocitosis, pleuropulmonary involvement, frequent exacerbations and resistant to conventional therapy. On the contrary, anti-IL-1 drugs are not indicated in patients with pericardial effusion whose cause is not attributable to inflammatory phenomena. Finally, many patients with RP are women of childbearing age, and the possibility for these women to become pregnant must be addressed by multidisciplinary teams.
Collapse
Affiliation(s)
- Emanuele Bizzi
- Internal Medicine Department, Fatebenefratelli Hospital, Piazzale Principessa Clotilde 3, 20121, Milano, Italy.
| | - Chiara Picchi
- Internal Medicine Department, Fatebenefratelli Hospital, Piazzale Principessa Clotilde 3, 20121, Milano, Italy.
| | - Greta Mastrangelo
- Department of Pediatrics, Fatebenefratelli Hospital, Piazzale Principessa Clotilde 3, 20121, Milano, Italy.
| | - Massimo Imazio
- Cardiology, Cardiothoracic Department, University Hospital Santa Maria della Misericordia, Piazzale Santa Maria della Misericordia, 15, 33100, Udine, Italy
| | - Antonio Brucato
- University of Milano, Department of biomedical and clinical sciences "Luigi Sacco", Fatebenefratelli Hospital, Piazzale Principessa Clotilde 3, 20121, Milano, Italy.
| |
Collapse
|
20
|
Cai Z, Wang S, Li J. Treatment of Inflammatory Bowel Disease: A Comprehensive Review. Front Med (Lausanne) 2021; 8:765474. [PMID: 34988090 PMCID: PMC8720971 DOI: 10.3389/fmed.2021.765474] [Citation(s) in RCA: 255] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD), as a global disease, has attracted much research interest. Constant research has led to a better understanding of the disease condition and further promoted its management. We here reviewed the conventional and the novel drugs and therapies, as well as the potential ones, which have shown promise in preclinical studies and are likely to be effective future therapies. The conventional treatments aim at controlling symptoms through pharmacotherapy, including aminosalicylates, corticosteroids, immunomodulators, and biologics, with other general measures and/or surgical resection if necessary. However, a considerable fraction of patients do not respond to available treatments or lose response, which calls for new therapeutic strategies. Diverse therapeutic options are emerging, involving small molecules, apheresis therapy, improved intestinal microecology, cell therapy, and exosome therapy. In addition, patient education partly upgrades the efficacy of IBD treatment. Recent advances in the management of IBD have led to a paradigm shift in the treatment goals, from targeting symptom-free daily life to shooting for mucosal healing. In this review, the latest progress in IBD treatment is summarized to understand the advantages, pitfalls, and research prospects of different drugs and therapies and to provide a basis for the clinical decision and further research of IBD.
Collapse
Affiliation(s)
- Zhaobei Cai
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
- Department of Gastroenterology and Hepatology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Shu Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Wu S, Hassan FU, Luo Y, Fatima I, Ahmed I, Ihsan A, Safdar W, Liu Q, Rehman SU. Comparative Genomic Characterization of Buffalo Fibronectin Type III Domain Proteins: Exploring the Novel Role of FNDC5/Irisin as a Ligand of Gonadal Receptors. BIOLOGY 2021; 10:1207. [PMID: 34827201 PMCID: PMC8615036 DOI: 10.3390/biology10111207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/29/2022]
Abstract
FN-III proteins are widely distributed in mammals and are usually involved in cellular growth, differentiation, and adhesion. The FNDC5/irisin regulates energy metabolism and is present in different tissues (liver, brain, etc.). The present study aimed to investigate the physiochemical characteristics and the evolution of FN-III proteins and FNDC5/irisin as a ligand targeting the gonadal receptors including androgen (AR), DDB1 and CUL4 associated factor 6 (DCAF6), estrogen-related receptor β (ERR-β), estrogen-related receptor γ (ERR-γ), Krüppel-like factor 15 (KLF15), and nuclear receptor subfamily 3 group C member 1 (NR3C1). Moreover, the putative role of irisin in folliculogenesis and spermatogenesis was also elucidated. We presented the molecular structure and function of 29 FN-III genes widely distributed in the buffalo genome. Phylogenetic analysis, motif, and conserved domain pattern demonstrated the evolutionary well-conserved nature of FN-III proteins with a variety of stable to unstable, hydrophobic to hydrophilic, and thermostable to thermo-unstable properties. The comparative structural configuration of FNDC5 revealed amino acid variations but still the FNDC5 structure of humans, buffalo, and cattle was quite similar to each other. For the first time, we predicted the binding scores and interface residues of FNDC5/irisin as a ligand for six representative receptors having a functional role in energy homeostasis, and a significant involvement in folliculogenesis and spermatogenesis in buffalo.
Collapse
Affiliation(s)
- Siwen Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530005, China; (S.W.); (Y.L.)
| | - Faiz-ul Hassan
- Institute of Animal and Dairy Sciences, University of Agriculture, Faisalabad 38040, Pakistan;
| | - Yuhong Luo
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530005, China; (S.W.); (Y.L.)
| | - Israr Fatima
- Department of Bioinformatics and Biotechnology, Govt. College University, Faisalabad 38000, Pakistan;
| | - Ishtiaq Ahmed
- School of Medical Science, Gold Coast Campus, Griffith University, Southport, QLD 4222, Australia;
| | - Awais Ihsan
- Department of Biosciences, COMSATS University Islamabad, Sahiwal Campus, Sahiwal 57000, Pakistan;
| | - Warda Safdar
- Department of Biochemistry, Bahauddin Zakariya University, Multan 60000, Pakistan;
| | - Qingyou Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530005, China; (S.W.); (Y.L.)
| | - Saif ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530005, China; (S.W.); (Y.L.)
| |
Collapse
|
22
|
Tsukayama I, Mega T, Hojo N, Toda K, Kawakami Y, Takahashi Y, Suzuki-Yamamoto T. Diosgenin suppresses COX-2 and mPGES-1 via GR and improves LPS-induced liver injury in mouse. Prostaglandins Other Lipid Mediat 2021; 156:106580. [PMID: 34252545 DOI: 10.1016/j.prostaglandins.2021.106580] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/28/2022]
Abstract
Using a wild yam (Dioscorea japonica), we previously found novel anti-inflammatory and anti-carcinogenic effects via the downregulation of cyclooxygenase (COX)-2 and microsomal prostaglandin E synthase (mPGES)-1. One of the substances in wild yam is a steroidal saponin, diosgenin. We demonstrated that diosgenin suppressed COX-2 in human non-small-cell lung carcinoma A549 cells via nuclear factor-kappa B (NF-κB) translocation and the effects were reversed by a glucocorticoid receptor antagonist, RU486. In lipopolysaccharide (LPS)-induced mouse liver injury, COX-2 and mPGES-1 were induced and localized in sinusoidal macrophages and endothelial cells; however, diosgenin administration significantly suppressed Ptgs2 and Ptges expression and decreased COX-2 and mPGES-1 immunopositive cells in the sinusoids. Multiple immunohistochemical analyses showed that diosgenin had an effect on COX-2 and mPGES-1, particularly in the macrophages. Thus, we showed that diosgenin downregulated COX-2 and mPGES-1 via the glucocorticoid receptor and suppressed COX-2 and mPGES-1 in the macrophages of LPS-induced acute mouse liver injury.
Collapse
Affiliation(s)
- Izumi Tsukayama
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Takuto Mega
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Nana Hojo
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Keisuke Toda
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Yuki Kawakami
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Yoshitaka Takahashi
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan
| | - Toshiko Suzuki-Yamamoto
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama, 719-1197, Japan.
| |
Collapse
|
23
|
Wang CC, Du L, Shi HH, Ding L, Yanagita T, Xue CH, Wang YM, Zhang TT. Dietary EPA-Enriched Phospholipids Alleviate Chronic Stress and LPS-Induced Depression- and Anxiety-Like Behavior by Regulating Immunity and Neuroinflammation. Mol Nutr Food Res 2021; 65:e2100009. [PMID: 34219360 DOI: 10.1002/mnfr.202100009] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/10/2021] [Indexed: 12/17/2022]
Abstract
SCOPE A growing number of studies have reported the effects of eicosapentaenoic acid (EPA) and terrestrial phospholipids on ameliorating mood disorders. Marine-derived EPA-enriched phospholipids (EPA-PL) exhibit the structural characteristics of EPA and phospholipids. However, the effect of dietary EPA-PL, and the differences between amphiphilic EPA-PL and lyophobic EPA on mood disorders had not been studied. METHODS AND RESULTS A comparative investigation to determine the effects of dietary EPA-enriched ethyl ester (EPA-EE) and EPA-PL on improving depression- and anxiety-like behavior in a mouse model is performed, induced by 4 week chronic unpredictable mild stress (CUMS) coupled with lipopolysaccharide (LPS) challenge. It is found that dietary 4 week 0.6% (w/w) EPA-PL rescued depression- and anxiety-like behavior to a greater extent than did EPA-EE. Moreover, dietary EPA-PL significantly reduced the immobility time by 56.6%, close to the normal level, in forced swimming test, which revealed a reversal of depression-like behavior. Further studies revealed that dietary EPA-PL regulated immunity, monoamine systems, and the hypothalamic-pituitary-adrenal (HPA) axis by multi-target interactions, including inhibition of neuroinflammation and apoptosis. CONCLUSION EPA-PL exerted superior effects to EPA-EE in alleviating depression- and anxiety-like behavior. The data suggest potential novel candidate or targeted dietary patterns to prevent and treat mood disorder.
Collapse
Affiliation(s)
- Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Lei Du
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, No. 44 Wenhuaxi Road, Jinan, Shandong, 250012, P. R. China
| | - Hao-Hao Shi
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Lin Ding
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
| | - Teruyoshi Yanagita
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong Province, 266237, P. R. China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
- Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science, Saga University, Saga, 840-8502, Japan
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
- Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science, Saga University, Saga, 840-8502, Japan
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, P. R. China
| |
Collapse
|
24
|
Húngaro TGR, Gregnani MF, Alves-Silva T, Herse F, Alenina N, Bader M, Araújo RC. Cortisol Dose-Dependently Impairs Migration and Tube-like Formation in a Trophoblast Cell Line and Modulates Inflammatory and Angiogenic Genes. Biomedicines 2021; 9:980. [PMID: 34440184 PMCID: PMC8393357 DOI: 10.3390/biomedicines9080980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/17/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022] Open
Abstract
Several stimuli can change maternal hormone levels during pregnancy. These changes may affect trophoblastic cells and modulate the development of the embryo and the placental tissue itself. Changes in cortisol levels are associated with impaired trophoblast implantation and function, in addition to other pregnancy complications. This study aims to analyze the effects of low and high doses of cortisol on an extravillous trophoblast cell line, and the effects of various exposures to this hormone. SGHPL-4 cells were treated with cortisol at five doses (0-1000 nM) and two exposures (continuous: 24 h/day; and intermittent: 2 h/day). In intermittent treatment, cortisol acted mainly as an anti-inflammatory hormone, repressing gene expression of kinin B1 receptors, interleukin-6, and interleukin-1β. Continuous treatment modulated inflammatory and angiogenic pathways, significantly repressing angiogenic factors and their receptors. Cortisol affected cell migration and tube-like structures formation. In conclusion, both continuous and intermittent exposure to cortisol repressed the expression of inflammatory genes, while only continuous exposure repressed the expression of angiogenic genes, suggesting that a sustained increase in the levels of this hormone is more harmful than a high short-term increase. Cortisol also impaired tube-like structures formation, and kinin receptors may be involved in this response.
Collapse
Affiliation(s)
- Talita Guerreiro Rodrigues Húngaro
- Nephrology Program, Laboratory of Genetics and Exercise Metabolism, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil;
| | - Marcos F. Gregnani
- Molecular Biology Program, Laboratory of Genetics and Exercise Metabolism, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil; (M.F.G.); (T.A.-S.)
| | - Thaís Alves-Silva
- Molecular Biology Program, Laboratory of Genetics and Exercise Metabolism, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil; (M.F.G.); (T.A.-S.)
| | - Florian Herse
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; (F.H.); (N.A.)
- Experimental and Clinical Research Center (ECRC), a Cooperation of Charité—Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine (MDC), Lindenberger Weg 80, 13125 Berlin, Germany
- Berlin Institute of Health, 10178 Berlin, Germany
| | - Natalia Alenina
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; (F.H.); (N.A.)
- Berlin Institute of Health, 10178 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10117 Berlin, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; (F.H.); (N.A.)
- Berlin Institute of Health, 10178 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10117 Berlin, Germany
- Max Delbrück Center of Molecular Medicine, Charité University Medicine, Charitéplatz 1, 10117 Berlin, Germany
- Institute for Biology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Ronaldo C. Araújo
- Nephrology Program, Laboratory of Genetics and Exercise Metabolism, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil;
- Molecular Biology Program, Laboratory of Genetics and Exercise Metabolism, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil; (M.F.G.); (T.A.-S.)
| |
Collapse
|
25
|
Bizzi E, Trotta L, Pancrazi M, Nivuori M, Giosia V, Matteucci L, Montori D, Brucato A. Autoimmune and Autoinflammatory Pericarditis: Definitions and New Treatments. Curr Cardiol Rep 2021; 23:128. [PMID: 34319478 DOI: 10.1007/s11886-021-01549-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE OF THE REVIEW The purpose of the review is to analyze the pathogenetic mechanisms that underlie acute pericarditis, with attention to autoimmune and autoinflammatory pericarditis, and, in addition, to review the available therapeutic armamentarium. RECENT FINDINGS Several studies have been published on the use of anti-IL-1 drugs in recurrent pericarditis, including anakinra and rilonacept. The latest, the RHAPSODY study, based on the use of rilonacept in recurrent pericarditis, has recently reached phase 3 with promising results in terms of efficacy and safety. Alterations in the function of the inflammasome and the consequent overproduction of IL-1 play a pivotal role in the genesis of autoinflammatory pericarditis. Recent studies added evidence to the importance of anti-IL-1 drugs in the treatment of recurrent pericarditis with raised C-reactive protein. In the era of tailored medicine, anti-IL-1 agents may be very useful in the subset of patients with recurrent pericarditis and a clear inflammatory phenotype.
Collapse
Affiliation(s)
- Emanuele Bizzi
- Internal Medicine, Fatebefratelli Hospital, Piazzale Principessa Clotilde 3, Milan, Italy.
| | - Lucia Trotta
- Internal Medicine, Fatebefratelli Hospital, Piazzale Principessa Clotilde 3, Milan, Italy
| | - Massimo Pancrazi
- Internal Medicine, Fatebefratelli Hospital, Piazzale Principessa Clotilde 3, Milan, Italy
| | - Mariangela Nivuori
- Internal Medicine, Fatebefratelli Hospital, Piazzale Principessa Clotilde 3, Milan, Italy
| | - Valeria Giosia
- Internal Medicine, Fatebefratelli Hospital, Piazzale Principessa Clotilde 3, Milan, Italy
| | - Luca Matteucci
- Internal Medicine, Fatebefratelli Hospital, Piazzale Principessa Clotilde 3, Milan, Italy
| | - Daniela Montori
- Internal Medicine, Fatebefratelli Hospital, Piazzale Principessa Clotilde 3, Milan, Italy
| | - Antonio Brucato
- Department of Biomedical and Clinical Sciences, Fatebenefratelli Hospital, Università di Milano, Milan, Italy
| |
Collapse
|
26
|
Glucocorticoids Improve Enteral Feeding Tolerance in Pediatric Short Bowel Syndrome With Chronic Intestinal Inflammation. J Pediatr Gastroenterol Nutr 2021; 73:17-22. [PMID: 33534364 DOI: 10.1097/mpg.0000000000003058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVES A group of short bowel syndrome (SBS) patients developed chronic intestinal inflammation while struggling weaning off parenteral nutrition (PN). They did not respond to standard management of SBS and food allergy. We treated them with glucocorticoids and described the outcome. METHODS Our study is a retrospective descriptive study. We reviewed records from the intestinal rehabilitation program from 2006 to 2017. We identified 15 patients whose lab values, pathology results, and clinic notes were reviewed. RESULTS We had more patients (n = 10) with diagnosis of gastroschisis, and more female patients (n = 9). Seven patients weaned off PN with median treatment duration of 5 months, 5 of which remained on budesonide for significant period of time (median: 7.5 months). One of these 7 patients relapsed, as the patient resumed glucocorticoids because of recurrence of chronic intestinal inflammation. Six of 15 children had significant eosinophils in their initial biopsy, 5 of these children weaned off PN whereas 1 child's gastrointestinal (GI) bleeding stopped. Four patients were not able to decrease PN calorie. Two of these patients' GI bleeding stopped, the other 2 had normalized histology. CONCLUSIONS For SBS children with histologically confirmed chronic intestinal inflammation, glucocorticoids may help promote enteral feeding tolerance. Glucocorticoids regimen should be chosen individually. Patients are more likely to respond if initial histology has significant eosinophilic infiltration. Patients may need to remain on glucocorticoids for over 6 months.
Collapse
|
27
|
Lin P, Fu S, Li W, Hu Y, Liang Z. Inhaled corticosteroids and risk of lung cancer: A systematic review and meta-analysis. Eur J Clin Invest 2021; 51:e13434. [PMID: 33053199 DOI: 10.1111/eci.13434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/09/2020] [Accepted: 09/21/2020] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Current studies investigating the association between inhaled corticosteroid (ICS) use and risk of lung cancer have yielded inconsistent findings. The aim of this systematic review and meta-analysis was to pool all currently available data to estimate this association. METHODS We systematically searched MEDLINE (1946 to July 2020), EMBASE (1974 to July 2020) and the Cochrane Library (June 2020) via Ovid to identify relevant articles investigating the association between the ICS use and the risk of lung cancer. Random-effects analysis was used to calculate pooled relative risks (RRs) with 95% confidence intervals (CIs). RESULTS Ten articles including 234 920 patients were analysed. ICS use was identified to have a decreased risk of lung cancer in chronic obstructive pulmonary disease (8 studies, 1806 patients; RR = 0.73, 95% CI: 0.61-0.87, P < .01; I2 = 60.0 %), asthma (1 study, 41 438 patients; RR = 0.44, 95% CI: 0.34-0.57, P < .01) and mixed (1 study, 46 225 patients; RR = 0.79, 95% CI: 0.69-0.90, P < .01) patients. The findings of reduced risk of lung cancer were consistent in all subgroup analyses except for the short-term follow-up (≤5 years) (RR = 0.94, 95% CI: 0.81-1.07, P = .34) and free of immortal time bias (RR = 0.94, 95% CI: 0.82-1.08, P = .38) subgroups. CONCLUSIONS The present study suggested that ICS use was associated with decreased risk of lung cancer. However, our findings should be interpreted with caution because most original studies were judged to be at high risk of immortal time bias.
Collapse
Affiliation(s)
- Ping Lin
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Siyu Fu
- Center of Infectious Diseases, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Weijing Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuehong Hu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zongan Liang
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Zozo B, Govender N, Moodley J, Naicker T. Expression of plasma nuclear factor-kappa B cells (NF-κB) and Inhibitory subunit kappa B alpha (IκB-α) in HIV-associated pre-eclampsia. Hypertens Pregnancy 2021; 40:15-20. [PMID: 33397161 DOI: 10.1080/10641955.2020.1854299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Objective:To investigate the expression of plasma NF-κB and the inhibitory subunit IκB-α in HIV-associated pre-eclampsia. Method: This retrospective study examined plasma NF-κB and IκB-α expression expressed as Mean Fluorescence Intensity (MFI) in normotensive (n = 32) and pre-eclamptic (n = 34) pregnant women stratified by HIV status. Results: A significant decrease in the level of plasma NF-κB expression between pre-eclamptic and normotensive pregnant women was observed, irrespective of HIV status (MFI = 82.8 vs 134.3; p = 0.03). Similarly, a significant decline in the level of plasma IκB-α expression was noted in the pre-eclamptic compared to normotensive pregnant women, irrespective of HIV status (MFI = 91.2 vs 120.1; p = 0.04). Plasma NF-κB (p = 0.7) and IκB-α (p = 0.8) expression was similar between HIV positive and HIV negative pregnant women, irrespective of pregnancy type. Conclusion: This study demonstrates decreased plasma NF-κB and IκB-α expression in preeclampsia compared to normotensive pregnant women irrespective of HIV status.
Collapse
Affiliation(s)
- Bambanani Zozo
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal , Durban, KwaZulu-Natal, South Africa
| | - Nalini Govender
- Women's Health and HIV Research Group, College of Health Sciences, University of KwaZulu-Natal , Durban, South Africa
| | - Jagidesa Moodley
- Dept of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology , Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal , Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
29
|
Shanker S, Saroj N, Cordova EJ, Jarillo-Luna RA, López-Sánchez P, Terrón JA. Chronic restraint stress induces serotonin transporter expression in the rat adrenal glands. Mol Cell Endocrinol 2020; 518:110935. [PMID: 32659440 DOI: 10.1016/j.mce.2020.110935] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
Chronic restraint stress (CRS) magnifies restraint-induced corticosterone secretion through a mechanism involving increased adrenocortical 5-HT content and turnover. We analysed the impact of CRS on serotonin transporter (SERT) expression and distribution in rat adrenal glands. Male Wistar rats were submitted to CRS (20 min/day) or undisturbed control conditions for 14 days. Exposure to CRS induced a remarkable increase in SERT-like immunoreactivity in the adrenal cortex, which closely matched that of chromogranin A immunostaining, along with a significant increase in SERT protein and mRNA levels in whole adrenals as determined by immunohistochemistry, Western blot and RT-PCR assays, respectively; all these CRS-induced changes occurred almost exclusively in left adrenals. Closely similar results were obtained in animals that received a 14-day chronic corticosterone treatment. These results unravel an interesting association between chronic stress exposure and SERT expression in adrenocortical chromogranin A-positive cells, which seems to be a glucocorticoid-dependent phenomenon.
Collapse
Affiliation(s)
- Shiv Shanker
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina-IPN, Plan de San Luis y Díaz Mirón s/n, Casco de Sto. Tomás, CP 11340, CDMX, Mexico
| | - Neeshu Saroj
- Departamento de Farmacología, CINVESTAV-IPN, Av. Instituto Politécnico Nacional 2508, col. La Laguna Ticomán, CP 07360, CDMX, Mexico
| | - Emilio J Cordova
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica, Periférico Sur 4809, col. Arenal Tepepan, CP 14610, CDMX, Mexico
| | - Rosa A Jarillo-Luna
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina-IPN, Plan de San Luis y Díaz Mirón s/n, Casco de Sto. Tomás, CP 11340, CDMX, Mexico
| | - Pedro López-Sánchez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina-IPN, Plan de San Luis y Díaz Mirón s/n, Casco de Sto. Tomás, CP 11340, CDMX, Mexico
| | - José A Terrón
- Departamento de Farmacología, CINVESTAV-IPN, Av. Instituto Politécnico Nacional 2508, col. La Laguna Ticomán, CP 07360, CDMX, Mexico.
| |
Collapse
|
30
|
Hajjo R, Sabbah DA, Bardaweel SK. Chemocentric Informatics Analysis: Dexamethasone Versus Combination Therapy for COVID-19. ACS OMEGA 2020; 5:29765-29779. [PMID: 33251412 PMCID: PMC7689662 DOI: 10.1021/acsomega.0c03597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/03/2020] [Indexed: 05/08/2023]
Abstract
COVID-19 is a biphasic infectious disease with no approved vaccine or pharmacotherapy. The first drug that has shown promise in reducing COVID-19 mortality in severely-ill patients is dexamethasone, a cheap, well-known anti-inflammatory glucocorticoid, approved for the treatment of inflammatory conditions including respiratory diseases such as asthma and tuberculosis. However, about 80% of COVID-19 patients requiring oxygenation, and about 67% of patients on ventilators, are not responsive to dexamethasone therapy mainly. Additionally, using higher doses of dexamethasone for prolonged periods of time can lead to severe side effects and some patients may develop corticosteroid resistance leading to treatment failure. In order to increase the therapeutic efficacy of dexamethasone in COVID-19 patients, while minimizing dexamethasone-related complications that could result from using higher doses of the drug, we applied a chemocentric informatics approach to identify combination therapies. Our results indicated that combining dexamethasone with fast long-acting beta-2 adrenergic agonists (LABAs), such as formoterol and salmeterol, can ease respiratory symptoms hastily, until dexamethasone's anti-inflammatory and immunosuppressant effects kick in. Our studies demonstrated that LABAs and dexamethasone (or other glucocorticoids) exert synergistic effects that will augment both anti-inflammatory and fibronectin-mediated anticoagulant effects. We also propose other alternatives to LABAs that are supported by sound systems biology evidence, such as nitric oxide. Other drugs such as sevoflurane and treprostinil interact with the SARS-CoV-2 interactome and deserve further exploration. Moreover, our chemocentric informatics approach provides systems biology evidence that combination therapies for COVID-19 will have higher chances of perturbing the SARS-CoV-2 human interactome, which may negatively impact COVID-19 disease pathways.
Collapse
Affiliation(s)
- Rima Hajjo
- Department
of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah
University of Jordan, P.O. Box 130, Amman 11733, Jordan
| | - Dima A. Sabbah
- Department
of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah
University of Jordan, P.O. Box 130, Amman 11733, Jordan
| | - Sanaa K. Bardaweel
- Department
of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| |
Collapse
|
31
|
McDonnell M, Harris RJ, Borca F, Mills T, Downey L, Dharmasiri S, Patel M, Zare B, Stammers M, Smith TR, Felwick R, Cummings JRF, Phan HTT, Gwiggner M. High incidence of glucocorticoid-induced hyperglycaemia in inflammatory bowel disease: metabolic and clinical predictors identified by machine learning. BMJ Open Gastroenterol 2020; 7:e000532. [PMID: 33187976 PMCID: PMC7668301 DOI: 10.1136/bmjgast-2020-000532] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/03/2020] [Accepted: 10/23/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Glucocorticosteroids (GC) are long-established, widely used agents for induction of remission in inflammatory bowel disease (IBD). Hyperglycaemia is a known complication of GC treatment with implications for morbidity and mortality. Published data on prevalence and risk factors for GC-induced hyperglycaemia in the IBD population are limited. We prospectively characterise this complication in our cohort, employing machine-learning methods to identify key predictors of risk. METHODS We conducted a prospective observational study of IBD patients receiving intravenous hydrocortisone (IVH). Electronically triggered three times daily capillary blood glucose (CBG) monitoring was recorded alongside diabetes mellitus (DM) history, IBD biomarkers, nutritional and IBD clinical activity scores. Hyperglycaemia was defined as CBG ≥11.1 mmol/L and undiagnosed DM as glycated haemoglobin ≥48 mmol/mol. Random forest (RF) regression models were used to extract predictor-patterns present within the dataset. RESULTS 94 consecutive IBD patients treated with IVH were included. 60% (56/94) of the cohort recorded an episode of hyperglycaemia, including 57% (50/88) of those with no history of DM, of which 19% (17/88) and 5% (4/88) recorded a CBG ≥14 mmol/L and ≥20 mmol/L, respectively. The RF models identified increased C-reactive protein (CRP) followed by a longer IBD duration as leading risk predictors for significant hyperglycaemia. CONCLUSION Hyperglycaemia is common in IBD patients treated with intravenous GC. Therefore, CBG monitoring should be included in routine clinical practice. Machine learning methods can identify key risk factors for clinical complications. Steroid-sparing treatment strategies may be considered for those IBD patients with higher admission CRP and greater disease duration, who appear to be at the greatest risk of hyperglycaemia.
Collapse
Affiliation(s)
- Martin McDonnell
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Human Health and Development, University of Southampton Faculty of Medicine, Southampton, UK
| | - Richard J Harris
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Florina Borca
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Clinical Informatics Research Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Tilly Mills
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Louise Downey
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Suranga Dharmasiri
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Mayank Patel
- Department of Diabetes and Endocrinology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Benjamin Zare
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Matt Stammers
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- NIHR Biomedical Research Facility, University of Southampton, Southampton, UK
| | - Trevor R Smith
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Richard Felwick
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - J R Fraser Cummings
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Human Health and Development, University of Southampton Faculty of Medicine, Southampton, UK
| | - Hang T T Phan
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Clinical Informatics Research Unit, University of Southampton Faculty of Medicine, Southampton, UK
| | - Markus Gwiggner
- Department of Gastroenterology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
32
|
Ide H, Miyamoto H. The Role of Steroid Hormone Receptors in Urothelial Tumorigenesis. Cancers (Basel) 2020; 12:cancers12082155. [PMID: 32759680 PMCID: PMC7465876 DOI: 10.3390/cancers12082155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/28/2020] [Accepted: 08/01/2020] [Indexed: 12/18/2022] Open
Abstract
Preclinical and/or clinical evidence has indicated a potential role of steroid hormone-mediated signaling pathways in the development of various neoplastic diseases, while precise mechanisms for the functions of specific receptors remain poorly understood. Specifically, in urothelial cancer where sex-related differences particularly in its incidence are noted, activation of sex hormone receptors, such as androgen receptor and estrogen receptor-β, has been associated with the induction of tumor development. More recently, glucocorticoid receptor has been implied to function as a suppressor of urothelial tumorigenesis. This article summarizes and discusses available data suggesting that steroid hormone receptors, including androgen receptor, estrogen receptor-α, estrogen receptor-β, glucocorticoid receptor, progesterone receptor and vitamin D receptor, as well as their related signals, contribute to modulating urothelial tumorigenesis.
Collapse
Affiliation(s)
- Hiroki Ide
- Department of Urology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Hiroshi Miyamoto
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Correspondence:
| |
Collapse
|
33
|
Darwitan A, Wong YS, Nguyen LTH, Czarny B, Vincent A, Nedumaran AM, Tan YF, Muktabar A, Tang JK, Ng KW, Venkatraman S. Liposomal Nanotherapy for Treatment of Atherosclerosis. Adv Healthc Mater 2020; 9:e2000465. [PMID: 32543010 DOI: 10.1002/adhm.202000465] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/21/2020] [Indexed: 02/02/2023]
Abstract
Atherosclerosis is a chronic disease that can lead to life-threatening events such as myocardial infarction and stroke, is characterized by the build-up of lipids and immune cells within the arterial wall. It is understood that inflammation is a hallmark of atherosclerosis and can be a target for therapy. In support of this concept, an injectable nanoliposomal formulation encapsulating fluocinolone acetonide (FA), a corticosteroid, is developed that allows for drug delivery to atherosclerotic plaques while reducing the systemic exposure to off-target tissues. In this study, FA is successfully incorporated into liposomal nanocarriers of around 100 nm in size with loading efficiency of 90% and the formulation exhibits sustained release up to 25 d. The anti-inflammatory effect and cholesterol efflux capability of FA-liposomes are demonstrated in vitro. In vivo studies carried out with an apolipoprotein E-knockout (Apoe-/- ) mouse model of atherosclerosis show accumulation of liposomes in atherosclerotic plaques, colocalization with plaque macrophages and anti-atherogenic effect over 3 weeks of treatment. This FA-liposomal-based nanocarrier represents a novel potent nanotherapeutic option for atherosclerosis.
Collapse
Affiliation(s)
- Anastasia Darwitan
- School of Materials Science & EngineeringNanyang Technological University Singapore 639798 Singapore
| | - Yee Shan Wong
- School of Materials Science & EngineeringNanyang Technological University Singapore 639798 Singapore
| | - Luong T. H. Nguyen
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State University Columbus OH 43210 USA
| | - Bertrand Czarny
- School of Materials Science & EngineeringNanyang Technological University Singapore 639798 Singapore
- Lee Kong Chian School of MedicineNanyang Technological University Singapore 636921 Singapore
| | - Anita Vincent
- School of Materials Science & EngineeringNanyang Technological University Singapore 639798 Singapore
| | - Anu Maashaa Nedumaran
- School of Materials Science & EngineeringNanyang Technological University Singapore 639798 Singapore
| | - Yang Fei Tan
- School of Materials Science & EngineeringNanyang Technological University Singapore 639798 Singapore
| | - Aristo Muktabar
- School of Materials Science & EngineeringNanyang Technological University Singapore 639798 Singapore
| | - Jin Kai Tang
- School of Materials Science & EngineeringNanyang Technological University Singapore 639798 Singapore
| | - Kee Woei Ng
- School of Materials Science & EngineeringNanyang Technological University Singapore 639798 Singapore
| | - Subbu Venkatraman
- Materials Science and EngineeringNational University of Singapore Singapore 117575 Singapore
| |
Collapse
|
34
|
A heretical view: rather than a solely placental protective function, placental 11β hydroxysteroid dehydrogenase 2 also provides substrate for fetal peripheral cortisol synthesis in obese pregnant ewes. J Dev Orig Health Dis 2020; 12:94-100. [PMID: 32151296 DOI: 10.1017/s2040174420000112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Exposure to glucocorticoid levels higher than appropriate for current developmental stages induces offspring metabolic dysfunction. Overfed/obese (OB) ewes and their fetuses display elevated blood cortisol, while fetal Adrenocorticotropic hormone (ACTH) remains unchanged. We hypothesized that OB pregnancies would show increased placental 11β hydroxysteroid dehydrogenase 2 (11β-HSD2) that converts maternal cortisol to fetal cortisone as it crosses the placenta and increased 11β-HSD system components responsible for peripheral tissue cortisol production, providing a mechanism for ACTH-independent increase in circulating fetal cortisol. Control ewes ate 100% National Research Council recommendations (CON) and OB ewes ate 150% CON diet from 60 days before conception until necropsy at day 135 gestation. At necropsy, maternal jugular and umbilical venous blood, fetal liver, perirenal fat, and cotyledonary tissues were harvested. Maternal plasma cortisol and fetal cortisol and cortisone were measured. Fetal liver, perirenal fat, cotyledonary 11β-HSD1, hexose-6-phosphate dehydrogenase (H6PD), and 11β-HSD2 protein abundance were determined by Western blot. Maternal plasma cortisol, fetal plasma cortisol, and cortisone were higher in OB vs. CON (p < 0.01). 11β-HSD2 protein was greater (p < 0.05) in OB cotyledonary tissue than CON. 11β-HSD1 abundance increased (p < 0.05) in OB vs. CON fetal liver and perirenal fat. Fetal H6PD, an 11β-HSD1 cofactor, also increased (p < 0.05) in OB vs. CON perirenal fat and tended to be elevated in OB liver (p < 0.10). Our data provide evidence for increased 11β-HSD system components responsible for peripheral tissue cortisol production in fetal liver and adipose tissue, thereby providing a mechanism for an ACTH-independent increase in circulating fetal cortisol in OB fetuses.
Collapse
|
35
|
Hu Y, Feng Y, Zhang L, Jia Y, Cai D, Qian SB, Du M, Zhao R. GR-mediated FTO transactivation induces lipid accumulation in hepatocytes via demethylation of m 6A on lipogenic mRNAs. RNA Biol 2020; 17:930-942. [PMID: 32116145 DOI: 10.1080/15476286.2020.1736868] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chronic stress or excessive exposure to glucocorticoids (GC) contributes to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Glucocorticoid receptor (GR) mediates the action of GC, but its downstream signalling is not fully understood. Fat mass and obesity associated (FTO) protein and its demethylation substrate N6-methyladenosine (m6A) are both reported to participate in the regulation of lipid metabolism, yet it remains unknown whether they are involved in GC-induced hepatic lipid accumulation as new components of GR signalling. In this study, we use both in vivo and in vitro models of GC-induced hepatic lipid accumulation and demonstrate that the activation of lipogenic genes and accumulation of lipid in liver cells are mediated by GR-dependent FTO transactivation and m6A demethylation on mRNA of lipogenic genes. Targeted mutation of m6A methylation sites and FTO knockdown further validated the role of m6A on 3'UTR of sterol regulatory element-binding transcription factor 1 and stearoyl-CoA desaturase mRNAs. Finally, FTO knockdown significantly alleviated dexamethasone-induced fatty liver in mice. These results demonstrate a role of GR-mediated FTO transactivation and m6A demethylation in the pathogenesis of NAFLD and provide new insight into GR signalling in the regulation of fat metablism in the liver.
Collapse
Affiliation(s)
- Yun Hu
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University , Nanjing, Jiangsu, China.,Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University , Nanjing, Jiangsu, China.,College of Animal Science and Technology, Yangzhou University , Yangzhou, China
| | - Yue Feng
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University , Nanjing, Jiangsu, China.,Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University , Nanjing, Jiangsu, China
| | - Luchu Zhang
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University , Nanjing, Jiangsu, China.,Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University , Nanjing, Jiangsu, China
| | - Yimin Jia
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University , Nanjing, Jiangsu, China.,Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University , Nanjing, Jiangsu, China
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University , Yangzhou, China
| | - Shu-Bing Qian
- Division of Nutritional Sciences, Cornell University , Ithaca, NY, USA
| | - Min Du
- Laboratory of Nutrigenomics and Growth Biology, Department of Animal Sciences, Washington State University , Pullman, WA, USA
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University , Nanjing, Jiangsu, China.,Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University , Nanjing, Jiangsu, China
| |
Collapse
|
36
|
Georgana I, Maluquer de Motes C. Cullin-5 Adaptor SPSB1 Controls NF-κB Activation Downstream of Multiple Signaling Pathways. Front Immunol 2020; 10:3121. [PMID: 32038638 PMCID: PMC6985365 DOI: 10.3389/fimmu.2019.03121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/20/2019] [Indexed: 01/03/2023] Open
Abstract
The initiation of innate immune responses against pathogens relies on the activation of pattern-recognition receptors (PRRs) and corresponding intracellular signaling cascades. To avoid inappropriate or excessive activation of PRRs, these responses are tightly controlled. Cullin-RING E3 ubiquitin ligases (CRLs) have emerged as critical regulators of many cellular functions including innate immune activation and inflammation. CRLs form multiprotein complexes in which a Cullin protein acts as a scaffold and recruits specific adaptor proteins, which in turn recognize specific substrate proteins for ubiquitylation, hence providing selectivity. CRLs are divided into 5 main groups, each of which uses a specific group of adaptor proteins. Here, we systematically depleted all predicted substrate adaptors for the CRL5 family (the so-called SOCS-box proteins) and assessed the impact on the activation of the inflammatory transcription factor NF-κB. Depletion of SPSB1 resulted in a significant increase in NF-κB activation, indicating the importance of SPSB1 as an NF-κB negative regulator. In agreement, overexpression of SPSB1 suppressed NF-κB activity in a potent, dose-dependent manner in response to various agonists. Inhibition by SPSB1 was specific to NF-κB, because other transcription factors related to innate immunity and interferon (IFN) responses such as IRF-3, AP-1, and STATs remained unaffected by SPSB1. SPSB1 suppressed NF-κB activation induced via multiple pathways including Toll-like receptors and RNA and DNA sensing adaptors, and required the presence of its SOCS-box domain. To provide mechanistic insight, we examined phosphorylation and degradation of the inhibitor of κB (IκBα) and p65 translocation into the nucleus. Both remained unaffected by SPSB1, indicating that SPSB1 exerts its inhibitory activity downstream, or at the level, of the NF-κB heterodimer. In agreement with this, SPSB1 was found to co-precipitate with p65 after over-expression and at endogenous levels. Additionally, A549 cells stably expressing SPSB1 presented lower cytokine levels including type I IFN in response to cytokine stimulation and virus infection. Taken together, our results reveal novel regulatory mechanisms in innate immune signaling and identify the prominent role of SPSB1 in limiting NF-κB activation. Our work thus provides insights into inflammation and inflammatory diseases and new opportunities for the therapeutic targeting of NF-κB transcriptional activity.
Collapse
|
37
|
Längericht J, Krämer I, Kahaly GJ. Glucocorticoids in Graves' orbitopathy: mechanisms of action and clinical application. Ther Adv Endocrinol Metab 2020; 11:2042018820958335. [PMID: 33403097 PMCID: PMC7745544 DOI: 10.1177/2042018820958335] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/20/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Graves' orbitopathy (GO) is the most frequent extrathyroidal manifestation of the autoimmune Graves' disease. GO significantly impacts quality of life and has a psycho-social morbidity. Inflammation and swelling of the orbital tissue often leads to proptosis, diplopia, and decrease of visual acuity. Due to the inflammatory background of the disease, glucocorticoids (GC) have been used as a first-line treatment for decades. METHODS PubMed and MeSH database were searched for original articles, clinical trials, reviews, and meta-analyses published between 1 January 2000 and 31 March 2020 and pertaining to both the mechanism of action and immunological effects of GC as well as to the treatment of GO by GC. The publications were evaluated according to their setting and study design. RESULTS GC act through genomic (trans-activation and trans-repression) and rapid non-genomic mechanisms. GC in general, and the intravenous (IV) administration of GC in particular, markedly decrease the activity and number of the most potent antigen-presenting dendritic cells. According to the internationally acknowledged European Thyroid Association Guidelines for the management of GO, weekly IVGC application over 12 weeks is recommended as first-line treatment for patients with active and severe GO. The daily and cumulative dose should be tailored according to clinical severity, for example, 4.5 g of IV methylprednisolone for the inflammatory component versus 7.5 g in the presence of diplopia and severe proptosis. Fast and significant improvements in orbital symptoms and signs are noted in 65-70% of patients. Long-term experience over decades, and worldwide availability at low cost, underline the clinical and therapeutic relevance of GC. Adverse events are rarely severe, dose-dependent, and usually reversible, hence easy to handle by medical investigators. Oral GC application on a daily basis is characterized by high bioavailability but reduced efficacy and increased toxicity. CONCLUSION IVGC still represents the standard of care in active/severe GO. Innovative biologicals, like monoclonal antibodies targeting the thyrotropin/Insulin-like growth factor-1 receptors or pro-inflammatory cytokines (e.g., Interleukin-6) should be compared with standard GC treatment with respect to short- and long-term efficacy, safety, costs, and global availability.
Collapse
Affiliation(s)
- Jan Längericht
- Department of Medicine I., Johannes Gutenberg University (JGU) Medical Center, Mainz, Rheinland-Pfalz, Germany
| | - Irene Krämer
- Department of Pharmacy, Johannes Gutenberg University (JGU) Medical Center, Mainz, Rheinland-Pfalz, Germany
| | | |
Collapse
|
38
|
Dimopoulou C, Lundgren JD, Sundal J, Ullum H, Aukrust P, Nielsen FC, Marvig RL. Variant in ERAP1 promoter region is associated with low expression in a patient with a Behçet-like MHC-I-opathy. J Hum Genet 2019; 65:325-335. [PMID: 31873220 DOI: 10.1038/s10038-019-0709-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/25/2019] [Accepted: 12/08/2019] [Indexed: 11/09/2022]
Abstract
Behçet disease (BD) is an immune-mediated disease. The cause of BD remains unknown, but the existence of multiple pathological pathways is suspected, including different genetic factors. Polymorphisms in ERAP1 gene have been associated with an increased risk of BD. However, while current BD-associated ERAP1 variants are suggested to contribute to disease by altering the activity of the encoded protein, there is no knowledge of variants that alter the expression level of ERAP1, despite previous associations between ERAP1 expression and BD. Here, we used whole-exome sequencing of a patient with a Behçet-like MHC-I-opathy to identify that the patient, unlike its healthy parents, was homozygous for a rare 1-bp deletion, rs140416843, in the promoter region of ERAP1. rs140416843 has not previously been associated with disease, but is linked to ERAP1 haplotype Hap10 which is associated with BD. The expression of ERAP1 by both RT-qPCR and RNA sequencing showed that ERAP1 mRNA expression correlated with the zygosity for the identified deletion and was decreased in comparison to a healthy cohort. In conclusion, we diagnosed the patient as having BD, and hypothesize that rs140416843-mediated changes in ERAP1 expression play a causative role in BD and that this risk factor is contributing to the association between Hap10 and BD. This is the first report to identify a variant that may cause BD by altering the expression of ERAP1, and our findings suggest that downregulation of ERAP1 expression can serve as a diagnostic marker for BD.
Collapse
Affiliation(s)
- Chrysoula Dimopoulou
- Centre of Excellence for Health, Immunity and Infection (CHIP), Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jens D Lundgren
- Centre of Excellence for Health, Immunity and Infection (CHIP), Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jon Sundal
- Department of Infectious Diseases, Stavanger University Hospital, Stavanger, Norway
| | - Henrik Ullum
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Insitute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Finn C Nielsen
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rasmus L Marvig
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| |
Collapse
|
39
|
Gouveia VM, Rizzello L, Nunes C, Poma A, Ruiz-Perez L, Oliveira A, Reis S, Battaglia G. Macrophage Targeting pH Responsive Polymersomes for Glucocorticoid Therapy. Pharmaceutics 2019; 11:pharmaceutics11110614. [PMID: 31731713 PMCID: PMC6920840 DOI: 10.3390/pharmaceutics11110614] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022] Open
Abstract
Glucocorticoid (GC) drugs are the cornerstone therapy used in the treatment of inflammatory diseases. Here, we report pH responsive poly(2-methacryloyloxyethyl phosphorylcholine)–poly(2-(diisopropylamino)ethyl methacrylate) (PMPC–PDPA) polymersomes as a suitable nanoscopic carrier to precisely and controllably deliver GCs within inflamed target cells. The in vitro cellular studies revealed that polymersomes ensure the stability, selectivity and bioavailability of the loaded drug within macrophages. At molecular level, we tested key inflammation-related markers, such as the nuclear factor-κB, tumour necrosis factor-α, interleukin-1β, and interleukin-6. With this, we demonstrated that pH responsive polymersomes are able to enhance the anti-inflammatory effect of loaded GC drug. Overall, we prove the potential of PMPC–PDPA polymersomes to efficiently promote the inflammation shutdown, while reducing the well-known therapeutic limitations in GC-based therapy.
Collapse
Affiliation(s)
- Virgínia M. Gouveia
- LAQV/REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.M.G.); (C.N.)
- Abel Salazar Biomedical Sciences Institute, University of Porto, Portugal, 4050-313 Porto, Portugal;
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK; (L.R.); (A.P.); (L.R.-P.)
- Institute of Physics of Living Systems, University College London, 20 Gordon Street, London WC1H 0AJ, UK
| | - Loris Rizzello
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK; (L.R.); (A.P.); (L.R.-P.)
- Institute of Physics of Living Systems, University College London, 20 Gordon Street, London WC1H 0AJ, UK
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Claudia Nunes
- LAQV/REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.M.G.); (C.N.)
| | - Alessandro Poma
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK; (L.R.); (A.P.); (L.R.-P.)
- Institute of Physics of Living Systems, University College London, 20 Gordon Street, London WC1H 0AJ, UK
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, 256 Gray’s Inn Road, London WC1X 8LD, UK
| | - Lorena Ruiz-Perez
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK; (L.R.); (A.P.); (L.R.-P.)
- Institute of Physics of Living Systems, University College London, 20 Gordon Street, London WC1H 0AJ, UK
- EPSRC/JEOL Centre for Liquid Phase Electron Microscopy, University College London, 20 Gordon Street, London WC1H 0AJ, UK
| | - António Oliveira
- Abel Salazar Biomedical Sciences Institute, University of Porto, Portugal, 4050-313 Porto, Portugal;
| | - Salette Reis
- LAQV/REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.M.G.); (C.N.)
- Correspondence: (S.R.); (G.B.); Tel.: +351-220-428-672 (S.R.); +44-20-7679-4688 (G.B.)
| | - Giuseppe Battaglia
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK; (L.R.); (A.P.); (L.R.-P.)
- Institute of Physics of Living Systems, University College London, 20 Gordon Street, London WC1H 0AJ, UK
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- EPSRC/JEOL Centre for Liquid Phase Electron Microscopy, University College London, 20 Gordon Street, London WC1H 0AJ, UK
- Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, 08010 Barcelona, Spain
- Correspondence: (S.R.); (G.B.); Tel.: +351-220-428-672 (S.R.); +44-20-7679-4688 (G.B.)
| |
Collapse
|
40
|
Imami L, Stanton SCE, Zilioli S, Tobin ET, Farrell AK, Luca F, Slatcher RB. Self-Disclosure and Perceived Responsiveness Among Youth With Asthma: Links to Affect and Anti-Inflammatory Gene Expression. PERSONALITY AND SOCIAL PSYCHOLOGY BULLETIN 2019; 45:1155-1169. [PMID: 30486748 PMCID: PMC9889136 DOI: 10.1177/0146167218808497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Self-disclosure and perceived responsiveness are important building blocks of social relationships that have long-lasting consequences for health and well-being. However, the conditions under which self-disclosure and responsiveness are likely to benefit health, and how early in life these benefits arise, remain unclear. Among 141 youth (aged 10-17) with asthma, we investigated how average daily levels of self-disclosure and responsiveness are linked to positive and negative affect and the expression of the glucocorticoid receptor gene NR3C1, a marker of improved regulation of stress physiology and immune functioning. Higher levels of self-disclosure were associated with higher NR3C1 expression and positive affect only when perceptions of responsiveness were high. Furthermore, perceived responsiveness was linked to NR3C1 expression for females but not males. These results suggest that the potential benefits of self-disclosure depend on the extent to which interaction partners are perceived as responsive and that these benefits emerge prior to adulthood.
Collapse
|
41
|
Profiling withanolide A for therapeutic targets in neurodegenerative diseases. Bioorg Med Chem 2019; 27:2508-2520. [DOI: 10.1016/j.bmc.2019.03.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/08/2019] [Accepted: 03/10/2019] [Indexed: 11/22/2022]
|
42
|
Johnson JD, Barnard DF, Kulp AC, Mehta DM. Neuroendocrine Regulation of Brain Cytokines After Psychological Stress. J Endocr Soc 2019; 3:1302-1320. [PMID: 31259292 PMCID: PMC6595533 DOI: 10.1210/js.2019-00053] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/06/2019] [Indexed: 12/22/2022] Open
Abstract
There is growing evidence that stress-induced brain cytokines are important in the etiology of depression and anxiety. Here, we review how the neuroendocrine responses to psychological stressors affect the immediate and long-term regulation of inflammatory cytokines within the brain and highlight how the regulation changes across time with repeated stress exposure. In doing so, we report on the percentage of studies in the literature that observed increases in either IL-1β, TNF-α, or IL-6 within the hypothalamus, hippocampus, or prefrontal cortex after either acute or chronic stress exposure. The key takeaway is that catecholamines and glucocorticoids play critical roles in the regulation of brain cytokines after psychological stress exposure. Central catecholamines stimulate the release of IL-1β from microglia, which is a key factor in the further activation of microglia and recruitment of monocytes into the brain. Meanwhile, the acute elevation of glucocorticoids inhibits the production of brain cytokines via two mechanisms: the suppression of noradrenergic locus coeruleus neurons and inhibition of the NFκB signaling pathway. However, glucocorticoids and peripheral catecholamines facilitate inflammatory responses to future stimuli by stimulating monocytes to leave the bone marrow, downregulating inhibitory receptors on microglia, and priming inflammatory responses mediated by peripheral monocytes or macrophages. The activation of microglia and the elevation of peripheral glucocorticoid and catecholamine levels are both necessary during times of stress exposure for the development of psychopathologies.
Collapse
Affiliation(s)
- John D Johnson
- Kent State University, Biological Sciences Department, School of Biomedical Sciences, Kent, Ohio
| | - David F Barnard
- Kent State University, Biological Sciences Department, School of Biomedical Sciences, Kent, Ohio
| | - Adam C Kulp
- Kent State University, Biological Sciences Department, School of Biomedical Sciences, Kent, Ohio
| | - Devanshi M Mehta
- Kent State University, Biological Sciences Department, School of Biomedical Sciences, Kent, Ohio
| |
Collapse
|
43
|
Neuroimmunomodulation in Major Depressive Disorder: Focus on Caspase 1, Inducible Nitric Oxide Synthase, and Interferon-Gamma. Mol Neurobiol 2018; 56:4288-4305. [PMID: 30306457 PMCID: PMC6505498 DOI: 10.1007/s12035-018-1359-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022]
Abstract
Major depressive disorder (MDD) is one of the leading causes of disability worldwide, and its incidence is expected to increase. Despite tremendous efforts to understand its underlying biological mechanisms, MDD pathophysiology remains elusive and pharmacotherapy outcomes are still far from ideal. Low-grade chronic inflammation seems to play a key role in mediating the interface between psychological stress, depressive symptomatology, altered intestinal microbiology, and MDD onset. We review the available pre-clinical and clinical evidence of an involvement of pro-inflammatory pathways in the pathogenesis, treatment, and remission of MDD. We focus on caspase 1, inducible nitric oxide synthase, and interferon gamma, three inflammatory systems dysregulated in MDD. Treatment strategies aiming at targeting such pathways alone or in combination with classical therapies could prove valuable in MDD. Further studies are needed to assess the safety and efficacy of immune modulation in MDD and other psychiatric disorders with neuroinflammatory components.
Collapse
|
44
|
Chung YH, Jeong SA, Choi HS, Ro S, Lee JS, Park JK. Protective effects of ginsenoside Rg2 and astaxanthin mixture against UVB-induced DNA damage. Anim Cells Syst (Seoul) 2018; 22:400-406. [PMID: 30533262 PMCID: PMC6282468 DOI: 10.1080/19768354.2018.1523806] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 09/04/2018] [Accepted: 09/11/2018] [Indexed: 12/03/2022] Open
Abstract
Ultraviolet B (UVB) radiation induces skin damage, skin matrix degradation, and wrinkle formation through photochemical reaction and oxidative stress. Therefore, protecting the skin from UVB can prevent skin aging. In this study, we investigated the effects of a mixture (RA) of Rg2, a ginsenoside, and astaxanthin, an antioxidant, on the responses of HaCaT cells exposed to UVB (700 J/m2). The cells were incubated for 24 h after UVB exposure and cell viability was determined by MTT assay. UVB decreased cell viability by 60% compared to that of untreated control cells, whereas RA increased cell viability in a concentration-dependent manner, and this increase was significantly higher than that in the single treatment groups. Further, UVB increased the levels of DNA lesions such as cyclobutane pyrimidine dimer (CPD) and 8-hydroxyguanine (8-OHdG). Conversely, RA decreased both CPD and 8-OHdG levels in a concentration-dependent manner. UVB exposure also increased phosphorylation of ataxia-telangiectasia mutated (ATM) protein kinase and p53 and subsequently increased the levels of GADD45α, p21, and matrix metalloproteinases (MMPs)-3, -9, and -13. Additionally, UVB exposure decreased the level of COL1A1. However, RA treatment decreased the levels of p-ATM, p-p53, GADD45α, p21, MMP-3, -9, and -13 and increased the level of COL1A1 in a concentration-dependent manner. These results suggest that RA reduces UVB-induced cytotoxicity and genotoxicity through up-regulation of DNA repair via the combined effects of Rg2 and astaxanthin.
Collapse
Affiliation(s)
- Yu Heon Chung
- Research Institute for Basic Science and Division of Biological Science, Wonkwang University, Iksan, Korea
| | - Seul A. Jeong
- Research Institute for Basic Science and Division of Biological Science, Wonkwang University, Iksan, Korea
| | - Hyun Seok Choi
- Research Institute for Basic Science and Division of Biological Science, Wonkwang University, Iksan, Korea
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Jung Sup Lee
- Department of Biomedical Science, BK21-plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, Gwangju, Republic of Korea
| | - Jong Kun Park
- Research Institute for Basic Science and Division of Biological Science, Wonkwang University, Iksan, Korea
| |
Collapse
|
45
|
Jhun EH, Sadhu N, Yao Y, He Y, Molokie RE, Wilkie DJ, Wang ZJ. Glucocorticoid receptor single nucleotide polymorphisms are associated with acute crisis pain in sickle cell disease. Pharmacogenomics 2018; 19:1003-1011. [PMID: 30079801 DOI: 10.2217/pgs-2018-0064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM Pain in sickle cell disease patients is heterogeneous and genetic polymorphisms may predispose an individual to varied vulnerability to painful events. We studied the association of SNPs in the glucocorticoid receptor gene (NR3C1) with pain in sickle cell disease. METHOD Acute pain was scored as the number of utilizations due to crisis pain in a 12-month period. Chronic pain was calculated as the Composite Pain Index score. RESULTS & CONCLUSION rs33389 T allele (IRR = 1.53, p = 0.014 additive; IRR = 1.64, p = 0.011 recessive), rs2963155 G allele (IRR = 1.80, p < 0.001 additive; IRR = 2.25, p = 0.021 dominant; IRR = 2.07, p < 0.001 recessive) and rs9324918 C allele (IRR = 1.43, p = 0.021 additive) were associated with higher utilization rates, indicating the potential contribution of NR3C1 polymorphisms to acute pain heterogeneity in sickle cell disease.
Collapse
Affiliation(s)
- Ellie H Jhun
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60612, USA
| | - Nilanjana Sadhu
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60612, USA
| | - Yingwei Yao
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, FL 32610, USA
| | - Ying He
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60612, USA.,Comprehensive Sickle Cell Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Robert E Molokie
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60612, USA.,Comprehensive Sickle Cell Center, University of Illinois at Chicago, Chicago, IL 60612, USA.,Jesse Brown Veteran's Administration Medical Center, Chicago, IL 60612, USA.,Division of Hematology/Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| | - Diana J Wilkie
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing, Gainesville, FL 32610, USA
| | - Zaijie Jim Wang
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago College of Pharmacy, Chicago, IL 60612, USA.,Comprehensive Sickle Cell Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
46
|
Farrell AK, Slatcher RB, Tobin ET, Imami L, Wildman DE, Luca F, Zilioli S. Socioeconomic status, family negative emotional climate, and anti-inflammatory gene expression among youth with asthma. Psychoneuroendocrinology 2018; 91. [PMID: 29529520 PMCID: PMC5903571 DOI: 10.1016/j.psyneuen.2018.02.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The glucocorticoid receptor gene NR3C1 is an important down-regulator of inflammation and is typically under-expressed in individuals with low socioeconomic status (SES). Negative emotionality has been suggested as a potential mediator of SES disparities in health outcomes. In this study, we expand this literature by naturalistically assessing negative emotionality in a key emotional environment: the family. In a sample of 104 youth with asthma (10-17 years) and their primary caregiver, we assessed SES via caregiver report, emotional expression by youth and parents in the home over four days using the electronically activated recorder (EAR), and NR3C1 expression via blood collected from youth. Although there was not a direct effect of SES on NR3C1 expression, bootstrapping mediation analyses showed a significant indirect path such that lower SES was associated with a more negative family emotional climate, which in turn predicted reduced NR3C1 expression. No mediation effects were found for family positive emotional climate. This research demonstrates the importance of examining the effects of SES on emotion expression in the family context and suggests a critical biopsychosocial pathway underlying SES-based health disparities that may extend beyond youth.
Collapse
Affiliation(s)
- Allison K. Farrell
- Department of Psychology, Wayne State University, 5057 Woodward Ave,
Detroit, MI 48202,Center for Molecular Medicine and Genetics and Department of
Obstetrics and Gynecology, Wayne State University, 540 East Canfield, Detroit, MI
48201
| | - Richard B. Slatcher
- Department of Psychology, Wayne State University, 5057 Woodward Ave,
Detroit, MI 48202
| | - Erin T. Tobin
- Department of Internal Medicine, Henry Ford Health System, Detroit,
MI
| | - Ledina Imami
- Department of Psychology, Wayne State University, 5057 Woodward Ave,
Detroit, MI 48202
| | - Derek E. Wildman
- Department of Molecular & Integrative Physiology and Carl R.
Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign,
1206 West Gregory Dr, Urbana, IL 61801
| | - Francesca Luca
- Center for Molecular Medicine and Genetics and Department of
Obstetrics and Gynecology, Wayne State University, 540 East Canfield, Detroit, MI
48201
| | - Samuele Zilioli
- Department of Psychology, Wayne State University, 5057 Woodward Ave, Detroit, MI 48202, United States; Department of Family Medicine and Public Health Sciences, Wayne State University, 3939 Woodward Ave, Detroit, MI 48201, United States.
| |
Collapse
|
47
|
Husta BC, Raoof S, Erzurum S, Mehta AC. Tracheobronchopathy From Inhaled Corticosteroids. Chest 2017; 152:1296-1305. [PMID: 28864055 PMCID: PMC6026226 DOI: 10.1016/j.chest.2017.08.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/06/2017] [Accepted: 08/01/2017] [Indexed: 12/17/2022] Open
Abstract
Inhaled corticosteroids (ICSs) have become the mainstay of asthma control. They are also recommended as an add-on therapy to long-acting beta agonists and anticholinergics in moderate to severe COPD with recurrent exacerbations. Ultimately this clinical practice has led to the widespread use of ICSs, which are supported by a more favorable side effect profile than that of systemic steroids.
Collapse
Affiliation(s)
- Bryan C Husta
- Lenox Hill Hospital, Hofstra Northwell School of Medicine, New York, NY
| | - Suhail Raoof
- Lenox Hill Hospital, Hofstra Northwell School of Medicine, New York, NY
| | - Serpil Erzurum
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Atul C Mehta
- Respiratory Institute, Cleveland Clinic, Cleveland, OH.
| |
Collapse
|
48
|
Cochlear Transcriptome Following Acoustic Trauma and Dexamethasone Administration Identified by a Combination of RNA-seq and DNA Microarray. Otol Neurotol 2017; 38:1032-1042. [DOI: 10.1097/mao.0000000000001373] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Cytokine profile of NALT during acute stress and its possible effect on IgA secretion. Immunol Lett 2017; 188:68-78. [PMID: 28472640 DOI: 10.1016/j.imlet.2017.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/27/2017] [Accepted: 04/28/2017] [Indexed: 12/20/2022]
Abstract
Stress stimuli affect the immune system responses that occur at mucosal membranes, particularly IgA secretion. It has been suggested that acute stress increases the levels of IgA and that sympathetic innervation plays an important role in this process. We herein explore in a murine model how acute stress affects the Th1/Th2/Treg cytokine balance in NALT, and the possible role of glucocorticoids in this effect. Nine-week-old male CD1 mice were divided into three groups: unstressed (control), stressed (subjected to 4h of immobilization), and stressed after pretreatment with a single dose of the corticosterone receptor antagonist RU-486. The parameters evaluated included plasma corticosterone and epinephrine, IgA levels in nasal fluid (by ELISA), the percentage of CD19+B220+IgA+ lymphocytes and CD138+IgA+ plasma cells, and the mRNA expression of heavy α chain, J chain and pIgR. Moreover, the gene and protein expression of Th1 cytokines (TNFα, IL-2 and INF-γ), Th2 cytokines (IL-4 and IL-5) and Treg cytokines (IL-10 and TGFβ) were determined in nasal mucosa. The results show that acute stress generated a shift towards the dominance of an anti-inflammatory immune response (Th2 and Treg cytokines), evidenced by a significant rise in the amount of T cells that produce IL4, IL-5 and IL-10. This immune environment may favor IgA biosynthesis by CD138+IgA+ plasma cells, a process mediated mostly by glucocorticoids.
Collapse
|
50
|
|