1
|
Díaz-Catalán D, Capó J, Vega-Beyhart A, Rodrigo-Calvo MT, Mora M, Vidal O, Squarcia M, Enseñat J, Casals G, Hanzu F. Sex-dependent effects of FGF21 on HPA axis regulation and adrenal regeneration after Cushing syndrome in mice. Mol Metab 2025; 96:102122. [PMID: 40154841 PMCID: PMC12013334 DOI: 10.1016/j.molmet.2025.102122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Cushing's syndrome (CS) results from prolonged exposure to excessive glucocorticoids (GCs), leading to metabolic disturbances and adrenal insufficiency (AI). Fibroblast growth factor 21 (FGF21) has shown promise as a potential therapeutic target for metabolic disorders. This study explores the effects of FGF21 on adrenal gland function in a mouse model of AI following chronic hypercortisolism and investigates sex-dependent differences in the hypothalamic-pituitary-adrenal (HPA) axis response. METHODS We employed a mouse model of AI after chronic corticosterone (CORT) treatment. The effects of recombinant human FGF21 (hFGF21) administration on adrenal function were evaluated in AI mice. Male and female wild-type (WT) and FGF21-overexpressing transgenic (Tg) mice were subjected to 5 weeks of CORT treatment, reaching CS phenotype, followed by immediate analysis or a 10-week recovery period. Metabolic parameters, HPA axis function, and adrenal gland morphology and gene expression were assessed. RESULTS Prolonged CORT exposure resulted in metabolic disturbances and HPA axis dysregulation. hFGF21 treatment increased CORT and ACTH secretion in AI mice. FGF21 overexpression influenced glucose homeostasis and insulin regulation during CORT treatment and recovery, with sex-specific effects. Tissue-specific regulation of Klb expression was observed across the HPA axis, with distinct patterns between males and females. Tg mice displayed altered adrenal progenitor cell activation and steroidogenic gene expression. Sex-specific differences were observed in adrenal capsule remodeling and gene expression patterns during recovery. CONCLUSIONS This study reveals the complex interplay between FGF21 signaling and GC-induced metabolic and endocrine changes, suggesting a potential sex-specific role of FGF21 in metabolic regulation and HPA axis recovery following after CS.
Collapse
Affiliation(s)
- Daniela Díaz-Catalán
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain.
| | - Júlia Capó
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain
| | - Arturo Vega-Beyhart
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain
| | | | - Mireia Mora
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain; Endocrinology Department, Hospital Clínic, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Oscar Vidal
- Endocrine Surgery Department, Hospital Clínic Barcelona, University of Barcelona, Spain
| | - Mattia Squarcia
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain; The Biomedical Diagnostic Center (CBD), Barcelona, Spain
| | - Joaquim Enseñat
- Neurosurgery Department, Hospital Clinic Barcelona, University of Barcelona, Spain
| | - Gregori Casals
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain; Biochemistry and Molecular Genetics Service, Hospital Clínic, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain
| | - Felicia Hanzu
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain; Endocrinology Department, Hospital Clínic, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain.
| |
Collapse
|
2
|
Song X, Cheng M, Gu C, Wang F, Ma K, Wang C, She X, Cui B. Research progress in modulating the auditory system by the cochlear circadian clock system in response to noise exposure. Front Neurosci 2025; 19:1507363. [PMID: 40171535 PMCID: PMC11958988 DOI: 10.3389/fnins.2025.1507363] [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: 10/07/2024] [Accepted: 03/05/2025] [Indexed: 04/03/2025] Open
Abstract
The circadian clock is an endogenous system evolved to adapt to environmental changes. Recent studies confirmed that the cochlea exhibits circadian oscillations regulating auditory function. These oscillations are linked to brain-derived neurotrophic factor and glucocorticoid levels. Circadian rhythms influence cochlear sensitivity to noise by regulating the secretion of brain-derived neurotrophic factors and glucocorticoids. This study explores the regulatory mechanism of the circadian clock system, its impact on the auditory system, and its potential role in noise-induced hearing loss. Understanding the regulatory mechanisms of circadian rhythms in auditory function will provide new ideas for developing treatments for noise-induced hearing loss.
Collapse
Affiliation(s)
- Xiaoqiong Song
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
- Academy of Military Medical Sciences, Tianjin, China
| | - Mengzhu Cheng
- Academy of Military Medical Sciences, Tianjin, China
- School of Public Health and Management, Binzhou Medical University, Yantai, Shandong, China
| | - Cui Gu
- Academy of Military Medical Sciences, Tianjin, China
- School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fenghan Wang
- Academy of Military Medical Sciences, Tianjin, China
- School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kefeng Ma
- Academy of Military Medical Sciences, Tianjin, China
| | - Chunping Wang
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
| | - Xiaojun She
- Academy of Military Medical Sciences, Tianjin, China
| | - Bo Cui
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, China
- Academy of Military Medical Sciences, Tianjin, China
| |
Collapse
|
3
|
Afshar AM, Sharma SK, Hamidi SH, Marznaki ZH, Mudgal SK, Kamyari N, Khalilizad M. Comparison of Dexamethasone at Three Doses Administered Postoperatively for Improving Pain Control and Sleep Quality in Patients Who Underwent Total Knee Arthroplasty: A Triple Blind Randomized Controlled Trial. J Arthroplasty 2025; 40:658-664. [PMID: 39284389 DOI: 10.1016/j.arth.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/31/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Dexamethasone has been shown to alleviate pain, yet the optimal dosing and safety profiles remain unclear. This study aimed to evaluate the analgesic efficacy and impact on sleep quality of three different doses of intravenous dexamethasone in patients undergoing total knee arthroplasty (TKA). METHODS In this randomized, triple-blind, clinical trial, we assessed the analgesic effects of three doses of intravenous dexamethasone (four, eight, and 16 mg) in adult patients who underwent TKA. Pain was measured using the visual analog scale at one, 12, 24, and 48 hours postoperatively, and sleep quality was assessed two weeks postsurgery. RESULTS A total of 90 participants were enrolled in the study, with 30 participants in each dosing group. The mean visual analog scale scores at 12, 24, and 48 hours postoperatively showed significant improvement from baseline in all groups. Notably, the 16 mg and eight mg dexamethasone groups demonstrated significantly greater pain reduction compared to the four mg group (P < 0.05). Additionally, sleep quality significantly improved in the 16 mg and eight mg groups (P < 0.05). CONCLUSIONS Dexamethasone at doses of four, eight, and 16 mg effectively reduces pain and enhances sleep quality in patients undergoing TKA, with the 16 mg dose showing the most pronounced effects at 12, 24, and 48 hours postoperatively.
Collapse
Affiliation(s)
- Amir M Afshar
- Resident of Orthopaedic, Department of Orthopaedic and Trauma Surgery, Babol University of Medical Sciences, Babol, Iran
| | - Suresh K Sharma
- College of Nursing, All India Institute of Medical Sciences, Jodhpur, India
| | - Seyed Hosseini Hamidi
- Department of Anesthesiology and Critical Care, Babol University of Medical Sciences, Babol, Iran
| | | | - Shiv K Mudgal
- College of Nursing, All India Institute of Medical Sciences, Deoghar, India
| | - Naser Kamyari
- Department of Biostatistics and Epidemiology, School of Health, Abadan University of Medical Sciences, Abadan, Iran
| | - Majid Khalilizad
- Department of Orthopaedic and Trauma Surgery, Shahid Beheshti Hospital and Mobility Impairment Research Centre, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
4
|
Wang Q, Zhang H, Chen Y, Lv X, Qiao Y, Zhu Q. Impact of baseline glucocorticoids (GCs) on cardiotoxic events and myocardial damage related to immune checkpoint inhibitors: a retrospective clinical research. Expert Opin Drug Saf 2025:1-12. [PMID: 39953683 DOI: 10.1080/14740338.2025.2467814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs)-associated cardiotoxic events (CEs) are of increasing concern. Existing research about glucocorticoids (GCs) on immunotherapy focused on ICIs' efficacy and patients' outcome. The influence of GCs on ICIs-associated CEs and myocardial damage (MD) remains unknown. RESEARCH DESIGN AND METHODS This single-center retrospective study included patients treated with ICIs from 2018 to 2022, with follow-up period ending on 30 June 2023. The incidence, risk factors of ICIs-associated CEs, especially MD were described. Additionally, the impact of baseline GCs was assessed by propensity score matching (PSM) to mitigate intergroup differences and ensure comparability. RESULTS Among 1018 patients, 204 (20.04%) experienced ICIs-associated CEs, including 71 (6.97%) with MD. The mean follow-up time was 40.39 (95% CI 38.47-42.31) weeks. The median time to onset of MD was the shortest at 12.57 weeks (IQR 5.29-25.14). Tumor type, co-medication with platinum and angiogenesis inhibitors may be influential factors of MD. After PSM, the relative risks of CEs (OR 0.4625,95%CI 0.2514-0.7235, p = 0.0020) and MD (OR 0.3254, 95% CI 0.1190-0.8898, p = 0.0378) in GCs1 ≥ 20 mg group were both significantly lower than those in GCs1 < 20 mg. CONCLUSION GCs ≥ 20 mg during the first ICIs treatment cycle is significantly associated with the reduced risks of both ICIs-associated CEs and MD.
Collapse
Affiliation(s)
- Qiaoyun Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Department of Pharmacy, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Haixia Zhang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, SAR, China
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| | - Yawen Chen
- Department of Pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin Lv
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yanli Qiao
- Department of Pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qiaoling Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, China
| |
Collapse
|
5
|
Hiltunen J, Helminen L, Paakinaho V. Glucocorticoid receptor action in prostate cancer: the role of transcription factor crosstalk. Front Endocrinol (Lausanne) 2024; 15:1437179. [PMID: 39027480 PMCID: PMC11254642 DOI: 10.3389/fendo.2024.1437179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Prostate cancer is one of the most prevalent malignancies and is primarily driven by aberrant androgen receptor (AR) signaling. While AR-targeted therapies form the cornerstone of prostate cancer treatment, they often inadvertently activate compensatory pathways, leading to therapy resistance. This resistance is frequently mediated through changes in transcription factor (TF) crosstalk, reshaping gene regulatory programs and ultimately weakening treatment efficacy. Consequently, investigating TF interactions has become crucial for understanding the mechanisms driving therapy-resistant cancers. Recent evidence has highlighted the crosstalk between the glucocorticoid receptor (GR) and AR, demonstrating that GR can induce prostate cancer therapy resistance by replacing the inactivated AR, thereby becoming a driver of the disease. In addition to this oncogenic role, GR has also been shown to act as a tumor suppressor in prostate cancer. Owing to this dual role and the widespread use of glucocorticoids as adjuvant therapy, it is essential to understand GR's actions across different stages of prostate cancer development. In this review, we explore the current knowledge of GR in prostate cancer, with a specific focus on its crosstalk with other TFs. GR can directly and indirectly interact with a variety of TFs, and these interactions vary significantly depending on the type of prostate cancer cells. By highlighting these crosstalk interactions, we aim to provide insights that can guide the research and development of new GR-targeted therapies to mitigate its harmful effects in prostate cancer.
Collapse
Affiliation(s)
| | | | - Ville Paakinaho
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
6
|
Heath H, Yoo JY, Akter S, Jain A, Sharma V, McGee H, Soliman A, Mahmoud AM, Matthews AK, Winn RA, Madak-Erdogan Z, Kim SJ. The Effect of Exposure to Neighborhood Violence on Glucocorticoid Receptor Signaling in Lung Tumors. CANCER RESEARCH COMMUNICATIONS 2024; 4:1643-1654. [PMID: 38912926 PMCID: PMC11221527 DOI: 10.1158/2767-9764.crc-24-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/02/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024]
Abstract
Despite lower rates and intensity of smoking, Black men experience a higher incidence of lung cancer compared to white men. The racial disparity in lung cancer is particularly pronounced in Chicago, a highly segregated urban city. Neighborhood conditions, particularly social stress, may play a role in lung tumorigenesis. Preliminary studies indicate that Black men residing in neighborhoods with higher rates of violent crime have significantly higher levels of hair cortisol, an indicator of stress response. To examine the relationship between social stress exposure and gene expression in lung tumors, we investigated glucocorticoid receptor (GR) binding in 15 lung tumor samples in relation to GR target gene expression levels and zip code level residential violent crime rates. Spatial transcriptomics and a version of ChIP sequencing known as CUT&RUN were used. Heatmap of genes, pathway analysis, and motif analysis were conducted at the statistical significance of P < 0.05. GR recruitment to chromatin was correlated with zip code level residential violent crime rate and overall GR binding increased with higher violent crime rates. Our findings suggest that exposure to residential violent crime may influence tumor biology via reprogramming GR recruitment. Prioritizing lung cancer screening in neighborhoods with increased social stress, such as high levels of violent crime, may reduce racial disparities in lung cancer. SIGNIFICANCE Exposure to neighborhood violent crime is correlated with glucocorticoid signaling and lung tumor gene expression changes associated with increased tumor aggressiveness, suggesting social conditions have downstream biophysical consequences that contribute to lung cancer disparities.
Collapse
Affiliation(s)
- Hannah Heath
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, Illinois.
| | - Jin Y. Yoo
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, Illinois.
| | - Sabrina Akter
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, Illinois.
| | - Atharva Jain
- National Center for Supercomputing Applications, University of Illinois Urbana-Champaign, Urbana, Illinois.
| | - Vani Sharma
- School of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois.
| | - Hannah McGee
- School of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois.
| | - Aiman Soliman
- National Center for Supercomputing Applications, University of Illinois Urbana-Champaign, Urbana, Illinois.
| | - Abeer M. Mahmoud
- Department of Medicine, University of Illinois Chicago, Chicago, Illinois.
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, Illinois.
| | | | - Robert A. Winn
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
| | - Zeynep Madak-Erdogan
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, Illinois.
- National Center for Supercomputing Applications, University of Illinois Urbana-Champaign, Urbana, Illinois.
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, Illinois.
- Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois.
| | - Sage J. Kim
- School of Public Health, University of Illinois Chicago, Chicago, Illinois.
- University of Illinois Cancer Center, Chicago, Illinois.
| |
Collapse
|
7
|
Xu W, Ye J, Cao Z, Zhao Y, Zhu Y, Li L. Glucocorticoids in lung cancer: Navigating the balance between immunosuppression and therapeutic efficacy. Heliyon 2024; 10:e32357. [PMID: 39022002 PMCID: PMC11252876 DOI: 10.1016/j.heliyon.2024.e32357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
Glucocorticoids (GCs), a class of hormones secreted by the adrenal glands, are released into the bloodstream to maintain homeostasis and modulate responses to various stressors. These hormones function by binding to the widely expressed GC receptor (GR), thereby regulating a wide range of pathophysiological processes, especially in metabolism and immunity. The role of GCs in the tumor immune microenvironment (TIME) of lung cancer (LC) has been a focal point of research. As immunosuppressive agents, GCs exert a crucial impact on the occurrence, progression, and treatment of LC. In the TIME of LC, GCs act as a constantly swinging pendulum, simultaneously offering tumor-suppressive properties while diminishing the efficacy of immune-based therapies. The present study reviews the role and mechanisms of GCs in the TIME of LC.
Collapse
Affiliation(s)
| | | | - Zhendong Cao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Yupei Zhao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Yimin Zhu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Lei Li
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| |
Collapse
|
8
|
Höpner L, Proschmann U, Inojosa H, Ziemssen T, Akgün K. Corticosteroid-depending effects on peripheral immune cell subsets vary according to disease modifying strategies in multiple sclerosis. Front Immunol 2024; 15:1404316. [PMID: 38938576 PMCID: PMC11208457 DOI: 10.3389/fimmu.2024.1404316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
Background The primary treatment for acute relapses in multiple sclerosis (MS) is the intravenous administration of high-dose methylprednisolone (IVMP). However, the mechanisms through which corticosteroid treatment impacts acute neuroinflammation in people with MS (pwMS) remain not fully understood. In particular, the changes induced by glucocorticoids (GCs) on cells of the innate immune system and the differences between patients with distinct immunotherapies have received little attention to date. Methods We conducted immunophenotyping using flow cytometry on peripheral blood mononuclear cells of pwMS who received IVMP treatment during a relapse. We compared the impact of an IVMP treatment on a broad variety of immune cell subsets within three groups: twelve patients who were treatment-naïve to disease modifying therapies (wDMT) to ten patients on platform therapies (PT) and eighteen patients on fingolimod therapy (FTY). Results We observed pronounced interindividual short- and intermediate-term effects of IVMP on distinct immune cells subsets. In addition to the well-documented decrease in T-helper cells (Th cells), we detected significant alterations after the first IVMP infusion within the innate immune response among neutrophil, eosinophil and basophil granulocytes, monocytes and plasmacytoid dendritic cells (pDCs). When comparing patients wDMT to the PT and FTY cohorts, we found that IVMP had a similar impact on innate immune cells across all treatment groups. However, we did not observe a significant further decline in T lymphocyte counts during IVMP in patients with pre-existing lymphopenia under FTY treatment. Although T cell apoptosis is considered the main mechanism of action of GCs, patients with FTY still reported symptom improvement following IVMP treatment. Conclusion In addition to T cell suppression, our data suggests that further immunoregulatory mechanisms of GC, particularly on cells of the innate immune response, are of greater significance than previously understood. Due to the regulation of the adaptive immune cells by DMTs, the impact of GC on these cells varies depending on the underlying DMT. Additional studies involving larger cohorts and cerebrospinal fluid samples are necessary to gain a deeper understanding of the immune response to GC in pwMS with different DMTs during relapse to define and explain differences in clinical response profiles.
Collapse
Affiliation(s)
| | | | | | | | - Katja Akgün
- Center of Clinical Neuroscience, Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| |
Collapse
|
9
|
de Lima AMDL, de Lima Rosa G, Guzzo EFM, Padilha RB, de Araujo MC, da Silva RC, Coitinho AS, Van Der Sand ST. Effect of prednisolone in a kindling model of epileptic seizures in rats on cytokine and intestinal microbiota diversity. Epilepsy Behav 2024; 155:109800. [PMID: 38657485 DOI: 10.1016/j.yebeh.2024.109800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/26/2024] [Accepted: 04/14/2024] [Indexed: 04/26/2024]
Abstract
Epilepsy is a neurological disease characterized by spontaneous and recurrent seizures. Epileptic seizures can be initiated and facilitated by inflammatory mechanisms. As the dysregulation of the immune system would be involved in epileptogenesis, it is suggested that anti-inflammatory medications could impact epileptic seizures. These medications could potentially have a side effect by altering the structure and composition of the intestinal microbiota. These changes can disrupt microbial homeostasis, leading to dysbiosis and potentially exacerbating intestinal inflammation. We hypothesize that prednisolone may affect the development of epileptic seizures, potentially influencing the diversity of the intestinal microbiota and the regulation of pro-inflammatory cytokines in intestinal tissue. This study aimed to evaluate the effects of prednisolone treatment on epileptic seizures and investigate the effect of this drug on the bacterial diversity of the intestinal microbiota and markers of inflammatory processes in intestinal tissue. We used Male Wistar rat littermates (n = 31, 90-day-old) divided into four groups: positive control treated with 2 mg/kg of diazepam (n = 6), negative control treated with 0.9 g% sodium chloride (n = 6), and the remaining two groups were subjected to treatment with prednisolone, with one receiving 1 mg/kg (n = 9) and the other 5 mg/kg (n = 10). All administrations were performed intraperitoneally (i.p.) over 14 days. To induce the chronic model of epileptic seizures, we administered pentylenetetrazole (PTZ) 25 mg/kg i.p. on alternate days. Seizure latency (n = 6 - 10) and TNF-α and IL-1β concentrations from intestinal samples were measured by ELISA (n = 6 per group), and intestinal microbiota was evaluated with intergenic ribosomal RNA (rRNA) spacer (RISA) analysis (n = 6 per group). The prednisolone treatment demonstrated an increase in the latency time of epileptic seizures and TNF-α and IL-1β concentrations compared to controls. There was no statistically significant difference in intestinal microbiota diversity between the different treatments. However, there was a strong positive correlation between microbial diversity and TNF-α and IL-1β concentrations. The administration of prednisolone yields comparable results to diazepam on increasing latency between seizures, exhibiting promise for its use in clinical studies. Although there were no changes in intestinal microbial diversity, the increase in the TNF-α and IL-1β cytokines in intestinal tissue may be linked to immune system signaling pathways involving the intestinal microbiota. Additional research is necessary to unravel the intricacies of these pathways and to understand their implications for clinical practice.
Collapse
Affiliation(s)
- Amanda Muliterno Domingues Lourenço de Lima
- Programa de Pós-Graduação em Microbiologia Agrícola e do Ambiente, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil
| | - Gabriel de Lima Rosa
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil
| | - Edson Fernando Müller Guzzo
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil
| | - Rafael Bremm Padilha
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil
| | - Milena Conci de Araujo
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil
| | - Rodrigo Costa da Silva
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil
| | - Adriana Simon Coitinho
- Programa de Pós-Graduação em Ciências Biológicas: Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil; Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil.
| | - Sueli Teresinha Van Der Sand
- Programa de Pós-Graduação em Microbiologia Agrícola e do Ambiente, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil; Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul, Instituto de Ciências Básicas da Saúde, Ramiro Barcelos Street, 2.600, Porto Alegre, RS, Brazil
| |
Collapse
|
10
|
Pokhylko V, Cherniavska Y, Fishchuk L, Rossokha Z, Popova O, Vershyhora V, Ievseienkova O, Soloviova H, Zhuk L, Gorovenko N. Association of the C3953T (rs1143634) variant of the interleukin 1 beta gene with the features of a complicated course of COVID-19-associated pneumonia. Mol Biol Rep 2024; 51:630. [PMID: 38720147 DOI: 10.1007/s11033-024-09569-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/19/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND The pro-inflammatory cytokine IL-1 plays an important role in severe COVID-19. A change in IL-1 production may be associated with a mutation in the IL1Β gene. Our study analyzed the impact of the IL1Β gene variants (rs1143634) on disease progression in patients with severe COVID-19 pneumonia, taking into account treatment strategies. METHODS AND RESULTS The study enrolled 117 patients with severe COVID-19 pneumonia. The IL1Β gene variants were identified using the polymerase chain reaction-restriction fragment length polymorphism method. In the group of patients, the following genotype frequencies were found based on the investigated rs1143634 variant of the IL1Β gene: CC-65.8%, CT-28.2%, and TT-6.0%. Our results showed that the group of patients with the T allele of the IL1Β gene had higher leukocyte counts (p = 0.040) and more pronounced lymphopenia (p = 0.007). It was determined that patients carrying the T allele stayed on ventilators significantly longer (p = 0.049) and required longer treatment with corticosteroids (p = 0.045). CONCLUSION Identifying variants of the IL1Β gene can be used as a predictive tool for assessing the severity of COVID-19 pneumonia and tailoring personalized treatment strategies. Further research with a larger patient cohort is required to validate these findings.
Collapse
Affiliation(s)
| | | | - Liliia Fishchuk
- Department of Genetic Diagnostics, Institute of Genetic and Regenerative Medicine, SI "M.D. Strazhesko National Scientific Center of the NAMS of Ukraine", Kyiv, Ukraine.
| | - Zoia Rossokha
- SI "Reference-Center for Molecular Diagnostics of the Ministry of Public Health of Ukraine", Kyiv, Ukraine
| | - Olena Popova
- SI "Reference-Center for Molecular Diagnostics of the Ministry of Public Health of Ukraine", Kyiv, Ukraine
| | - Viktoriia Vershyhora
- SI "Reference-Center for Molecular Diagnostics of the Ministry of Public Health of Ukraine", Kyiv, Ukraine
| | - Olena Ievseienkova
- Department of Genetic Diagnostics, Institute of Genetic and Regenerative Medicine, SI "M.D. Strazhesko National Scientific Center of the NAMS of Ukraine", Kyiv, Ukraine
| | | | | | - Nataliia Gorovenko
- Department of Genetic Diagnostics, Institute of Genetic and Regenerative Medicine, SI "M.D. Strazhesko National Scientific Center of the NAMS of Ukraine", Kyiv, Ukraine
| |
Collapse
|
11
|
Lee J, Song J, Kim SG, Yun D, Kang MW, Kim DK, Oh KH, Joo KW, Kim YS, Han SS, Kim YC. Mortality associated with the neutrophil-lymphocyte ratio in septic acute kidney injury requiring continuous renal replacement therapy. Kidney Res Clin Pract 2024; 43:337-347. [PMID: 38325867 PMCID: PMC11181042 DOI: 10.23876/j.krcp.23.116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/10/2023] [Accepted: 10/24/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Sepsis is an important cause of acute kidney injury in intensive care unit patients, accounting for 15% to 20% of renal replacement therapy prescriptions. The neutrophil-lymphocyte ratio (NLR), a marker of systemic inflammation and immune response, was previously associated with the mortality rate in multiple conditions. Herein, we aimed to examine how the NLR relates to the mortality rate in septic acute kidney injury patients requiring continuous renal replacement therapy (CRRT). METHODS The NLRs of 6 and 18 were used for dividing NLRs into three groups and, thus, were set higher than those in previous studies accounting for steroid use in sepsis. Cox proportional hazard models were used to calculate hazard ratios of mortality outcomes before and after matching their propensity scores. RESULTS A total of 798 septic acute kidney injury patients requiring CRRT were classified into three NLR groups (low, <6 [n = 277]; medium, ≥6 and <18 [n = 115], and high, ≥18 [n = 406], respectively). The in-hospital mortality rates per group were 83.4%, 74.8%, and 70.4%, respectively (p < 0.001). Per the univariable Cox survival analysis after propensity score matching, a high NLR was related to approximately 24% reduced mortality. The survival benefit of the high NLR group compared with the other two groups remained consistent across all subgroups, showing any p for interactions of >0.05. CONCLUSION A high NLR is associated with better clinical outcomes, such as low mortality, in septic acute kidney injury patients undergoing CRRT.
Collapse
Affiliation(s)
- Jinwoo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeongin Song
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seong Geun Kim
- Department of Internal Medicine, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Donghwan Yun
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Min Woo Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
12
|
Marinho A, Seabra CL, Lima SAC, Lobo-da-Cunha A, Reis S, Nunes C. Empowering Naringin's Anti-Inflammatory Effects through Nanoencapsulation. Int J Mol Sci 2024; 25:4152. [PMID: 38673736 PMCID: PMC11050564 DOI: 10.3390/ijms25084152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Abundant in citrus fruits, naringin (NAR) is a flavonoid that has a wide spectrum of beneficial health effects, including its anti-inflammatory activity. However, its use in the clinic is limited due to extensive phase I and II first-pass metabolism, which limits its bioavailability. Thus, lipid nanoparticles (LNPs) were used to protect and concentrate NAR in inflamed issues, to enhance its anti-inflammatory effects. To target LNPs to the CD44 receptor, overexpressed in activated macrophages, functionalization with hyaluronic acid (HA) was performed. The formulation with NAR and HA on the surface (NAR@NPsHA) has a size below 200 nm, a polydispersity around 0.245, a loading capacity of nearly 10%, and a zeta potential of about 10 mV. In vitro studies show the controlled release of NAR along the gastrointestinal tract, high cytocompatibility (L929 and THP-1 cell lines), and low hemolytic activity. It was also shown that the developed LNPs can regulate inflammatory mediators. In fact, NAR@NPsHA were able to decrease TNF-α and CCL-3 markers expression by 80 and 90% and manage to inhibit the effects of LPS by around 66% for IL-1β and around 45% for IL-6. Overall, the developed LNPs may represent an efficient drug delivery system with an enhanced anti-inflammatory effect.
Collapse
Affiliation(s)
- Andreia Marinho
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal; (A.M.); (C.L.S.); (S.R.)
- LAQV, REQUIMTE, Faculdade de Ciências, Universidade do Porto, R. do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Catarina Leal Seabra
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal; (A.M.); (C.L.S.); (S.R.)
| | - Sofia A. C. Lima
- LAQV, REQUIMTE, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Alexandre Lobo-da-Cunha
- Departamento de Microscopia, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Salette Reis
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal; (A.M.); (C.L.S.); (S.R.)
| | - Cláudia Nunes
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal; (A.M.); (C.L.S.); (S.R.)
- LAQV, REQUIMTE, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| |
Collapse
|
13
|
Mehta V, Dwivedi AR, Ludhiadch A, Rana V, Goel KK, Uniyal P, Joshi G, Kumar A, Kumar B. A decade of USFDA-approved small molecules as anti-inflammatory agents: Recent trends and Commentaries on the "industrial" perspective. Eur J Med Chem 2024; 263:115942. [PMID: 38000212 DOI: 10.1016/j.ejmech.2023.115942] [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: 10/08/2023] [Revised: 11/05/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023]
Abstract
Inflammation is the human body's defence process against various pathogens, toxic substances, irradiation, and physically injured cells that have been damaged. Inflammation is characterized by swelling, pain, redness, heat, as well as diminished tissue function. Multiple important inflammatory markers determine the prognosis of inflammatory processes, which include likes of pro-inflammatory cytokines which are controlled by nuclear factor kappa-B (NF-kB), mitogen-activated protein kinase (MAPK), Janus kinase signal transducer and activator of transcription (JAK-STAT) pathway, all of which are activated in response to the stimulation of specific receptors. Besides these, the cyclooxygenase (COX) enzyme family also plays a significant role in inflammation. The current review is kept forth to compile a summary of small molecules-based drugs approved by the USFDA during the study period of 2013-2023. A thorough discussion has also been made to focus on biologics, macromolecules, and small chemical entities approved during this study period and their greener synthetic routes with a brief discussion on the chemical spacing parameters of anti-inflammatory drugs. The compilation is expected to assist the medicinal chemist and the scientist actively engaged in drug discovery and development of anti-inflammatory agents from newer perspectives during the current years.
Collapse
Affiliation(s)
- Vikrant Mehta
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, Texas, 78229, USA
| | | | - Abhilash Ludhiadch
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, 10032, USA
| | - Vikas Rana
- School of Pharmacy, Graphic Era Hill University, Clement town, Dehradun, 248002, Uttarakhand, India
| | - Kapil Kumar Goel
- Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to Be University), Haridwar, 249404, Uttarakhand, India
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Clement town, Dehradun, 248002, Uttarakhand, India
| | - Gaurav Joshi
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Garhwal, Srinagar, Uttarakhand, 246174, India; Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, 248002, Uttarakhand, India.
| | - Asim Kumar
- Amity Institute of Pharmacy (AIP), Amity University Haryana, Panchgaon, Manesar, 122413, India.
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Garhwal, Srinagar, Uttarakhand, 246174, India.
| |
Collapse
|
14
|
Nieratschker M, Liepins R, Honeder C, Auinger AB, Gausterer JC, Baumgartner WD, Riss D, Arnoldner C, Dahm V. A Single Intratympanic Triamcinolone Acetonide Administration Elicits Long-Term Reduction in Impedances Following Cochlear Implantation. J Otolaryngol Head Neck Surg 2024; 53:19160216241288819. [PMID: 39415405 PMCID: PMC11526200 DOI: 10.1177/19160216241288819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 07/27/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Intracochlear fibrosis and inflammation remain important limitations in cochlear implantation (CI). Glucocorticoids are routinely used to ameliorate the inflammatory response following CI. This study investigates the long-term effects of an intratympanically-applied triamcinolone-acetonide suspension on intracochlear impedance changes in CI recipients and investigates differences in drug concentrations and timepoints of injection. METHODS A total of 87 patients were included in the study, of whom 39 received an intratympanically-applied triamcinolone-acetonide suspension at either 10 or 40 mg/ml, 1 hour or 24 hours prior to cochlear implantation, while 48 patients served as an untreated control group. Electrode impedances were measured and compared over a period of 3 years following cochlear implantation. RESULTS The preoperative intratympanic application of a triamcinolone-acetonide suspension resulted in significantly lower mean impedances following cochlear implantation compared with an untreated control group at first fitting (4.66 ± 1.3 kΩ to 5.90 ± 1.4 kΩ, P = .0001), with sustained reduction observed over 3 months. A sustained reduction was observed after spatial grouping of the electrode contacts, with significant improvements in both the middle cochlear region over a 24 month period (from 3.97 ± 1.3 kΩ to 5.85 ± 1.3 kΩ, P = .049) and the basal region over a 6 month period (from 5.02 ± 1.3 kΩ to 5.85 ± 1.3 kΩ, P = .008). The injection of 10 mg/ml of triamcinolone-acetonide 1 hour prior to cochlear implantation resulted in higher impedances compared with 40 mg/ml and 24 hour time interval until surgery. CONCLUSION A single preoperative intratympanic injection of triamcinolone-acetonide significantly reduces electrode impedances across the entire cochlea. This effect is sustained for up to 2 years, after which impedances gradually equalize between the groups. A preoperative triamcinolone-acetonide injection could therefore be a favorable approach to attenuate the immediate tissue response following cochlear implantation.
Collapse
Affiliation(s)
- Michael Nieratschker
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Rudolfs Liepins
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Clemens Honeder
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Alice Barbara Auinger
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Julia Clara Gausterer
- Division of Pharmaceutical Technology and Biopharmaceutics, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Wolf-Dieter Baumgartner
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Dominik Riss
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Christoph Arnoldner
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| | - Valerie Dahm
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Dodonova SA, Zhidkova EM, Kryukov AA, Valiev TT, Kirsanov KI, Kulikov EP, Budunova IV, Yakubovskaya MG, Lesovaya EA. Synephrine and Its Derivative Compound A: Common and Specific Biological Effects. Int J Mol Sci 2023; 24:17537. [PMID: 38139366 PMCID: PMC10744207 DOI: 10.3390/ijms242417537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
This review is focused on synephrine, the principal phytochemical found in bitter orange and other medicinal plants and widely used as a dietary supplement for weight loss/body fat reduction. We examine different aspects of synephrine biology, delving into its established and potential molecular targets, as well as its mechanisms of action. We present an overview of the origin, chemical composition, receptors, and pharmacological properties of synephrine, including its anti-inflammatory and anti-cancer activity in various in vitro and animal models. Additionally, we conduct a comparative analysis of the molecular targets and effects of synephrine with those of its metabolite, selective glucocorticoid receptor agonist (SEGRA) Compound A (CpdA), which shares a similar chemical structure with synephrine. SEGRAs, including CpdA, have been extensively studied as glucocorticoid receptor activators that have a better benefit/risk profile than glucocorticoids due to their reduced adverse effects. We discuss the potential of synephrine usage as a template for the synthesis of new generation of non-steroidal SEGRAs. The review also provides insights into the safe pharmacological profile of synephrine.
Collapse
Affiliation(s)
- Svetlana A. Dodonova
- Research Institute of Experimental Medicine, Department of Pathophysiology, Kursk State Medical University, 305041 Kursk, Russia; (S.A.D.); (A.A.K.)
| | - Ekaterina M. Zhidkova
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (E.M.Z.); (T.T.V.); (K.I.K.); (M.G.Y.)
| | - Alexey A. Kryukov
- Research Institute of Experimental Medicine, Department of Pathophysiology, Kursk State Medical University, 305041 Kursk, Russia; (S.A.D.); (A.A.K.)
| | - Timur T. Valiev
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (E.M.Z.); (T.T.V.); (K.I.K.); (M.G.Y.)
| | - Kirill I. Kirsanov
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (E.M.Z.); (T.T.V.); (K.I.K.); (M.G.Y.)
- Faculty of Oncology, Ryazan State Medical University Named after Academician I.P. Pavlov, 390026 Ryazan, Russia
| | - Evgeny P. Kulikov
- Laboratory of Single Cell Biology, Russian University of People’s Friendship (RUDN) University, 117198 Moscow, Russia;
| | - Irina V. Budunova
- Department of Dermatology, Northwestern University, Chicago, IL 60611, USA;
| | - Marianna G. Yakubovskaya
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (E.M.Z.); (T.T.V.); (K.I.K.); (M.G.Y.)
- Faculty of Oncology, Ryazan State Medical University Named after Academician I.P. Pavlov, 390026 Ryazan, Russia
| | - Ekaterina A. Lesovaya
- Department of Chemical Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (E.M.Z.); (T.T.V.); (K.I.K.); (M.G.Y.)
- Faculty of Oncology, Ryazan State Medical University Named after Academician I.P. Pavlov, 390026 Ryazan, Russia
- Laboratory of Single Cell Biology, Russian University of People’s Friendship (RUDN) University, 117198 Moscow, Russia;
| |
Collapse
|
16
|
Akbari-Gharalari N, Ghahremani-Nasab M, Naderi R, Aliyari-Serej Z, Karimipour M, Shahabi P, Ebrahimi-Kalan A. Improvement of spinal cord injury symptoms by targeting the Bax/Bcl2 pathway and modulating TNF-α/IL-10 using Platelet-Rich Plasma exosomes loaded with dexamethasone. AIMS Neurosci 2023; 10:332-353. [PMID: 38188010 PMCID: PMC10767060 DOI: 10.3934/neuroscience.2023026] [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: 09/26/2023] [Revised: 11/12/2023] [Accepted: 11/16/2023] [Indexed: 01/09/2024] Open
Abstract
Spinal cord injury (SCI) is a debilitating condition that results in impaired sensory and motor function due to the limited self-regenerative ability of the spinal cord. To address this issue, combination therapy has been proposed as an effective treatment strategy for SCI regeneration. In this study, Platelet-Rich Plasma (PRP)-derived exosomes loaded with dexamethasone were utilized in a mouse model of SCI compression. PRP-derived exosomes loaded with dexamethasone (Dex) were prepared using ultracentrifugation and sonication methods and were administered to the mice via intravenous injection. Following a four-week duration, behavioral assessments were administered to assess functional recuperation, and diverse metrics encompassing the expression of genes associated with apoptosis and antiapoptosis, serum cytokine concentrations and tissue sampling were subjected to thorough examination. The results of this study demonstrated that mice treated with PRP-derived exosomes loaded with Dex (ExoDex) exhibited altered levels of TNF-α and IL-10, along with decreased Bax and increased Bcl2 expression in comparison to the model group. Furthermore, intravenously injected ExoDex reduced the size of the lesion site, lymphocyte infiltration, vacuolation, cavity size and tissue disorganization while also improving locomotor recovery. We propose that the utilization of exosome-loaded Dex therapy holds potential as a promising and clinically relevant approach for injured spinal cord repair. However, further extensive research is warranted in this domain to validate and substantiate the outcomes presented in this study.
Collapse
Affiliation(s)
- Naeimeh Akbari-Gharalari
- Department of Neurosciences and Cognition, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Ghahremani-Nasab
- Department of Tissue Engineering, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Naderi
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zeinab Aliyari-Serej
- Department of Applied Cell Sciences, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Shahabi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi-Kalan
- Department of Neurosciences and Cognition, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Cignarella A, Vegeto E, Bolego C, Trabace L, Conti L, Ortona E. Sex-oriented perspectives in immunopharmacology. Pharmacol Res 2023; 197:106956. [PMID: 37820857 DOI: 10.1016/j.phrs.2023.106956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/27/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
Several immunopharmacological agents are effective in the treatment of cancer and immune-mediated conditions, with a favorable impact on life expectancy and clinical outcomes for a large number of patients. Nevertheless, response variation and undesirable effects of these drugs represent major issues, and overall efficacy remains unpredictable. Males and females show a distinct difference in immune system responses, with females generally mounting stronger responses to a variety of stimuli. Therefore, exploring sex differences in the efficacy and safety of immunopharmacological agents would strengthen the practice of precision medicine. As a pharmacological target highlight, programmed cell death 1 ligand 1 (PD-L1) is the first functionally characterized ligand of the coinhibitory programmed death receptor 1 (PD-1). The PD-L1/PD-1 crosstalk plays an important role in the immune response and is relevant in cancer, infectious and autoimmune disease. Sex differences in the response to immune checkpoint inhibitors are well documented, with male patients responding better than female patients. Similarly, higher efficacy of and adherence to tumor necrosis factor inhibitors in chronic inflammatory conditions including rheumatoid arthritis and Crohn's disease have been reported in male patients. The pharmacological basis of sex-specific responses to immune system modulating drugs is actively investigated in other settings such as stroke and type 1 diabetes. Advances in therapeutics targeting the endothelium could soon be wielded against autoimmunity and metabolic disorders. Based on the established sexual dimorphism in immune-related pathophysiology and disease presentation, sex-specific immunopharmacological protocols should be integrated into clinical guidelines.
Collapse
Affiliation(s)
| | - Elisabetta Vegeto
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Lucia Conti
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Ortona
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
18
|
Sgambellone S, Febo M, Durante M, Marri S, Villano S, Bereshchenko O, Migliorati G, Masini E, Riccardi C, Bruscoli S, Lucarini L. Role of histamine H 4 receptor in the anti-inflammatory pathway of glucocorticoid-induced leucin zipper (GILZ) in a model of lung fibrosis. Inflamm Res 2023; 72:2037-2052. [PMID: 37815550 PMCID: PMC10611623 DOI: 10.1007/s00011-023-01802-3] [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: 04/26/2023] [Revised: 09/11/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023] Open
Abstract
INTRODUCTION This study investigates the interactions between histaminergic system and glucocorticoid-induced leucin zipper (GILZ) in the inflammatory process and glucocorticoid modulation in lung fibrosis. METHODS Wild-type (WT) and GILZ Knock-Out (KO) mice were treated with bleomycin (0.05 IU) or saline, delivered by intra-tracheal injection. After surgery, mice received a continuous infusion of JNJ7777120 (JNJ, 2 mg/kg b.wt.) or vehicle for 21 days. Lung function was studied by measuring airway resistance to air insufflation through the analysis of pressure at airway opening (PAO). Lung samples were collected to evaluate the expression of histamine H4R, Anx-A1, and p65-NF-kB, the activity of myeloperoxidase (MPO), and the production of pro-inflammatory cytokines. RESULTS Airway fibrosis and remodeling were assessed by measuring TGF-β production and α-SMA deposition. JNJ reduces PAO in WT but not in GILZ KO mice (from 22 ± 1 mm to 15 ± 0.5 and from 24 ± 1.5 to 19 ± 0.5 respectively), MPO activity (from 204 ± 3.13 pmol/mg to 73.88 ± 2.63 in WT and from 221 ± 4.46 pmol/mg to 107 ± 5.54 in GILZ KO), the inflammatory response, TGF-β production, and α-SMA deposition in comparison to WT and GILZ KO vehicle groups. CONCLUSION In conclusion, the role of H4R and GILZ in relation to glucocorticoids could pave the way for innovative therapies to counteract pulmonary fibrosis.
Collapse
Affiliation(s)
- Silvia Sgambellone
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Gaetano Pieraccini, 6, 50139, Florence, Italy
| | - Marta Febo
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, Piazzale Severi, 1 06132 S. Andrea Delle Fratte, Perugia, Italy
| | - Mariaconcetta Durante
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Gaetano Pieraccini, 6, 50139, Florence, Italy
| | - Silvia Marri
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Gaetano Pieraccini, 6, 50139, Florence, Italy
| | - Serafina Villano
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Gaetano Pieraccini, 6, 50139, Florence, Italy
| | - Oxana Bereshchenko
- Department of Philosophy, Social Sciences and Education, University of Perugia, 06100, Perugia, Italy
| | - Graziella Migliorati
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, Piazzale Severi, 1 06132 S. Andrea Delle Fratte, Perugia, Italy
| | - Emanuela Masini
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Gaetano Pieraccini, 6, 50139, Florence, Italy
| | - Carlo Riccardi
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, Piazzale Severi, 1 06132 S. Andrea Delle Fratte, Perugia, Italy
| | - Stefano Bruscoli
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, Piazzale Severi, 1 06132 S. Andrea Delle Fratte, Perugia, Italy
| | - Laura Lucarini
- Section of Pharmacology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Gaetano Pieraccini, 6, 50139, Florence, Italy.
| |
Collapse
|
19
|
Inceu AI, Neag MA, Catinean A, Bocsan CI, Craciun CI, Melincovici CS, Muntean DM, Onofrei MM, Pop RM, Buzoianu AD. The Effects of Probiotic Bacillus Spores on Dexamethasone-Treated Rats. Int J Mol Sci 2023; 24:15111. [PMID: 37894792 PMCID: PMC10606902 DOI: 10.3390/ijms242015111] [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: 09/15/2023] [Revised: 10/03/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Glucocorticoids are effective anti-inflammatory and immunosuppressive agents. Long-term exposure is associated with multiple metabolic side effects. Spore-forming probiotic bacteria have shown modulatory properties regarding glycolipid metabolism and inflammation. The aim of this study was to evaluate, for the first time, the effects of Bacillus species spores (B. licheniformis, B. indicus, B. subtilis, B. clausii, and B. coagulans) alone and in combination with metformin against dexamethasone-induced systemic disturbances. A total of 30 rats were randomly divided into 5 groups: group 1 served as control (CONTROL), group 2 received dexamethasone (DEXA), group 3 received DEXA and MegaSporeBiotic (MSB), group 4 received DEXA and metformin (MET), and group 5 received DEXA, MSB, and MET. On the last day of the experiment, blood samples and liver tissue samples for histopathological examination were collected. We determined serum glucose, total cholesterol, triglycerides, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-10 (IL-10), catalase, total antioxidant capacity (TAC), and metformin concentration. DEXA administration caused hyperglycemia and hyperlipidemia, increased inflammation cytokines, and decreased antioxidant markers. Treatment with MSB reduced total cholesterol, suggesting that the administration of Bacillus spores-based probiotics to DEXA-treated rats could ameliorate metabolic parameters.
Collapse
Affiliation(s)
- Andreea Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| | - Maria Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| | - Adrian Catinean
- Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
| | - Corina Ioana Bocsan
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| | - Cristian Ioan Craciun
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| | - Carmen Stanca Melincovici
- Department of Histology, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (C.S.M.); (M.M.O.)
| | - Dana Maria Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Iuliu Hatieganu University of Medicine and Pharmacy, 400010 Cluj-Napoca, Romania;
| | - Mădălin Mihai Onofrei
- Department of Histology, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (C.S.M.); (M.M.O.)
| | - Raluca Maria Pop
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| | - Anca Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| |
Collapse
|
20
|
Son Y, Kim BY, Kim M, Kim J, Kwon RJ, Kim K. Glucocorticoids Impair the 7α-Hydroxycholesterol-Enhanced Innate Immune Response. Immune Netw 2023; 23:e40. [PMID: 37970232 PMCID: PMC10643330 DOI: 10.4110/in.2023.23.e40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/17/2023] Open
Abstract
Glucocorticoids suppress the vascular inflammation that occurs under hypercholesterolemia, as demonstrated in an animal model fed a high-cholesterol diet. However, the molecular mechanisms underlying these beneficial effects remain poorly understood. Because cholesterol is oxidized to form cholesterol oxides (oxysterols) that are capable of inducing inflammation, we investigated whether glucocorticoids affect the immune responses evoked by 7α-hydroxycholesterol (7αOHChol). The treatment of human THP-1 monocytic cells with dexamethasone (Dex) and prednisolone (Pdn) downregulated the expression of pattern recognition receptors (PRRs), such as TLR6 and CD14, and diminished 7αOHChol-enhanced response to FSL-1, a TLR2/6 ligand, and lipopolysaccharide, which interacts with CD14 to initiate immune responses, as determined by the reduced secretion of IL-23 and CCL2, respectively. Glucocorticoids weakened the 7αOHChol-induced production of CCL2 and CCR5 ligands, which was accompanied by decreased migration of monocytic cells and CCR5-expressing Jurkat T cells. Treatment with Dex or Pdn also reduced the phosphorylation of the Akt-1 Src, ERK1/2, and p65 subunits. These results indicate that both Dex and Pdn impair the expression of PRRs and their downstream products, chemokine production, and phosphorylation of signaling molecules. Collectively, glucocorticoids suppress the innate immune response and activation of monocytic cells to an inflammatory phenotype enhanced or induced by 7αOHChol, which may contribute to the anti-inflammatory effects in hypercholesterolemic conditions.
Collapse
Affiliation(s)
- Yonghae Son
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Bo-Young Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Miran Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Jaesung Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Ryuk Jun Kwon
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
21
|
Hu S, Sun Q, Xu F, Jiang N, Gao J. Age-related hearing loss and its potential drug candidates: a systematic review. Chin Med 2023; 18:121. [PMID: 37730634 PMCID: PMC10512576 DOI: 10.1186/s13020-023-00825-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Age-related hearing loss (ARHL) is one of the main illnesses afflicting the aged population and has a significant negative impact on society, economy, and health. However, there is presently no appropriate therapeutic treatment of ARHL due to the absence of comprehensive trials. OBJECTIVES The goal of this review is to systematically evaluate and analyze recent statistics on the pathologic classifications, risk factors, treatment strategies, and drug candidates of ARHL, including that from traditional Chinese medicine (TCM), to provide potential new approaches for preventing and treating ARHL. METHODS Literature related to ARHL was conducted in databases such as PubMed, WOS, China National Knowledge Infrastructure (CNKI), and Wanfang from the establishment of the database to Jan, 2023. The pathology, causal factor, pathophysiological mechanism, treatment strategy, and the drug candidate of ARHL were extracted and pooled for synthesis. RESULTS Many hypotheses about the etiology of ARHL are based on genetic and environmental elements. Most of the current research on the pathology of ARHL focuses on oxidative damage, mitochondrial dysfunction, inflammation, cochlear blood flow, ion homeostasis, etc. In TCM, herbs belonging to the kidney, lung, and liver meridians exhibit good hearing protection. Seven herbs belonging to the kidney meridian, 9 belonging to the lung meridian, and 4 belonging to the liver meridian were ultimately retrieved in this review, such as Polygonum multiflorum Thunb., Panax ginseng C.A. Mey, and Pueraria lobata (Willd.) Ohwi. Their active compounds, 2,3,4',5-Tetrahydroxystilbene-2-O-D-glucoside, ginsenoside Rb1, and puerarin, may act as the molecular substance for their anti-ARHL efficacy, and show anti-oxidative, neuroprotective, anti-inflammatory, anti-apoptotic, or mitochondrial protective effects. CONCLUSION Anti-oxidants, modulators of mitochondrial function, anti-inflammation agents, vasodilators, K+ channel openers, Ca2+ channel blockers, JNK inhibitors, and nerve growth factors/neurotrophic factors all contribute to hearing protection, and herbs are an important source of potential anti-ARHL drugs.
Collapse
Affiliation(s)
- Shiyu Hu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Qingru Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Fei Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China
| | - Ninghua Jiang
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, People's Republic of China
| | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| |
Collapse
|
22
|
Paglialunga M, Flamini S, Contini R, Febo M, Ricci E, Ronchetti S, Bereshchenko O, Migliorati G, Riccardi C, Bruscoli S. Anti-Inflammatory Effects of Synthetic Peptides Based on Glucocorticoid-Induced Leucine Zipper (GILZ) Protein for the Treatment of Inflammatory Bowel Diseases (IBDs). Cells 2023; 12:2294. [PMID: 37759516 PMCID: PMC10528232 DOI: 10.3390/cells12182294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Glucocorticoids (GCs) are commonly used to treat autoimmune and inflammatory diseases, but their clinical effects and long-term use can lead to serious side effects. New drugs that can replace GCs are needed. Glucocorticoid-induced leucine zipper (GILZ) is induced by GCs and mediates many of their anti-inflammatory effects, such as inhibiting the pro-inflammatory molecule NF-κB. The GILZ C-terminal domain (PER region) is responsible for GILZ/p65NF-κB interaction and consequent inhibition of its transcriptional activity. A set of five short peptides spanning different parts of the PER region of GILZ protein was designed, and their anti-inflammatory activity was tested, both in vitro and in vivo. We tested the biological activity of GILZ peptides in human lymphocytic and monocytic cell lines to evaluate their inhibitory effect on the NF-κB-dependent expression of pro-inflammatory cytokines. Among the tested peptides, the peptide named PEP-1 demonstrated the highest efficacy in inhibiting cell activation in vitro. Subsequently, PEP-1 was further evaluated in two in vivo experimental colitis models (chemically induced by DNBS administration and spontaneous colitis induced in IL-10 knock-out (KO) mice (to assess its effectiveness in counteracting inflammation. Results show that PEP-1 reduced disease severity in both colitis models associated with reduced NF-κB pro-inflammatory activity in colon lamina propria lymphocytes. This study explored GILZ-based 'small peptides' potential efficacy in decreasing lymphocyte activation and inflammation associated with experimental inflammatory bowel diseases (IBDs). Small peptides have several advantages over the entire protein, including higher selectivity, better stability, and bioavailability profile, and are easy to synthesize and cost-effective. Thus, identifying active GILZ peptides could represent a new class of drugs for treating IBD patients.
Collapse
Affiliation(s)
- Musetta Paglialunga
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.F.); (R.C.); (M.F.); (E.R.); (S.R.); (G.M.); (C.R.)
| | - Sara Flamini
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.F.); (R.C.); (M.F.); (E.R.); (S.R.); (G.M.); (C.R.)
| | - Raffaele Contini
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.F.); (R.C.); (M.F.); (E.R.); (S.R.); (G.M.); (C.R.)
| | - Marta Febo
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.F.); (R.C.); (M.F.); (E.R.); (S.R.); (G.M.); (C.R.)
| | - Erika Ricci
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.F.); (R.C.); (M.F.); (E.R.); (S.R.); (G.M.); (C.R.)
| | - Simona Ronchetti
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.F.); (R.C.); (M.F.); (E.R.); (S.R.); (G.M.); (C.R.)
| | - Oxana Bereshchenko
- Department of Philosophy, Social Sciences and Education, University of Perugia, 06123 Perugia, Italy;
| | - Graziella Migliorati
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.F.); (R.C.); (M.F.); (E.R.); (S.R.); (G.M.); (C.R.)
| | - Carlo Riccardi
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.F.); (R.C.); (M.F.); (E.R.); (S.R.); (G.M.); (C.R.)
| | - Stefano Bruscoli
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy; (M.P.); (S.F.); (R.C.); (M.F.); (E.R.); (S.R.); (G.M.); (C.R.)
| |
Collapse
|
23
|
Liu Z, Chen H, Tan C, Zha J, Liu H, Chen G. Activation of CD3+TIM3+ T Cells Contributes to Excessive Inflammatory Response During Glucocorticoid Treatment. Biochem Pharmacol 2023; 212:115551. [PMID: 37044297 DOI: 10.1016/j.bcp.2023.115551] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023]
Abstract
Glucocorticoids (GCs) are widely used to treat autoimmune and inflammatory diseases, but recent research has challenged the notion that GCs are universally anti-inflammatory. In this study, we investigated the effects of long-term GC exposure on circulating T cells in a retrospective cohort of 5,476 patients with primary glomerular diseases. Our results revealed that GCs altered the composition pattern of circulating leukocytes and the correlation between circulating lymphocytes and serum cytokines in response to infections, as well as the subsets of CD4+ T cells. Specifically, GCs promoted the loss of CD4+ T cells and increased the proportions of CD3+TIM3+ T cells in response to infections, which correlated with the expression of serum inflammatory cytokines, such as IFNG and IL-10. Using animal models of cecal ligation and puncture, we demonstrated that long-term GC exposure exacerbated apoptosis of CD4+ T cells and cytokine storm during sepsis, which was mechanistically linked to the increase of CD3+TIM3+ T cells. Notably, we found that CD3+TIM3+ T cells expressed high levels of multiple cytokine genes during infections, suggesting a potent role of TIM3 in the regulation of T cell biology. In vitro studies further showed that engagement of anti-TIM3 treatment enhanced the inflammatory activity of CD3+ T cells. Our findings suggest a causal relationship between chronic exposure to GCs and an excessive inflammatory response mediated by T cells during infections, which is, at least partly, driven by dysregulation of CD3+TIM3+ T cells.
Collapse
Affiliation(s)
- Zhiwen Liu
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Huihui Chen
- Clinical Immunology Research Center of Central South University, Changsha, China; Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Chongqing Tan
- Department of Pharmacy, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Jie Zha
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Hong Liu
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Guochun Chen
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China; Clinical Immunology Research Center of Central South University, Changsha, China.
| |
Collapse
|
24
|
Systemic glucocorticoids as an adjunct to treatment of postoperative pain after total hip and knee arthroplasty: A systematic review with meta-analysis and trial sequential analysis. Ugeskr Laeger 2023; 40:155-170. [PMID: 36325886 DOI: 10.1097/eja.0000000000001768] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Postoperative analgesic effects of systemic glucocorticoids given as an adjunct to treatment are largely undetermined in alloplastic procedures. OBJECTIVES To investigate the beneficial and harmful effects of peri-operative systemic glucocorticoid treatment for pain after total hip arthroplasty (THA) or total knee arthroplasty (TKA). DESIGN A systematic review of randomised clinical trials (RCTs) with meta-analyses, trial sequential analyses and GRADE. Primary outcome was 24 h intravenous (i.v.) morphine (or equivalent) consumption with a predefined minimal important difference (MID) of 5 mg. Secondary outcomes included pain at rest and during mobilisation (MID, VAS 10 mm), adverse and serious adverse events (SAEs). DATA SOURCES We searched EMBASE, Cochrane CENTRAL, PubMed and Google Scholar up to October 2021. ELIGIBILITY CRITERIA RCTs investigating peri-operative systemic glucocorticoid versus placebo or no intervention, for analgesic pain management of patients at least 18 years undergoing planned THA or TKA, irrespective of publication date and language. RESULTS We included 32 RCTs with 3521 patients. Nine trials were at a low risk of bias. Meta-analyses showed evidence of a reduction in 24 h cumulative morphine consumption with glucocorticoids by 5.0 mg (95% CI 2.2 to 7.7; P = 0.0004). Pain at rest was reduced at 6 h by 7.8 mm (95% CI 5.5 to 10.2; P < 0.00001), and at 24 h by 6.3 mm (95% CI 3.8 to 8.8; P < 0.00001). Pain during mobilisation was reduced at 6 h by 9.8 mm (95% CI 6.9 to 12.8; P < 0.00001), and at 24 h by 9.0 mm (95% CI 5.5 to 12.4, P < 0.00001). Incidence of adverse events was generally lower in the glucocorticoid treatment group. SAEs were rarely reported. The GRADE rated quality of evidence was low to very low. CONCLUSION Peri-operative systemic glucocorticoid treatment reduced postoperative morphine consumption to an individually relevant level following hip and knee arthroplasty. Pain levels were reduced but were below the predefined MID. The quality of evidence was generally low. REGISTRATION PROSPERO ID: CRD42019135034.
Collapse
|
25
|
Zhang H, Yan J, Nie G, Xie D, Luo B, Niu J, Wang H, Li X. Effects of cadmium and lead co-exposure on glucocorticoid levels in rural residents of northwest China. CHEMOSPHERE 2023; 317:137783. [PMID: 36638928 DOI: 10.1016/j.chemosphere.2023.137783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 06/17/2023]
Abstract
Cadmium (Cd) and lead (Pb) are important environmental endocrine disruptors that are associated with adverse health problems. However, the effects of co-exposure to Cd and Pb on glucocorticoids (GCs) in the body at environmental levels are limited. A total of 468 subjects from the Dongdagou-Xinglong cohort (DDG-XL) were included in this study. We measured the serum levels of two representative endogenous GCs [cortisol (CRL) and cortisone (CRN)], and whole blood levels of Cd and Pb. Multiple linear regression models were constructed to explore the associations of single or combined Cd and Pb exposure with serum CRL and CRN levels. The interactive effects of Cd and Pb on GCs were further assessed using mediation analysis and moderation analysis. Single-heavy metal exposure analysis with adjustment for potential confounders showed that the serum CRL level decreased when the blood Cd or Pb concentration gradually increased (P trend <0.01). Additionally, subjects with high Cd or Pb exposure (Q4) had significantly reduced serum CRN levels compared to those with low Cd or Pb exposure (Q1) (P < 0.05). In Cd and Pb co-exposure analysis, significant negative dose-response relationships were observed between co-exposure to Cd and Pb and serum CRL and CRN levels. Furthermore, mediation analysis showed that Cd completely mediated the relationship between Pb and GCs, and moderation analysis suggested that Pb might weaken the negative relationship between Cd and GCs. These findings suggest that single or combined exposure to Cd and Pb interferes with the homeostasis of serum CRL and CRN levels. Furthermore, we innovatively propose that Cd and Pb may have interactive effects on GCs levels, and Pb can antagonize the negative relationship between Cd and GCs, which may provide clues for further studies on endocrine and metabolic disorders related to these heavy metals.
Collapse
Affiliation(s)
- Honglong Zhang
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jun Yan
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China; Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, Gansu, People's Republic of China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Medical College Cancer Center of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Guole Nie
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Danna Xie
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Bin Luo
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Jingping Niu
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Haiping Wang
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China; Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, Gansu, People's Republic of China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Medical College Cancer Center of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Xun Li
- The First School of Clinical Medical, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China; Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, 730000, Gansu, People's Republic of China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Medical College Cancer Center of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
26
|
Prednisone acetate modulates Th1/Th2 and Th17/Treg cell homeostasis in experimental autoimmune uveitis via orchestrating the Notch signaling pathway. Int Immunopharmacol 2023; 116:109809. [PMID: 36753985 DOI: 10.1016/j.intimp.2023.109809] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/09/2023]
Abstract
Uveitis is an immune eye disease that can seriously impair vision. Glucocorticoids (GCS) have been extensively used to treat uveitis, though the mechanisms have not been fully elucidated. In this study, we investigated the regulatory effects of prednisone acetate (PA) on the Th1/Th2 and Th17/Treg balance in experimental autoimmune uveitis (EAU) through modulating the Notch signaling pathway. Briefly, Lewis rats were randomly divided into the normal control (NC), EAU, and EAU + PA groups. Rats in EAU and EAU + PA groups were induced EAU, while those in the EAU + PA group were treated with PA. Clinical and histopathological scores were employed to assess the progression of EAU. The expression levels of Notch signaling-related molecules (Notch1, Notch2, Dll3, Dll4, and Rbpj) and Th-associated cytokines (IFN-γ, IL-4, IL-10, and IL-17) were assessed via quantitative PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA). In addition, the frequencies of Th1, Th2, Th17 and Treg cells were detected by flow cytometry. These experimental results indicated that activation of the Notch signaling pathway occurred in EAU rats and resulted in a severe imbalance of the Th17/Treg and Th1/Th2 ratios. PA treatment significantly alleviated ocular inflammation, inhibited activation of the Notch signaling pathway, and declined Th1, and Th17 cell differentiation, thereby restoring the Th1/Th2 and Th17/Treg balance. Collectively, PA can positively enhance the systemic immune response and improve the intraocular microenvironmental homeostasis by inhibiting activation of the Notch signaling pathway and by restoring Th1/Th2 and Th17/Treg balance, thus achieving the goal of treating uveitis.
Collapse
|
27
|
Canali MM, Guyot M, Simon T, Daoudlarian D, Chabry J, Panzolini C, Petit-Paitel A, Hypolite N, Nicolas S, Bourdely P, Schmid-Antomarchi H, Schmid-Alliana A, Soria J, Karimdjee Soilihi B, Hofman P, Prevost-Blondel A, Kato M, Mougneau E, Glaichenhaus N, Blancou P. Environmental signals perceived by the brain abate pro-metastatic monocytes by dampening glucocorticoids receptor signaling. Cancer Cell Int 2023; 23:15. [PMID: 36726173 PMCID: PMC9893572 DOI: 10.1186/s12935-023-02855-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/19/2023] [Indexed: 02/03/2023] Open
Abstract
While positive social-behavioral factors predict longer survival in cancer patients, the underlying mechanisms are unknown. Since tumor metastasis are the major cancer mortality factor, we investigated how an enriched environment (EE) conductive to enhanced sensory, cognitive and motor stimulation impact metastatic progression in lungs following intravasation in the circulation. We find that mice housed in EE exhibited reduced number of lung metastatic foci compared to control mice housed in a standard environment (SE). Compared to SE mice, EE mice increased lung inflammation as early as 4 days after circulating tumor cells extravasation. The impact of environmental signals on lung metastasis is independent of adrenergic receptors signaling. By contrast, we find that serum corticosterone levels are lower in EE mice and that glucocorticoid receptor (GR) antagonist reduces the number of lung metastasis in SE mice. In addition, the difference of the number of lung metastasis between SE and EE mice is abolished when inflammatory monocytes are rendered deficient in GR signaling. This decreased GR signaling in inflammatory monocytes of SE mice results in an exacerbated inflammatory profile in the lung. Our study shows that not only EE reduces late stages of metastatic progression in lungs but disclose a novel anti-tumor mechanism whereby GR-dependent reprogramming of inflammatory monocytes can inhibit metastatic progression in lungs. Moreover, while inflammatory monocytes have been shown to promote cancer progression, they also have an anti-tumor effect, suggesting that their role is more complex than currently thought.
Collapse
Affiliation(s)
- María Magdalena Canali
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Mélanie Guyot
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Thomas Simon
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Douglas Daoudlarian
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Joelle Chabry
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Clara Panzolini
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Agnès Petit-Paitel
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Nicolas Hypolite
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Sarah Nicolas
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Pierre Bourdely
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Heidy Schmid-Antomarchi
- grid.460782.f0000 0004 4910 6551Université Côte d’Azur, CNRS, INSERM, Valrose Biology Institute, 28 Avenue de Valombrose, Nice, France
| | - Annie Schmid-Alliana
- grid.460782.f0000 0004 4910 6551Université Côte d’Azur, CNRS, INSERM, Valrose Biology Institute, 28 Avenue de Valombrose, Nice, France
| | - Javier Soria
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Babou Karimdjee Soilihi
- grid.460782.f0000 0004 4910 6551Université Côte d’Azur, CNRS, INSERM, Valrose Biology Institute, 28 Avenue de Valombrose, Nice, France ,Polyclinique Saint Jean, Cagnes sur mer, France
| | - Paul Hofman
- grid.410528.a0000 0001 2322 4179Laboratory of Clinical and Experimental Pathology and Biobank, Nice University Hospital, Nice, France ,grid.460782.f0000 0004 4910 6551Research Institute on Cancer and Aging, Université Côte d’Azur, CNRS, INSERM, 28 Avenue de Valombrose, Nice, France
| | - Armelle Prevost-Blondel
- grid.462098.10000 0004 0643 431XUniversité Paris Descartes, CNRS, INSERM, Institut Cochin, 22 rue Méchain, 75014 Paris, France
| | - Masashi Kato
- grid.27476.300000 0001 0943 978XDepartment of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya, Aichi Japan
| | - Evelyne Mougneau
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Nicolas Glaichenhaus
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| | - Philippe Blancou
- grid.460782.f0000 0004 4910 6551Molecular and Cellular Pharmacology Institute, Université Côte d’Azur, CNRS, 660 Route des Lucioles, Valbonne, France
| |
Collapse
|
28
|
Silva LP, Santos IP, Silva DKC, dos Reis BPZC, Meira CS, Castro MVBDS, dos Santos Filho JM, de Araujo-Neto JH, Ellena JA, da Silveira RG, Soares MBP. Molecular Hybridization Strategy on the Design, Synthesis, and Structural Characterization of Ferrocene- N-acyl Hydrazones as Immunomodulatory Agents. Molecules 2022; 27:molecules27238343. [PMID: 36500436 PMCID: PMC9737981 DOI: 10.3390/molecules27238343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022] Open
Abstract
Immunomodulatory agents are widely used for the treatment of immune-mediated diseases, but the range of side effects of the available drugs makes necessary the search for new immunomodulatory drugs. Here, we investigated the immunomodulatory activity of new ferrocenyl-N-acyl hydrazones derivatives (SintMed(141−156). The evaluated N-acyl hydrazones did not show cytotoxicity at the tested concentrations, presenting CC50 values greater than 50 µM. In addition, all ferrocenyl-N-acyl hydrazones modulated nitrite production in immortalized macrophages, showing inhibition values between 14.4% and 74.2%. By presenting a better activity profile, the ferrocenyl-N-acyl hydrazones SintMed149 and SintMed150 also had their cytotoxicity and anti-inflammatory effect evaluated in cultures of peritoneal macrophages. The molecules were not cytotoxic at any of the concentrations tested in peritoneal macrophages and were able to significantly reduce (p < 0.05) the production of nitrite, TNF-α, and IL-1β. Interestingly, both molecules significantly reduced the production of IL-2 and IFN-γ in cultured splenocytes activated with concanavalin A. Moreover, SintMed150 did not show signs of acute toxicity in animals treated with 50 or 100 mg/kg. Finally, we observed that ferrocenyl-N-acyl hydrazone SintMed150 at 100 mg/kg reduced the migration of neutrophils (44.6%) in an acute peritonitis model and increased animal survival by 20% in an LPS-induced endotoxic shock model. These findings suggest that such compounds have therapeutic potential to be used to treat diseases of inflammatory origin.
Collapse
Affiliation(s)
- Laís Peres Silva
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil
| | - Ivanilson Pimenta Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
| | | | | | - Cássio Santana Meira
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
- Institute for Innovation in Advanced Health Systems (CIMATEC ISI SAS—University Center SENAI/CIMATEC), Salvador 41650-010, BA, Brazil
| | - Marcos Venícius Batista de Souza Castro
- Laboratory of Design and Synthesis Applied to Medicinal Chemistry-SintMed®, Center for Technology and Geosciences, Federal University of Pernambuco, Recife 50740-521, PE, Brazil
| | - José Maurício dos Santos Filho
- Laboratory of Design and Synthesis Applied to Medicinal Chemistry-SintMed®, Center for Technology and Geosciences, Federal University of Pernambuco, Recife 50740-521, PE, Brazil
| | - João Honorato de Araujo-Neto
- Multiuser Laboratory of Structural Crystallography, Institute of São Carlos, University of São Paulo, São Carlos 13566-590, SP, Brazil
| | - Javier Alcides Ellena
- Multiuser Laboratory of Structural Crystallography, Institute of São Carlos, University of São Paulo, São Carlos 13566-590, SP, Brazil
| | - Rafael Gomes da Silveira
- Multiuser Laboratory of Structural Crystallography, Institute of São Carlos, University of São Paulo, São Carlos 13566-590, SP, Brazil
- Department of Chemistry, Federal Institute of Goiás, Campus Ceres, Ceres 76300-000, GO, Brazil
| | - Milena Botelho Pereira Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
- Institute for Innovation in Advanced Health Systems (CIMATEC ISI SAS—University Center SENAI/CIMATEC), Salvador 41650-010, BA, Brazil
- Correspondence:
| |
Collapse
|
29
|
Bruscoli S, Puzzovio PG, Zaimi M, Tiligada K, Levi-Schaffer F, Riccardi C. Glucocorticoids and COVID-19. Pharmacol Res 2022; 185:106511. [PMID: 36243331 PMCID: PMC9556882 DOI: 10.1016/j.phrs.2022.106511] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/10/2022] [Accepted: 10/10/2022] [Indexed: 12/15/2022]
Abstract
Coronavirus Disease 19 (COVID-19) is associated with high morbidity and mortality rates globally, representing the greatest health and economic challenge today. Several drugs are currently approved for the treatment of COVID-19. Among these, glucocorticoids (GCs) have received particular attention due to their anti-inflammatory and immunosuppressive effects. In fact, GC are widely used in current clinical practice to treat inflammatory, allergic and autoimmune diseases. Major mechanisms of GC action include inhibition of innate and adaptive immune activity. In particular, an important role is played by the inhibition of pro-inflammatory cytokines and chemokines, and the induction of proteins with anti-inflammatory activity. Overall, as indicated by various national and international regulatory agencies, GCs are recommended for the treatment of COVID-19 in patients requiring oxygen therapy, with or without mechanical ventilation. Regarding the use of GCs for the COVID-19 treatment of non-hospitalized patients at an early stage of the disease, many controversial studies have been reported and regulatory agencies have not recommended their use. The decision to start GC therapy should be based not only on the severity of COVID-19 disease, but also on careful considerations of the benefit/risk profile in individual patients, including monitoring of adverse events. In this review we summarize the effects of GCs on the major cellular and molecular components of the inflammatory/immune system, the benefits and the adverse common reactions in the treatment of inflammatory/autoimmune diseases, as well as in the management of COVID-19.
Collapse
Affiliation(s)
- Stefano Bruscoli
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Pier Giorgio Puzzovio
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maria Zaimi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Katerina Tiligada
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Francesca Levi-Schaffer
- Pharmacology and Experimental Therapeutics Unit, School of Pharmacy, Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Carlo Riccardi
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, Perugia, Italy.
| |
Collapse
|
30
|
Wang X, Tang M, Zhang Y, Li Y, Mao J, Deng Q, Li S, Jia Z, Du L. Dexamethasone enhances glucose uptake by SGLT1 and GLUT1 and boosts ATP generation through the PPP-TCA cycle in bovine neutrophils. J Vet Sci 2022; 23:e76. [PMID: 36174980 PMCID: PMC9523333 DOI: 10.4142/jvs.22112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/18/2022] [Accepted: 07/26/2022] [Indexed: 11/20/2022] Open
Abstract
Background Clinical dexamethasone (DEX) treatment or stress in bovines results in extensive physiological changes with prominent hyperglycemia and neutrophils dysfunction. Objectives To elucidate the effects of DEX treatment in vivo on cellular energy status and the underlying mechanism in circulating neutrophils. Methods We selected eight-month-old male bovines and injected DEX for 3 consecutive days (1 time/d). The levels of glucose, total protein (TP), total cholesterol (TC), and the proinflammatory cytokines interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α in blood were examined, and we then detected glycogen and adenosine triphosphate (ATP) content, phosphofructosekinase-1 (PFK1) and glucose-6-phosphate dehydrogenase (G6PDH) activity, glucose transporter (GLUT)1, GLUT4, sodium/glucose cotransporter (SGLT)1 and citrate synthase (CS) protein expression and autophagy levels in circulating neutrophils. Results DEX injection markedly increased blood glucose, TP and TC levels, the Ca2+/P5+ ratio and the neutrophil/lymphocyte ratio and significantly decreased blood IL-1β, IL-6 and TNF-α levels. Particularly in neutrophils, DEX injection inhibited p65-NFκB activation and elevated glycogen and ATP contents and SGLT1, GLUT1 and GR expression while inhibiting PFK1 activity, enhancing G6PDH activity and CS expression and lowering cell autophagy levels. Conclusions DEX induced neutrophils glucose uptake by enhancing SGLT1 and GLUT1 expression and the transformation of energy metabolism from glycolysis to pentose phosphate pathway (PPP)-tricarboxylic acid (TCA) cycle. This finding gives us a new perspective on deeper understanding of clinical anti-inflammatory effects of DEX on bovine.
Collapse
Affiliation(s)
- Xinbo Wang
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Mingyu Tang
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Yuming Zhang
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China.,Key Laboratory for Prevention and Control of Herbivorous Livestock Perinatal Diseases, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Yansong Li
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Jingdong Mao
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Qinghua Deng
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China.,Key Laboratory for Prevention and Control of Herbivorous Livestock Perinatal Diseases, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Shusen Li
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Zhenwei Jia
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Liyin Du
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China.,Key Laboratory for Prevention and Control of Herbivorous Livestock Perinatal Diseases, Inner Mongolia Minzu University, Tongliao 028000, China.
| |
Collapse
|
31
|
Cole AB, Montgomery K, Bale TL, Thompson SM. What the hippocampus tells the HPA axis: Hippocampal output attenuates acute stress responses via disynaptic inhibition of CRF+ PVN neurons. Neurobiol Stress 2022; 20:100473. [PMID: 35982732 PMCID: PMC9379952 DOI: 10.1016/j.ynstr.2022.100473] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/11/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
The hippocampus exerts inhibitory feedback on the release of glucocorticoids. Because the major hippocampal efferent projections are excitatory, it has been hypothesized that this inhibition is mediated by populations of inhibitory neurons in the hypothalamus or elsewhere. These regions would be excited by hippocampal efferents and project to corticotropin-releasing factor (CRF) cells in the paraventricular nucleus of the hypothalamus (PVN). A direct demonstration of the synaptic responses elicited by hippocampal outputs in PVN cells or upstream GABAergic interneurons has not been provided previously. Here, we used viral vectors to express channelrhodopsin (ChR) and enhanced yellow fluorescent protein (EYFP) in pyramidal cells in the ventral hippocampus (vHip) in mice expressing tdTomato in GABA- or CRF-expressing neurons. We observed dense innervation of the bed nucleus of the stria terminalis (BNST) by labeled vHip axons and sparse labeling within the PVN. Using whole-cell voltage-clamp recording in parasagittal brain slices containing the BNST and PVN, photostimulation of vHip terminals elicited rapid excitatory postsynaptic currents (EPSCs) and longer-latency inhibitory postsynaptic currents (IPSCs) in both CRF+ and GAD + cells. The ratio of synaptic excitation and inhibition was maintained in CRF + cells during 20 Hz stimulus trains. Photostimulation of hippocampal afferents to the BNST and PVN in vivo inhibited the rise in blood glucocorticoid levels produced by acute restraint stress. We thus provide functional evidence suggesting that hippocampal output to the BNST contributes to a net inhibition of the hypothalamic-pituitary axis, providing further mechanistic insights into this process using methods with enhanced spatial and temporal resolution.
Collapse
Affiliation(s)
- Anthony B. Cole
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Medical Scientist Training Program, Departments of University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Kristen Montgomery
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Tracy L. Bale
- Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Scott M. Thompson
- Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD, USA
| |
Collapse
|
32
|
Shafiei SH, Siavashi B, Ghasemi M, Golbakhsh MR, Baghdadi S. Single High-Dose Systemic Methylprednisolone Administered Preoperatively Improves Pain Control and Sleep Quality After Total Hip Arthroplasty: A Double-Blind, Randomized Controlled Trial. Arthroplast Today 2022; 16:78-82. [PMID: 35662994 PMCID: PMC9160665 DOI: 10.1016/j.artd.2022.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 11/29/2022] Open
Abstract
Background This study was performed to evaluate the efficacy of preoperative high-dose methylprednisolone on pain levels and sleep quality following primary total hip arthroplasty. Material and methods A double-blind, randomized controlled trial was performed in adults underdoing total hip arthroplasty. A computer-generated, permuted-block randomization scheme with a 1:1 ratio between the intervention (125 mg methylprednisolone) and control groups (normal saline) was utilized. Patients underwent a similar preoperative and postoperative protocol. Pain was assessed using the visual analog scale (VAS). Sleep quality was assessed at the 2-week postoperative visit using the Pittsburgh Sleep Quality Index (PSQI). Results With a total of 70 patients, 35 patients were included in the intervention and placebo groups. Hospital stay was significantly shorter in the intervention group (1.5 ± 0.7 vs 2.0 ± 0.5 days, P = .03). Preoperative pain levels were similar between groups, while satisfactory pain control was achieved in a significantly larger number of patients in the intervention group (18 vs 8 patients, P = .009). The intervention group was significantly more likely to have a good sleep quality than the placebo groups (74% vs 31%, P = .001). No significant differences were found between preoperative and postoperative blood sugar levels. We did not observe any cases of early postoperative wound complication, infection, or deep vein thrombosis among our patients. Conclusion In this randomized controlled trial, preoperative administration of 125 mg of methylprednisolone was found to improve pain control, as measured by VAS, 24 hours after surgery, and sleep quality, as measured by PSQI, 2 weeks following surgery.
Collapse
Affiliation(s)
- Seyyed Hossein Shafiei
- Orthopedic Surgery Research Centre, Sina University Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Siavashi
- Orthopedic Surgery Research Centre, Sina University Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Ghasemi
- Orthopedic Surgery Research Centre, Sina University Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Golbakhsh
- Orthopedic Surgery Research Centre, Sina University Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soroush Baghdadi
- Joint Reconstruction Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Corresponding author. Joint Reconstruction Research Center, End of Keshavarz Blvd, Tehran, Iran. Tel.: +98 21 66581586.
| |
Collapse
|
33
|
Leoncini G, Gentili M, Lusenti E, Caruso L, Calafà C, Migliorati G, Riccardi C, Villanacci V, Ronchetti S. The novel role of glucocorticoid-induced leucine zipper as a marker of mucosal healing in inflammatory bowel diseases. Pharmacol Res 2022; 182:106353. [PMID: 35835370 DOI: 10.1016/j.phrs.2022.106353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/22/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
Glucocorticoid-induced leucin zipper (GILZ) mediates the effects of glucocorticoids in immune cells, but little is known about its role in both the gastro-intestinal (GI) mucosa and inflammatory bowel diseases (IBD) in humans. To investigate the GILZ protein expression profile in the GI tract, mucosal biopsies from 80 patients were retrospectively enrolled in this study and subdivided into three groups: 1) patients without clinical-endoscopic and histological evidence of IBD; 2) IBD patients; 3) patients with chronic atrophic gastritis (CAG) and Barrett esophagus (BE), both characterized by intestinal metaplasia (IM). GILZ expression was assessed by immunohistochemical and immunofluorescence methods. Our results showed that GILZ protein was strongly expressed in the secretory cells in healthy mucosa. GILZ expression was reduced in goblet cells in active disease, whereas it was restored in quiescent diseases. Conversely, entero-endocrine cells were not involved in such inflammation-driven dynamics, as GILZ expression remained detectable in active disease. Moreover, GILZ was expressed in IM, but was limited to CAG, and was not detected in BE. In summary, GILZ acts as a secretory protein in the GI mucosa in healthy, hyperplastic and metaplastic conditions. Its secretion by goblet cells is mostly affected by neutrophils mucosal infiltration and seems to be directly related to active mucosal inflammation in IBD. Overall, our findings suggest that GILZ is a suitable molecule to be considered as a histological marker of mucosal healing.
Collapse
Affiliation(s)
- Giuseppe Leoncini
- Pathology Unit, Department of Pathology and Laboratory Medicine, ASST del Garda Desenzano del Garda, Brescia, Italy
| | - Marco Gentili
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Eleonora Lusenti
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Laura Caruso
- Pathology Unit, Department of Pathology and Laboratory Medicine, ASST del Garda Desenzano del Garda, Brescia, Italy
| | - Cristina Calafà
- Pathology Unit, Department of Pathology and Laboratory Medicine, ASST del Garda Desenzano del Garda, Brescia, Italy
| | - Graziella Migliorati
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Simona Ronchetti
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
34
|
Amratia DA, Viola H, Ioachimescu OC. Glucocorticoid therapy in respiratory illness: bench to bedside. J Investig Med 2022; 70:1662-1680. [PMID: 35764344 DOI: 10.1136/jim-2021-002161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2022] [Indexed: 11/07/2022]
Abstract
Each year, hundreds of millions of individuals are affected by respiratory disease leading to approximately 4 million deaths. Most respiratory pathologies involve substantially dysregulated immune processes that either fail to resolve the underlying process or actively exacerbate the disease. Therefore, clinicians have long considered immune-modulating corticosteroids (CSs), particularly glucocorticoids (GCs), as a critical tool for management of a wide spectrum of respiratory conditions. However, the complex interplay between effectiveness, risks and side effects can lead to different results, depending on the disease in consideration. In this comprehensive review, we present a summary of the bench and the bedside evidence regarding GC treatment in a spectrum of respiratory illnesses. We first describe here the experimental evidence of GC effects in the distal airways and/or parenchyma, both in vitro and in disease-specific animal studies, then we evaluate the recent clinical evidence regarding GC treatment in over 20 respiratory pathologies. Overall, CS remain a critical tool in the management of respiratory illness, but their benefits are dependent on the underlying pathology and should be weighed against patient-specific risks.
Collapse
|
35
|
Liao S, Luo J, Kadier T, Ding K, Chen R, Meng Q. Mitochondrial DNA Release Contributes to Intestinal Ischemia/Reperfusion Injury. Front Pharmacol 2022; 13:854994. [PMID: 35370747 PMCID: PMC8966724 DOI: 10.3389/fphar.2022.854994] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondria release many damage-associated molecular patterns (DAMPs) when cells are damaged or stressed, with mitochondrial DNA (mtDNA) being. MtDNA activates innate immune responses and induces inflammation through the TLR-9, NLRP3 inflammasome, and cGAS-STING signaling pathways. Released inflammatory factors cause damage to intestinal barrier function. Many bacteria and endotoxins migrate to the circulatory system and lymphatic system, leading to systemic inflammatory response syndrome (SIRS) and even damaging the function of multiple organs throughout the body. This process may ultimately lead to multiple organ dysfunction syndrome (MODS). Recent studies have shown that various factors, such as the release of mtDNA and the massive infiltration of inflammatory factors, can cause intestinal ischemia/reperfusion (I/R) injury. This destroys intestinal barrier function, induces an inflammatory storm, leads to SIRS, increases the vulnerability of organs, and develops into MODS. Mitophagy eliminates dysfunctional mitochondria to maintain cellular homeostasis. This review discusses mtDNA release during the pathogenesis of intestinal I/R and summarizes methods for the prevention or treatment of intestinal I/R. We also discuss the effects of inflammation and increased intestinal barrier permeability on drugs.
Collapse
Affiliation(s)
- Shishi Liao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Luo
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tulanisa Kadier
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingtao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
36
|
Afshari AR, Sanati M, Aminyavari S, Shakeri F, Bibak B, Keshavarzi Z, Soukhtanloo M, Jalili-Nik M, Sadeghi MM, Mollazadeh H, Johnston TP, Sahebkar A. Advantages and drawbacks of dexamethasone in glioblastoma multiforme. Crit Rev Oncol Hematol 2022; 172:103625. [PMID: 35158070 DOI: 10.1016/j.critrevonc.2022.103625] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 12/25/2022] Open
Abstract
The most widespread, malignant, and deadliest type of glial tumor is glioblastoma multiforme (GBM). Despite radiation, chemotherapy, and radical surgery, the median survival of afflicted individuals is about 12 months. Unfortunately, existing therapeutic interventions are abysmal. Dexamethasone (Dex), a synthetic glucocorticoid, has been used for many years to treat brain edema and inflammation caused by GBM. Several investigations have recently shown that Dex also exerts antitumoral effects against GBM. On the other hand, more recent disputed findings have questioned the long-held dogma of Dex treatment for GBM. Unfortunately, steroids are associated with various undesirable side effects, including severe immunosuppression and metabolic changes like hyperglycemia, which may impair the survival of GBM patients. Current ideas and concerns about Dex's effects on GBM cerebral edema, cell proliferation, migration, and its clinical outcomes were investigated in this study.
Collapse
Affiliation(s)
- Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Samaneh Aminyavari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Shakeri
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Bahram Bibak
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zakieh Keshavarzi
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Jalili-Nik
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Montazami Sadeghi
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, Australia; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
37
|
Zhu J, Feng C, Zhang W, Wang Z, Zhong M, Tang W, Wang Z, Shi H, Yin Z, Shi J, Huang Y, Xiao L, Geng D, Wang Z. Activation of dopamine receptor D1 promotes osteogenic differentiation and reduces glucocorticoid-induced bone loss by upregulating the ERK1/2 signaling pathway. Mol Med 2022; 28:23. [PMID: 35189819 PMCID: PMC8862482 DOI: 10.1186/s10020-022-00453-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 02/08/2022] [Indexed: 11/16/2022] Open
Abstract
Background The inhibition of osteogenic differentiation is a major factor in glucocorticoid-induced bone loss, but there is currently no effective treatment. Dopamine, a major neurotransmitter, transmits signals via five different seven-transmembrane G protein-coupled receptors termed D1 to D5. Although the relevance of the neuroendocrine system in bone metabolism has emerged, the precise effects of dopamine receptor signaling on osteoblastogenesis remain unknown. Methods In vitro, western blotting and immunofluorescence staining were used to observe the expression of dopamine receptors in MC3T3-E1 and BMSCs cells treated with dexamethasone (Dex). In addition, Alizarin red S (ARS) and alkaline phosphatase (ALP) staining and western blotting were used to evaluate the effect of D1R activation on osteogenic differentiation in Dex-induced MC3T3-E1 cells via the ERK1/2 signaling pathway. In vivo, micro-CT and hematoxylin and eosin (H&E), toluidine blue and immunohistochemical staining were used to determine the effect of D1R activation on Dex-induced bone loss. Results We demonstrated that the trend in D1R but not D2-5R was consistent with that of osteogenic markers in the presence of Dex. We also demonstrated that the activation of D1R promoted Dex-induced osteogenic differentiation by activating the ERK1/2 pathway in vitro. We further demonstrated that a D1R agonist could reduce Dex-induced bone loss, while pretreatment with a D1R inhibitor blocked the effect of a D1R agonist in vivo. Conclusions Activation of D1R promotes osteogenic differentiation and reduces Dex-induced bone loss by activating the ERK1/2 pathway. Hence, D1R could serve as a potential therapeutic target for glucocorticoid-induced osteoporosis. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00453-0.
Collapse
|
38
|
Cárdenas G, Chávez-Canales M, Espinosa AM, Jordán-Ríos A, Malagon DA, Murillo MFM, Araujo LVT, Campos RLB, Wong-Chew RM, González LER, Cresencio KI, Velázquez EG, de la Cerda MR, Leyva Y, Hernández-Ruiz J, Hernández-Medel ML, León-Hernández M, Quero KM, Monciváis AS, Díaz SH, Martínez IRZ, Martínez-Cuazitl A, Salazar INM, Sarmiento EB, Peña AF, Hernández PS, Reynoso RIA, Reyes DM, del Río Ambriz LR, Bonilla RAA, Cruz J, Huerta L, Fierro NA, Hernández M, Pérez-Tapia M, Meneses G, Espíndola-Arriaga E, Rosas G, Chinney A, Mendoza SR, Hernández-Aceves JA, Cervantes-Torres J, Rodríguez AF, Alor RO, Francisco SO, Salazar EA, Besedovsky H, Romano MC, Bobes RJ, Jung H, Soldevila G, López-Alvarenga J, Fragoso G, Laclette JP, Sciutto E. Intranasal dexamethasone: a new clinical trial for the control of inflammation and neuroinflammation in COVID-19 patients. Trials 2022; 23:148. [PMID: 35164840 PMCID: PMC8845269 DOI: 10.1186/s13063-022-06075-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 02/01/2022] [Indexed: 12/15/2022] Open
Abstract
Background By end December of 2021, COVID-19 has infected around 276 million individuals and caused over 5 million deaths worldwide. Infection results in dysregulated systemic inflammation, multi-organ dysfunction, and critical illness. Cells of the central nervous system are also affected, triggering an uncontrolled neuroinflammatory response. Low doses of glucocorticoids, administered orally or intravenously, reduce mortality among moderate and severe COVID-19 patients. However, low doses administered by these routes do not reach therapeutic levels in the CNS. In contrast, intranasally administered dexamethasone can result in therapeutic doses in the CNS even at low doses. Methods This is an approved open-label, multicenter, randomized controlled trial to compare the effectiveness of intranasal versus intravenous dexamethasone administered in low doses to moderate and severe COVID-19 adult patients. The protocol is conducted in five health institutions in Mexico City. A total of 120 patients will be randomized into two groups (intravenous vs. intranasal) at a 1:1 ratio. Both groups will be treated with the corresponding dexamethasone scheme for 10 days. The primary outcome of the study will be clinical improvement, defined as a statistically significant reduction in the NEWS-2 score of patients with intranasal versus intravenous dexamethasone administration. The secondary outcome will be the reduction in mortality during hospitalization. Conclusions This protocol is currently in progress to improve the efficacy of the standard therapeutic dexamethasone regimen for moderate and severe COVID-19 patients. Trial registration ClinicalTrials.govNCT04513184. Registered November 12, 2020. Approved by La Comisión Federal para la Protección contra Riesgos Sanitarios (COFEPRIS) with identification number DI/20/407/04/36. People are currently being recruited. Graphical abstract ![]()
REVIVAL is a multicenter, open-label, randomized, controlled study to compare the standard low doses of intravenous dexamethasone with weight-adjusted low doses of intranasal dexamethasone. Intranasal dexamethasone can reach the respiratory tract more effectively than intravenous administration. Intranasal dexamethasone can reach the central nervous system in therapeutic concentrations, even at low doses. REVIVAL aims to reduce central failures and sequelae by controlling not only systemic inflammation but also neuroinflammation.
Collapse
|
39
|
Miura T, Okizaki A, Hasuo H, Satomi E, Tagami K, Imai K, Kojima T, Satake H, Ishiki H, Inoue A, Yamaguchi T. Dexamethasone 8 mg for Cancer-Related Fatigue in Inpatients with Advanced Cancer Undergoing Palliative Care: A Multicenter Phase II Trial. Palliat Med Rep 2021; 2:316-323. [PMID: 34927157 PMCID: PMC8675272 DOI: 10.1089/pmr.2021.0053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 11/26/2022] Open
Abstract
Objective: No standard treatment for cancer-related fatigue (CRF) for inpatients in a palliative care setting exists. The aim of this study was to validate the previous study-derived efficacy of dexamethasone 8 mg for CRF among inpatients in a palliative care setting. Methods: Inpatients with moderate fatigue (≥4/10) were enrolled in a multicenter phase II trial. Dexamethasone 8 mg p.o. or 6.6 mg i.v. was administered for seven days and 4 mg p.o. or 3.3 mg i.v. for seven consecutive days. The primary endpoint was a threshold average change of Functional Assessment of Chronic Illness Therapy (FACIT)-fatigue subscale score of 3. The secondary endpoints were evaluated with the anorexia-cachexia subscale (ACS), and the Edmonton symptom assessment scale-revised Japanese version. Results: A total of 32 patients were enrolled. On day 8, the mean change of FACIT-fatigue subscale from day 1 was 5.2 (95% confidence interval 0.8-10.0), in which the lower bound was above 0 but not above the prespecified threshold value of 3.0 (p = 0.72). Edmonton symptom assessment system (ESAS)-fatigue was significantly improved by day 3 (p = 0.02), but not on day 8 or day 15. ACS, physical well-being, and ESAS-lack of appetite significantly improved by day 8 and day 15. Adverse events were tolerable. Conclusion: This study showed that dexamethasone 8 mg failed to achieve the preset efficacy for CRF among inpatients in a palliative care setting. However, this treatment improved fatigue and would be an option for CRF. jRCT (jRCTs031180068).
Collapse
Affiliation(s)
- Tomofumi Miura
- Department of Palliative Medicine, National Cancer Center Hospital East, Kashiwa, Japan
| | - Ayumi Okizaki
- Innovation Center for Supportive, Palliative and Psychosocial Care, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Hideaki Hasuo
- Department of Psychosomatic Medicine, Kansai Medical University, Hirakata, Japan
| | - Eriko Satomi
- Department of Palliative Medicine, National Cancer Center Hospital, Tokyo, Japan
| | - Keita Tagami
- Department of Palliative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kengo Imai
- Seirei Hospice, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hironaga Satake
- Cancer Treatment Center, Kansai Medical University Hospital, Hirakata, Japan
| | - Hiroto Ishiki
- Department of Palliative Medicine, National Cancer Center Hospital, Tokyo, Japan
| | - Akira Inoue
- Department of Palliative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takuhiro Yamaguchi
- Division of Biostatistics, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
40
|
Mesenchymal Stem Cell-Based Therapy for Rheumatoid Arthritis. Int J Mol Sci 2021; 22:ijms222111592. [PMID: 34769021 PMCID: PMC8584240 DOI: 10.3390/ijms222111592] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have great potential to differentiate into various types of cells, including but not limited to, adipocytes, chondrocytes and osteoblasts. In addition to their progenitor characteristics, MSCs hold unique immunomodulatory properties that provide new opportunities in the treatment of autoimmune diseases, and can serve as a promising tool in stem cell-based therapy. Rheumatoid arthritis (RA) is a chronic systemic autoimmune disorder that deteriorates quality and function of the synovium membrane, resulting in chronic inflammation, pain and progressive cartilage and bone destruction. The mechanism of RA pathogenesis is associated with dysregulation of innate and adaptive immunity. Current conventional treatments by steroid drugs, antirheumatic drugs and biological agents are being applied in clinical practice. However, long-term use of these drugs causes side effects, and some RA patients may acquire resistance to these drugs. In this regard, recently investigated MSC-based therapy is considered as a promising approach in RA treatment. In this study, we review conventional and modern treatment approaches, such as MSC-based therapy through the understanding of the link between MSCs and the innate and adaptive immune systems. Moreover, we discuss recent achievements in preclinical and clinical studies as well as various strategies for the enhancement of MSC immunoregulatory properties.
Collapse
|
41
|
Li H, Gao Y, Xie L, Wang R, Duan R, Li Z, Chen B, Zhu L, Wang X, Su W. Prednisone Reprograms the Transcriptional Immune Cell Landscape in CNS Autoimmune Disease. Front Immunol 2021; 12:739605. [PMID: 34484247 PMCID: PMC8414592 DOI: 10.3389/fimmu.2021.739605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 07/29/2021] [Indexed: 11/13/2022] Open
Abstract
Glucocorticoids (GCs) are widely used immunosuppressive drugs for autoimmune diseases, although considerable gaps exist between current knowledge of the mechanisms of GCs and their conclusive immune-regulatory effects. Here we generated a single-cell transcriptional immune cell atlas based on prednisone-treated or untreated experimental autoimmune uveitis (EAU) mice. Immune cells were globally activated in EAU, and prednisone partially reversed this effect in terms of cell composition, gene expression, transcription factor regulation, and cell-cell communication. Prednisone exerted considerable rescue effects on T and B cells and increased the proportion of neutrophils. Besides commonly regulated transcriptional factors (Fosb, Jun, Jund), several genes were only regulated in certain cell types (e.g. Cxcr4 and Bhlhe40 in T cells), suggesting cell-type-dependent immunosuppressive properties of GC. These findings provide new insights into the mechanisms behind the properties and cell-specific effects of GCs and can potentially benefit immunoregulatory therapy development.
Collapse
Affiliation(s)
- He Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yuehan Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lihui Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Rong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Runping Duan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Binyao Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lei Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xianggui Wang
- Eye Center of Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
42
|
Egi M, Ogura H, Yatabe T, Atagi K, Inoue S, Iba T, Kakihana Y, Kawasaki T, Kushimoto S, Kuroda Y, Kotani J, Shime N, Taniguchi T, Tsuruta R, Doi K, Doi M, Nakada TA, Nakane M, Fujishima S, Hosokawa N, Masuda Y, Matsushima A, Matsuda N, Yamakawa K, Hara Y, Sakuraya M, Ohshimo S, Aoki Y, Inada M, Umemura Y, Kawai Y, Kondo Y, Saito H, Taito S, Takeda C, Terayama T, Tohira H, Hashimoto H, Hayashida K, Hifumi T, Hirose T, Fukuda T, Fujii T, Miura S, Yasuda H, Abe T, Andoh K, Iida Y, Ishihara T, Ide K, Ito K, Ito Y, Inata Y, Utsunomiya A, Unoki T, Endo K, Ouchi A, Ozaki M, Ono S, Katsura M, Kawaguchi A, Kawamura Y, Kudo D, Kubo K, Kurahashi K, Sakuramoto H, Shimoyama A, Suzuki T, Sekine S, Sekino M, Takahashi N, Takahashi S, Takahashi H, Tagami T, Tajima G, Tatsumi H, Tani M, Tsuchiya A, Tsutsumi Y, Naito T, Nagae M, Nagasawa I, Nakamura K, Nishimura T, Nunomiya S, Norisue Y, Hashimoto S, Hasegawa D, Hatakeyama J, Hara N, Higashibeppu N, Furushima N, Furusono H, Matsuishi Y, Matsuyama T, Minematsu Y, Miyashita R, Miyatake Y, Moriyasu M, Yamada T, et alEgi M, Ogura H, Yatabe T, Atagi K, Inoue S, Iba T, Kakihana Y, Kawasaki T, Kushimoto S, Kuroda Y, Kotani J, Shime N, Taniguchi T, Tsuruta R, Doi K, Doi M, Nakada TA, Nakane M, Fujishima S, Hosokawa N, Masuda Y, Matsushima A, Matsuda N, Yamakawa K, Hara Y, Sakuraya M, Ohshimo S, Aoki Y, Inada M, Umemura Y, Kawai Y, Kondo Y, Saito H, Taito S, Takeda C, Terayama T, Tohira H, Hashimoto H, Hayashida K, Hifumi T, Hirose T, Fukuda T, Fujii T, Miura S, Yasuda H, Abe T, Andoh K, Iida Y, Ishihara T, Ide K, Ito K, Ito Y, Inata Y, Utsunomiya A, Unoki T, Endo K, Ouchi A, Ozaki M, Ono S, Katsura M, Kawaguchi A, Kawamura Y, Kudo D, Kubo K, Kurahashi K, Sakuramoto H, Shimoyama A, Suzuki T, Sekine S, Sekino M, Takahashi N, Takahashi S, Takahashi H, Tagami T, Tajima G, Tatsumi H, Tani M, Tsuchiya A, Tsutsumi Y, Naito T, Nagae M, Nagasawa I, Nakamura K, Nishimura T, Nunomiya S, Norisue Y, Hashimoto S, Hasegawa D, Hatakeyama J, Hara N, Higashibeppu N, Furushima N, Furusono H, Matsuishi Y, Matsuyama T, Minematsu Y, Miyashita R, Miyatake Y, Moriyasu M, Yamada T, Yamada H, Yamamoto R, Yoshida T, Yoshida Y, Yoshimura J, Yotsumoto R, Yonekura H, Wada T, Watanabe E, Aoki M, Asai H, Abe T, Igarashi Y, Iguchi N, Ishikawa M, Ishimaru G, Isokawa S, Itakura R, Imahase H, Imura H, Irinoda T, Uehara K, Ushio N, Umegaki T, Egawa Y, Enomoto Y, Ota K, Ohchi Y, Ohno T, Ohbe H, Oka K, Okada N, Okada Y, Okano H, Okamoto J, Okuda H, Ogura T, Onodera Y, Oyama Y, Kainuma M, Kako E, Kashiura M, Kato H, Kanaya A, Kaneko T, Kanehata K, Kano KI, Kawano H, Kikutani K, Kikuchi H, Kido T, Kimura S, Koami H, Kobashi D, Saiki I, Sakai M, Sakamoto A, Sato T, Shiga Y, Shimoto M, Shimoyama S, Shoko T, Sugawara Y, Sugita A, Suzuki S, Suzuki Y, Suhara T, Sonota K, Takauji S, Takashima K, Takahashi S, Takahashi Y, Takeshita J, Tanaka Y, Tampo A, Tsunoyama T, Tetsuhara K, Tokunaga K, Tomioka Y, Tomita K, Tominaga N, Toyosaki M, Toyoda Y, Naito H, Nagata I, Nagato T, Nakamura Y, Nakamori Y, Nahara I, Naraba H, Narita C, Nishioka N, Nishimura T, Nishiyama K, Nomura T, Haga T, Hagiwara Y, Hashimoto K, Hatachi T, Hamasaki T, Hayashi T, Hayashi M, Hayamizu A, Haraguchi G, Hirano Y, Fujii R, Fujita M, Fujimura N, Funakoshi H, Horiguchi M, Maki J, Masunaga N, Matsumura Y, Mayumi T, Minami K, Miyazaki Y, Miyamoto K, Murata T, Yanai M, Yano T, Yamada K, Yamada N, Yamamoto T, Yoshihiro S, Tanaka H, Nishida O. The Japanese Clinical Practice Guidelines for Management of Sepsis and Septic Shock 2020 (J-SSCG 2020). J Intensive Care 2021; 9:53. [PMID: 34433491 PMCID: PMC8384927 DOI: 10.1186/s40560-021-00555-7] [Show More Authors] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/10/2021] [Indexed: 02/08/2023] Open
Abstract
The Japanese Clinical Practice Guidelines for Management of Sepsis and Septic Shock 2020 (J-SSCG 2020), a Japanese-specific set of clinical practice guidelines for sepsis and septic shock created as revised from J-SSCG 2016 jointly by the Japanese Society of Intensive Care Medicine and the Japanese Association for Acute Medicine, was first released in September 2020 and published in February 2021. An English-language version of these guidelines was created based on the contents of the original Japanese-language version. The purpose of this guideline is to assist medical staff in making appropriate decisions to improve the prognosis of patients undergoing treatment for sepsis and septic shock. We aimed to provide high-quality guidelines that are easy to use and understand for specialists, general clinicians, and multidisciplinary medical professionals. J-SSCG 2016 took up new subjects that were not present in SSCG 2016 (e.g., ICU-acquired weakness [ICU-AW], post-intensive care syndrome [PICS], and body temperature management). The J-SSCG 2020 covered a total of 22 areas with four additional new areas (patient- and family-centered care, sepsis treatment system, neuro-intensive treatment, and stress ulcers). A total of 118 important clinical issues (clinical questions, CQs) were extracted regardless of the presence or absence of evidence. These CQs also include those that have been given particular focus within Japan. This is a large-scale guideline covering multiple fields; thus, in addition to the 25 committee members, we had the participation and support of a total of 226 members who are professionals (physicians, nurses, physiotherapists, clinical engineers, and pharmacists) and medical workers with a history of sepsis or critical illness. The GRADE method was adopted for making recommendations, and the modified Delphi method was used to determine recommendations by voting from all committee members.As a result, 79 GRADE-based recommendations, 5 Good Practice Statements (GPS), 18 expert consensuses, 27 answers to background questions (BQs), and summaries of definitions and diagnosis of sepsis were created as responses to 118 CQs. We also incorporated visual information for each CQ according to the time course of treatment, and we will also distribute this as an app. The J-SSCG 2020 is expected to be widely used as a useful bedside guideline in the field of sepsis treatment both in Japan and overseas involving multiple disciplines.
Collapse
Affiliation(s)
- Moritoki Egi
- Department of Surgery Related, Division of Anesthesiology, Kobe University Graduate School of Medicine, Kusunoki-cho 7-5-2, Chuo-ku, Kobe, Hyogo, Japan.
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Medical School, Yamadaoka 2-15, Suita, Osaka, Japan.
| | - Tomoaki Yatabe
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kazuaki Atagi
- Department of Intensive Care Unit, Nara Prefectural General Medical Center, Nara, Japan
| | - Shigeaki Inoue
- Department of Disaster and Emergency Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University, Tokyo, Japan
| | - Yasuyuki Kakihana
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tatsuya Kawasaki
- Department of Pediatric Critical Care, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Shigeki Kushimoto
- Division of Emergency and Critical Care Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Kuroda
- Department of Emergency, Disaster, and Critical Care Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Joji Kotani
- Department of Surgery Related, Division of Disaster and Emergency Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Nobuaki Shime
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takumi Taniguchi
- Department of Anesthesiology and Intensive Care Medicine, Kanazawa University, Kanazawa, Japan
| | - Ryosuke Tsuruta
- Acute and General Medicine, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Kent Doi
- Department of Acute Medicine, The University of Tokyo, Tokyo, Japan
| | - Matsuyuki Doi
- Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Taka-Aki Nakada
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masaki Nakane
- Department of Emergency and Critical Care Medicine, Yamagata University Hospital, Yamagata, Japan
| | - Seitaro Fujishima
- Center for General Medicine Education, Keio University School of Medicine, Tokyo, Japan
| | - Naoto Hosokawa
- Department of Infectious Diseases, Kameda Medical Center, Kamogawa, Japan
| | - Yoshiki Masuda
- Department of Intensive Care Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Asako Matsushima
- Department of Advancing Acute Medicine, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Naoyuki Matsuda
- Department of Emergency and Critical Care Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuma Yamakawa
- Department of Emergency Medicine, Osaka Medical College, Osaka, Japan
| | - Yoshitaka Hara
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masaaki Sakuraya
- Department of Emergency and Intensive Care Medicine, JA Hiroshima General Hospital, Hatsukaichi, Japan
| | - Shinichiro Ohshimo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yoshitaka Aoki
- Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mai Inada
- Member of Japanese Association for Acute Medicine, Tokyo, Japan
| | - Yutaka Umemura
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | - Yusuke Kawai
- Department of Nursing, Fujita Health University Hospital, Toyoake, Japan
| | - Yutaka Kondo
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Hiroki Saito
- Department of Emergency and Critical Care Medicine, St. Marianna University School of Medicine, Yokohama City Seibu Hospital, Yokohama, Japan
| | - Shunsuke Taito
- Division of Rehabilitation, Department of Clinical Support and Practice, Hiroshima University Hospital, Hiroshima, Japan
| | - Chikashi Takeda
- Department of Anesthesia, Kyoto University Hospital, Kyoto, Japan
| | - Takero Terayama
- Department of Psychiatry, School of Medicine, National Defense Medical College, Tokorozawa, Japan
| | | | - Hideki Hashimoto
- Department of Emergency and Critical Care Medicine/Infectious Disease, Hitachi General Hospital, Hitachi, Japan
| | - Kei Hayashida
- The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Toru Hifumi
- Department of Emergency and Critical Care Medicine, St. Luke's International Hospital, Tokyo, Japan
| | - Tomoya Hirose
- Emergency and Critical Care Medical Center, Osaka Police Hospital, Osaka, Japan
| | - Tatsuma Fukuda
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Tomoko Fujii
- Intensive Care Unit, Jikei University Hospital, Tokyo, Japan
| | - Shinya Miura
- The Royal Children's Hospital Melbourne, Melbourne, Australia
| | - Hideto Yasuda
- Department of Emergency and Critical Care Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Toshikazu Abe
- Department of Emergency and Critical Care Medicine, Tsukuba Memorial Hospital, Tsukuba, Japan
| | - Kohkichi Andoh
- Division of Anesthesiology, Division of Intensive Care, Division of Emergency and Critical Care, Sendai City Hospital, Sendai, Japan
| | - Yuki Iida
- Department of Physical Therapy, School of Health Sciences, Toyohashi Sozo University, Toyohashi, Japan
| | - Tadashi Ishihara
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Kentaro Ide
- Critical Care Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Kenta Ito
- Department of General Pediatrics, Aichi Children's Health and Medical Center, Obu, Japan
| | - Yusuke Ito
- Department of Infectious Disease, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Yu Inata
- Department of Intensive Care Medicine, Osaka Women's and Children's Hospital, Izumi, Japan
| | - Akemi Utsunomiya
- Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Unoki
- Department of Acute and Critical Care Nursing, School of Nursing, Sapporo City University, Sapporo, Japan
| | - Koji Endo
- Department of Pharmacoepidemiology, Kyoto University Graduate School of Medicine and Public Health, Kyoto, Japan
| | - Akira Ouchi
- College of Nursing, Ibaraki Christian University, Hitachi, Japan
| | - Masayuki Ozaki
- Department of Emergency and Critical Care Medicine, Komaki City Hospital, Komaki, Japan
| | - Satoshi Ono
- Gastroenterological Center, Shinkuki General Hospital, Kuki, Japan
| | | | | | - Yusuke Kawamura
- Department of Rehabilitation, Showa General Hospital, Tokyo, Japan
| | - Daisuke Kudo
- Division of Emergency and Critical Care Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kenji Kubo
- Department of Emergency Medicine and Department of Infectious Diseases, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Kiyoyasu Kurahashi
- Department of Anesthesiology and Intensive Care Medicine, International University of Health and Welfare School of Medicine, Narita, Japan
| | | | - Akira Shimoyama
- Department of Emergency and Critical Care Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Takeshi Suzuki
- Department of Anesthesiology, Tokai University School of Medicine, Isehara, Japan
| | - Shusuke Sekine
- Department of Anesthesiology, Tokyo Medical University, Tokyo, Japan
| | - Motohiro Sekino
- Division of Intensive Care, Nagasaki University Hospital, Nagasaki, Japan
| | - Nozomi Takahashi
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Sei Takahashi
- Center for Innovative Research for Communities and Clinical Excellence (CiRC2LE), Fukushima Medical University, Fukushima, Japan
| | - Hiroshi Takahashi
- Department of Cardiology, Steel Memorial Muroran Hospital, Muroran, Japan
| | - Takashi Tagami
- Department of Emergency and Critical Care Medicine, Nippon Medical School Musashi Kosugi Hospital, Kawasaki, Japan
| | - Goro Tajima
- Nagasaki University Hospital Acute and Critical Care Center, Nagasaki, Japan
| | - Hiroomi Tatsumi
- Department of Intensive Care Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masanori Tani
- Division of Critical Care Medicine, Saitama Children's Medical Center, Saitama, Japan
| | - Asuka Tsuchiya
- Department of Emergency and Critical Care Medicine, National Hospital Organization Mito Medical Center, Ibaraki, Japan
| | - Yusuke Tsutsumi
- Department of Emergency and Critical Care Medicine, National Hospital Organization Mito Medical Center, Ibaraki, Japan
| | - Takaki Naito
- Department of Emergency and Critical Care Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Masaharu Nagae
- Department of Intensive Care Medicine, Kobe University Hospital, Kobe, Japan
| | | | - Kensuke Nakamura
- Department of Emergency and Critical Care Medicine, Hitachi General Hospital, Hitachi, Japan
| | - Tetsuro Nishimura
- Department of Traumatology and Critical Care Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shin Nunomiya
- Department of Anesthesiology and Intensive Care Medicine, Division of Intensive Care, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Yasuhiro Norisue
- Department of Emergency and Critical Care Medicine, Tokyo Bay Urayasu Ichikawa Medical Center, Urayasu, Japan
| | - Satoru Hashimoto
- Department of Anesthesiology and Intensive Care Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Hasegawa
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Junji Hatakeyama
- Department of Emergency and Critical Care Medicine, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Naoki Hara
- Department of Pharmacy, Yokohama Rosai Hospital, Yokohama, Japan
| | - Naoki Higashibeppu
- Department of Anesthesiology and Nutrition Support Team, Kobe City Medical Center General Hospital, Kobe City Hospital Organization, Kobe, Japan
| | - Nana Furushima
- Department of Anesthesiology, Kobe University Hospital, Kobe, Japan
| | - Hirotaka Furusono
- Department of Rehabilitation, University of Tsukuba Hospital/Exult Co., Ltd., Tsukuba, Japan
| | - Yujiro Matsuishi
- Doctoral program in Clinical Sciences. Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Tasuku Matsuyama
- Department of Emergency Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yusuke Minematsu
- Department of Clinical Engineering, Osaka University Hospital, Suita, Japan
| | - Ryoichi Miyashita
- Department of Intensive Care Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yuji Miyatake
- Department of Clinical Engineering, Kakogawa Central City Hospital, Kakogawa, Japan
| | - Megumi Moriyasu
- Division of Respiratory Care and Rapid Response System, Intensive Care Center, Kitasato University Hospital, Sagamihara, Japan
| | - Toru Yamada
- Department of Nursing, Toho University Omori Medical Center, Tokyo, Japan
| | - Hiroyuki Yamada
- Department of Primary Care and Emergency Medicine, Kyoto University Hospital, Kyoto, Japan
| | - Ryo Yamamoto
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takeshi Yoshida
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuhei Yoshida
- Nursing Department, Osaka General Medical Center, Osaka, Japan
| | - Jumpei Yoshimura
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, Japan
| | | | - Hiroshi Yonekura
- Department of Clinical Anesthesiology, Mie University Hospital, Tsu, Japan
| | - Takeshi Wada
- Department of Anesthesiology and Critical Care Medicine, Division of Acute and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Eizo Watanabe
- Department of Emergency and Critical Care Medicine, Eastern Chiba Medical Center, Togane, Japan
| | - Makoto Aoki
- Department of Emergency Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hideki Asai
- Department of Emergency and Critical Care Medicine, Nara Medical University, Kashihara, Japan
| | - Takakuni Abe
- Department of Anesthesiology and Intensive Care, Oita University Hospital, Yufu, Japan
| | - Yutaka Igarashi
- Department of Emergency and Critical Care Medicine, Nippon Medical School Hospital, Tokyo, Japan
| | - Naoya Iguchi
- Department of Anesthesiology and Intensive Care Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Masami Ishikawa
- Department of Anesthesiology, Emergency and Critical Care Medicine, Kure Kyosai Hospital, Kure, Japan
| | - Go Ishimaru
- Department of General Internal Medicine, Soka Municipal Hospital, Soka, Japan
| | - Shutaro Isokawa
- Department of Emergency and Critical Care Medicine, St. Luke's International Hospital, Tokyo, Japan
| | - Ryuta Itakura
- Department of Emergency and Critical Care Medicine, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Hisashi Imahase
- Department of Biomedical Ethics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruki Imura
- Department of Infectious Diseases, Rakuwakai Otowa Hospital, Kyoto, Japan
- Department of Health Informatics, School of Public Health, Kyoto University, Kyoto, Japan
| | | | - Kenji Uehara
- Department of Anesthesiology, National Hospital Organization Iwakuni Clinical Center, Iwakuni, Japan
| | - Noritaka Ushio
- Advanced Medical Emergency Department and Critical Care Center, Japan Red Cross Maebashi Hospital, Maebashi, Japan
| | - Takeshi Umegaki
- Department of Anesthesiology, Kansai Medical University, Hirakata, Japan
| | - Yuko Egawa
- Advanced Emergency and Critical Care Center, Saitama Red Cross Hospital, Saitama, Japan
| | - Yuki Enomoto
- Department of Emergency and Critical Care Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kohei Ota
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yoshifumi Ohchi
- Department of Anesthesiology and Intensive Care, Oita University Hospital, Yufu, Japan
| | - Takanori Ohno
- Department of Emergency and Critical Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hiroyuki Ohbe
- Department of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo, Tokyo, Japan
| | | | - Nobunaga Okada
- Department of Emergency Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yohei Okada
- Department of Primary care and Emergency medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiromu Okano
- Department of Anesthesiology, Kyorin University School of Medicine, Tokyo, Japan
| | - Jun Okamoto
- Department of ER, Hashimoto Municipal Hospital, Hashimoto, Japan
| | - Hiroshi Okuda
- Department of Community Medical Supports, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Takayuki Ogura
- Tochigi prefectural Emergency and Critical Care Center, Imperial Gift Foundation Saiseikai, Utsunomiya Hospital, Utsunomiya, Japan
| | - Yu Onodera
- Department of Anesthesiology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Yuhta Oyama
- Department of Internal Medicine, Dialysis Center, Kichijoji Asahi Hospital, Tokyo, Japan
| | - Motoshi Kainuma
- Anesthesiology, Emergency Medicine, and Intensive Care Division, Inazawa Municipal Hospital, Inazawa, Japan
| | - Eisuke Kako
- Department of Anesthesiology and Intensive Care Medicine, Nagoya-City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masahiro Kashiura
- Department of Emergency and Critical Care Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Hiromi Kato
- Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Akihiro Kanaya
- Department of Anesthesiology, Sendai Medical Center, Sendai, Japan
| | - Tadashi Kaneko
- Emergency and Critical Care Center, Mie University Hospital, Tsu, Japan
| | - Keita Kanehata
- Advanced Medical Emergency Department and Critical Care Center, Japan Red Cross Maebashi Hospital, Maebashi, Japan
| | - Ken-Ichi Kano
- Department of Emergency Medicine, Fukui Prefectural Hospital, Fukui, Japan
| | - Hiroyuki Kawano
- Department of Gastroenterological Surgery, Onga Hospital, Fukuoka, Japan
| | - Kazuya Kikutani
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hitoshi Kikuchi
- Department of Emergency and Critical Care Medicine, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Takahiro Kido
- Department of Pediatrics, University of Tsukuba Hospital, Tsukuba, Japan
| | - Sho Kimura
- Division of Critical Care Medicine, Saitama Children's Medical Center, Saitama, Japan
| | - Hiroyuki Koami
- Center for Translational Injury Research, University of Texas Health Science Center at Houston, Houston, USA
| | - Daisuke Kobashi
- Advanced Medical Emergency Department and Critical Care Center, Japan Red Cross Maebashi Hospital, Maebashi, Japan
| | - Iwao Saiki
- Department of Anesthesiology, Tokyo Medical University, Tokyo, Japan
| | - Masahito Sakai
- Department of General Medicine Shintakeo Hospital, Takeo, Japan
| | - Ayaka Sakamoto
- Department of Emergency and Critical Care Medicine, University of Tsukuba Hospital, Tsukuba, Japan
| | - Tetsuya Sato
- Tohoku University Hospital Emergency Center, Sendai, Japan
| | - Yasuhiro Shiga
- Department of Orthopaedic Surgery, Center for Advanced Joint Function and Reconstructive Spine Surgery, Graduate school of Medicine, Chiba University, Chiba, Japan
| | - Manabu Shimoto
- Department of Primary care and Emergency medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinya Shimoyama
- Department of Pediatric Cardiology and Intensive Care, Gunma Children's Medical Center, Shibukawa, Japan
| | - Tomohisa Shoko
- Department of Emergency and Critical Care Medicine, Tokyo Women's Medical University Medical Center East, Tokyo, Japan
| | - Yoh Sugawara
- Department of Anesthesiology, Yokohama City University, Yokohama, Japan
| | - Atsunori Sugita
- Department of Acute Medicine, Division of Emergency and Critical Care Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Satoshi Suzuki
- Department of Intensive Care, Okayama University Hospital, Okayama, Japan
| | - Yuji Suzuki
- Department of Anesthesiology and Intensive Care Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomohiro Suhara
- Department of Anesthesiology, Keio University School of Medicine, Tokyo, Japan
| | - Kenji Sonota
- Department of Intensive Care Medicine, Miyagi Children's Hospital, Sendai, Japan
| | - Shuhei Takauji
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Kohei Takashima
- Critical Care Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Sho Takahashi
- Department of Cardiology, Fukuyama City Hospital, Fukuyama, Japan
| | - Yoko Takahashi
- Department of General Internal Medicine, Koga General Hospital, Koga, Japan
| | - Jun Takeshita
- Department of Anesthesiology, Osaka Women's and Children's Hospital, Izumi, Japan
| | - Yuuki Tanaka
- Fukuoka Prefectural Psychiatric Center, Dazaifu Hospital, Dazaifu, Japan
| | - Akihito Tampo
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Taichiro Tsunoyama
- Department of Emergency Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Kenichi Tetsuhara
- Emergency and Critical Care Center, Kyushu University Hospital, Fukuoka, Japan
| | - Kentaro Tokunaga
- Department of Intensive Care Medicine, Kumamoto University Hospital, Kumamoto, Japan
| | - Yoshihiro Tomioka
- Department of Anesthesiology and Intensive Care Unit, Todachuo General Hospital, Toda, Japan
| | - Kentaro Tomita
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Naoki Tominaga
- Department of Emergency and Critical Care Medicine, Nippon Medical School Hospital, Tokyo, Japan
| | - Mitsunobu Toyosaki
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yukitoshi Toyoda
- Department of Emergency and Critical Care Medicine, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan
| | - Hiromichi Naito
- Department of Emergency, Critical Care, and Disaster Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Isao Nagata
- Intensive Care Unit, Yokohama City Minato Red Cross Hospital, Yokohama, Japan
| | - Tadashi Nagato
- Department of Respiratory Medicine, Tokyo Yamate Medical Center, Tokyo, Japan
| | - Yoshimi Nakamura
- Department of Emergency and Critical Care Medicine, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Yuki Nakamori
- Department of Clinical Anesthesiology, Mie University Hospital, Tsu, Japan
| | - Isao Nahara
- Department of Anesthesiology and Critical Care Medicine, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Hiromu Naraba
- Department of Emergency and Critical Care Medicine, Hitachi General Hospital, Hitachi, Japan
| | - Chihiro Narita
- Department of Emergency Medicine and Intensive Care Medicine, Shizuoka General Hospital, Shizuoka, Japan
| | - Norihiro Nishioka
- Department of Preventive Services, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoya Nishimura
- Advanced Medical Emergency Department and Critical Care Center, Japan Red Cross Maebashi Hospital, Maebashi, Japan
| | - Kei Nishiyama
- Division of Emergency and Critical Care Medicine Niigata University Graduate School of Medical and Dental Science, Niigata, Japan
| | - Tomohisa Nomura
- Department of Emergency and Critical Care Medicine, Juntendo University Nerima Hospital, Tokyo, Japan
| | - Taiki Haga
- Department of Pediatric Critical Care Medicine, Osaka City General Hospital, Osaka, Japan
| | - Yoshihiro Hagiwara
- Department of Emergency and Critical Care Medicine, Saiseikai Utsunomiya Hospital, Utsunomiya, Japan
| | - Katsuhiko Hashimoto
- Research Associate of Minimally Invasive Surgical and Medical Oncology, Fukushima Medical University, Fukushima, Japan
| | - Takeshi Hatachi
- Department of Intensive Care Medicine, Osaka Women's and Children's Hospital, Izumi, Japan
| | - Toshiaki Hamasaki
- Department of Emergency Medicine, Japanese Red Cross Society Wakayama Medical Center, Wakayama, Japan
| | - Takuya Hayashi
- Division of Critical Care Medicine, Saitama Children's Medical Center, Saitama, Japan
| | - Minoru Hayashi
- Department of Emergency Medicine, Fukui Prefectural Hospital, Fukui, Japan
| | - Atsuki Hayamizu
- Department of Emergency Medicine, Saitama Saiseikai Kurihashi Hospital, Kuki, Japan
| | - Go Haraguchi
- Division of Intensive Care Unit, Sakakibara Heart Institute, Tokyo, Japan
| | - Yohei Hirano
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Ryo Fujii
- Department of Emergency Medicine and Critical Care Medicine, Tochigi Prefectural Emergency and Critical Care Center, Imperial Foundation Saiseikai Utsunomiya Hospital, Utsunomiya, Japan
| | - Motoki Fujita
- Acute and General Medicine, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Naoyuki Fujimura
- Department of Anesthesiology, St. Mary's Hospital, Our Lady of the Snow Social Medical Corporation, Kurume, Japan
| | - Hiraku Funakoshi
- Department of Emergency and Critical Care Medicine, Tokyo Bay Urayasu Ichikawa Medical Center, Urayasu, Japan
| | - Masahito Horiguchi
- Department of Emergency and Critical Care Medicine, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Jun Maki
- Department of Critical Care Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Naohisa Masunaga
- Department of Healthcare Epidemiology, School of Public Health in the Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yosuke Matsumura
- Department of Intensive Care, Chiba Emergency Medical Center, Chiba, Japan
| | - Takuya Mayumi
- Department of Internal Medicine, Kanazawa Municipal Hospital, Kanazawa, Japan
| | - Keisuke Minami
- Ishikawa Prefectual Central Hospital Emergency and Critical Care Center, Kanazawa, Japan
| | - Yuya Miyazaki
- Department of Emergency and General Internal Medicine, Saiseikai Kawaguchi General Hospital, Kawaguchi, Japan
| | - Kazuyuki Miyamoto
- Department of Emergency and Disaster Medicine, Showa University, Tokyo, Japan
| | - Teppei Murata
- Department of Cardiology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Machi Yanai
- Department of Emergency Medicine, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Takao Yano
- Department of Critical Care and Emergency Medicine, Miyazaki Prefectural Nobeoka Hospital, Nobeoka, Japan
| | - Kohei Yamada
- Department of Traumatology and Critical Care Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Naoki Yamada
- Department of Emergency Medicine, University of Fukui Hospital, Fukui, Japan
| | - Tomonori Yamamoto
- Department of Intensive Care Unit, Nara Prefectural General Medical Center, Nara, Japan
| | - Shodai Yoshihiro
- Pharmaceutical Department, JA Hiroshima General Hospital, Hatsukaichi, Japan
| | - Hiroshi Tanaka
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Osamu Nishida
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
43
|
Bruscoli S, Febo M, Riccardi C, Migliorati G. Glucocorticoid Therapy in Inflammatory Bowel Disease: Mechanisms and Clinical Practice. Front Immunol 2021; 12:691480. [PMID: 34149734 PMCID: PMC8209469 DOI: 10.3389/fimmu.2021.691480] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease (IBD) comprises ulcerative colitis (UC) and Crohn's disease (CD). IBD etiopathology is multifactorial and involves alteration of immune cells and chronic activation of the inflammatory cascade against yet unknown environmental factors that trigger the disease. IBD therapy aims at improving the quality of life and reducing the risk of disease-related complications to avoid the need for surgery. There is no specific cure for IBDs, and the focus of therapy is supportive measures and use of anti-inflammatory and immunosuppressive drugs. Glucocorticoids (GCs) are powerful anti-inflammatory and immunomodulatory agents used to treat many acute and chronic inflammatory diseases. GCs remain basic treatment for moderate-to-severe IBD, but their use is limited by several important adverse drug effects. Topical administration of a second-generation of GCs, such as budesonide and beclomethasone dipropionate (BDP), represents a valid alternative to use of older, systemic GCs. Administration of second-generation GCs shows promisingly high topical activity and less systemic toxicity, but maintenance therapy with these new GCs in IBD patients is associated with multiple adverse effects. In this review, we make a comparative analysis of the efficacy of first-generation and second-generation GCs in IBD treatment. Unraveling GC biology at the molecular level to uncouple their clinical benefits from detrimental effects is important. One approach is to consider new GC mediators, such as glucocorticoid-induced leucine zipper, which may have similar anti-inflammatory properties, but avoids the side effects of GCs. This in-depth analysis can help to improve the development and the clinical outcomes of GC therapies in IBD.
Collapse
Affiliation(s)
- Stefano Bruscoli
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | | | | |
Collapse
|
44
|
Cavone L, McCann T, Drake LK, Aguzzi EA, Oprişoreanu AM, Pedersen E, Sandi S, Selvarajah J, Tsarouchas TM, Wehner D, Keatinge M, Mysiak KS, Henderson BEP, Dobie R, Henderson NC, Becker T, Becker CG. A unique macrophage subpopulation signals directly to progenitor cells to promote regenerative neurogenesis in the zebrafish spinal cord. Dev Cell 2021; 56:1617-1630.e6. [PMID: 34033756 DOI: 10.1016/j.devcel.2021.04.031] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 03/15/2021] [Accepted: 04/28/2021] [Indexed: 12/14/2022]
Abstract
Central nervous system injury re-initiates neurogenesis in anamniotes (amphibians and fishes), but not in mammals. Activation of the innate immune system promotes regenerative neurogenesis, but it is fundamentally unknown whether this is indirect through the activation of known developmental signaling pathways or whether immune cells directly signal to progenitor cells using mechanisms that are unique to regeneration. Using single-cell RNA-seq of progenitor cells and macrophages, as well as cell-type-specific manipulations, we provide evidence for a direct signaling axis from specific lesion-activated macrophages to spinal progenitor cells to promote regenerative neurogenesis in zebrafish. Mechanistically, TNFa from pro-regenerative macrophages induces Tnfrsf1a-mediated AP-1 activity in progenitors to increase regeneration-promoting expression of hdac1 and neurogenesis. This establishes the principle that macrophages directly communicate to spinal progenitor cells via non-developmental signals after injury, providing potential targets for future interventions in the regeneration-deficient spinal cord of mammals.
Collapse
Affiliation(s)
- Leonardo Cavone
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Tess McCann
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Louisa K Drake
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Erika A Aguzzi
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Ana-Maria Oprişoreanu
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Elisa Pedersen
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Soe Sandi
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Jathurshan Selvarajah
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Themistoklis M Tsarouchas
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Daniel Wehner
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Max Planck Institute for the Science of Light, Staudtstraße 2, Erlangen 91058, Germany; Max-Planck-Zentrum für Physik und Medizin, Staudtstraße 2, Erlangen 91058, Germany
| | - Marcus Keatinge
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Karolina S Mysiak
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Beth E P Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK.
| | - Catherina G Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Euan MacDonald Centre for Motor Neurone Disease Research University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
45
|
Glucocorticoid-induced leucine zipper regulates liver fibrosis by suppressing CCL2-mediated leukocyte recruitment. Cell Death Dis 2021; 12:421. [PMID: 33927191 PMCID: PMC8085011 DOI: 10.1038/s41419-021-03704-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 12/16/2022]
Abstract
Liver fibrosis (LF) is a dangerous clinical condition with no available treatment. Inflammation plays a critical role in LF progression. Glucocorticoid-induced leucine zipper (GILZ, encoded in mice by the Tsc22d3 gene) mimics many of the anti-inflammatory effects of glucocorticoids, but its role in LF has not been directly addressed. Here, we found that GILZ deficiency in mice was associated with elevated CCL2 production and pro-inflammatory leukocyte infiltration at the early LF stage, resulting in enhanced LF development. RNA interference-mediated in vivo silencing of the CCL2 receptor CCR2 abolished the increased leukocyte recruitment and the associated hepatic stellate cell activation in the livers of GILZ knockout mice. To highlight the clinical relevance of these findings, we found that TSC22D3 mRNA expression was significantly downregulated and was inversely correlated with that of CCL2 in the liver samples of patients with LF. Altogether, these data demonstrate a protective role of GILZ in LF and uncover the mechanism, which can be targeted therapeutically. Therefore, modulating GILZ expression and its downstream targets represents a novel avenue for pharmacological intervention for treating LF and possibly other liver inflammatory disorders.
Collapse
|
46
|
Braz-de-Melo HA, Faria SS, Pasquarelli-do-Nascimento G, Santos IDO, Kobinger GP, Magalhães KG. The Use of the Anticoagulant Heparin and Corticosteroid Dexamethasone as Prominent Treatments for COVID-19. Front Med (Lausanne) 2021; 8:615333. [PMID: 33968948 PMCID: PMC8102695 DOI: 10.3389/fmed.2021.615333] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/26/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is spreading worldwide at disturbing rates, overwhelming global healthcare. Mounting death cases due to disease complications highlight the necessity of describing efficient drug therapy strategies for severe patients. COVID-19 severity associates with hypercoagulation and exacerbated inflammation, both influenced by ACE2 downregulation and cytokine storm occurrence. In this review, we discuss the applicability of the anticoagulant heparin and the anti-inflammatory corticosteroid dexamethasone for managing severe COVID-19 patients. The upregulated inflammation and blood clotting may be mitigated by administrating heparin and its derivatives. Heparin enhances the anticoagulant property of anti-thrombin (AT) and may be useful in conjunction with fibrinolytic drugs for severe COVID-19 patients. Besides, heparin can also modulate immune responses, alleviating TNF-α-mediated inflammation, impairing IL-6 production and secretion, and binding to complement proteins and leukotriene B4 (LTB4). Moreover, heparin may present anti-SARS-CoV-2 potential once it can impact viral infectivity and alter SARS-CoV-2 Spike protein architecture. Another feasible approach is the administration of the glucocorticoid dexamethasone. Although glucocorticoid's administration for viral infection managing is controversial, there is increasing evidence demonstrating that dexamethasone treatment is capable of drastically diminishing the death rate of patients presenting with Acute Respiratory Distress Syndrome (ARDS) that required invasive mechanical ventilation. Importantly, dexamethasone may be detrimental by impairing viral clearance and inducing hyperglycemia and sodium retention, hence possibly being deleterious for diabetics and hypertensive patients, two major COVID-19 risk groups. Therefore, while heparin's multitarget capacity shows to be strongly beneficial for severe COVID-19 patients, dexamethasone should be carefully administered taking into consideration underlying medical conditions and COVID-19 disease severity. Therefore, we suggest that the multitarget impact of heparin as an anti-viral, antithrombotic and anti-inflammatory drug in the early stage of the COVID-19 could significantly reduce the need for dexamethasone treatment in the initial phase of this disease. If the standard treatment of heparins fails on protecting against severe illness, dexamethasone must be applied as a potent anti-inflammatory shutting-down the uncontrolled and exacerbated inflammation.
Collapse
Affiliation(s)
| | - Sara Socorro Faria
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | | | - Igor de Oliveira Santos
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Gary P Kobinger
- Département de Microbiologie-Infectiologie et d'Immunologie, Université Laval, Quebec City, QC, Canada.,Centre de Recherche en Infectiologie du CHU de Québec, Université Laval, Quebec City, QC, Canada
| | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
47
|
Stoynova MA, Shinkov AD, Georgiev GK, Kovatcheva RD. Association between Clinical Activity Score and Serum Interleukin-6, Interleukin-8 and Interleukin-10 during Systemic Glucocorticoid Treatment for Active Moderate-to-Severe Graves' Orbitopathy. Curr Eye Res 2021; 46:1503-1508. [PMID: 33849364 DOI: 10.1080/02713683.2021.1912783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Purpose: Some interleukins (ILs) play an important role in Graves' orbitopathy (GO) pathogenesis. We aimed to compare serum IL-6, IL-8 and IL-10 in GO patients, patients with Graves' disease (GD) without GO and healthy controls (HC); to follow IL changes during glucocorticoid (GC) treatment for GO; to examine associations between ILs and Clinical Activity Score (CAS).Materials and Methods: Thirty-one patients with active moderate-to-severe GO (GO(+) group), 30 patients with GD without GO (GO(-) group) and 30 HC were enrolled. At baseline, ILs were measured in all groups, CAS was evaluated in GO(+) patients, who were then treated with systemic GCs for 12 weeks. ILs and CAS were reassessed after the first week of treatment (W2) and at the end of the therapy (W12).Results: At baseline, IL-6 was significantly higher in GO(+) and GO(-) patients, IL-8 - higher in GO(-) patients and IL-10 - lower in GO(+) patients compared to HC. Baseline ILs did not correlate with CAS. At W2, all ILs and CAS decreased significantly. At W12, CAS decreased further, IL-6 remained low, IL-8 and IL-10 returned to baseline. CAS reduction correlated positively with IL-6 reduction at W12 (ρ = 0.38, p = .04).GO(+) patients with overall CAS reduction≥2 had higher baseline IL-6 (3.4 vs 2.6 pg/ml, p = .15), smaller IL-10 reduction at W2 (10.5 vs 18.2%, p = .09), lower IL-6 (1.4 vs 2.4 pg/ml, p < .01) and higher IL-6 reduction at W12 (48.6 vs 21.4%, p = .01) compared to patients with CAS reduction<2. Logistic regression analysis confirmed that overall CAS reduction≥2 was associated with higher baseline IL-6, lower IL-6 at W12 and smaller IL-10 reduction at W2 (R2 = 0.66).Conclusions: Higher baseline IL-6, lower IL-6 at W12 and smaller IL-10 reduction at W2 were associated with higher probability of significant overall CAS reduction. IL-6 might be a potential additional marker for assessing disease activity.
Collapse
|
48
|
Dubashynskaya NV, Bokatyi AN, Skorik YA. Dexamethasone Conjugates: Synthetic Approaches and Medical Prospects. Biomedicines 2021; 9:341. [PMID: 33801776 PMCID: PMC8067246 DOI: 10.3390/biomedicines9040341] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/20/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Dexamethasone (DEX) is the most commonly prescribed glucocorticoid (GC) and has a wide spectrum of pharmacological activity. However, steroid drugs like DEX can have severe side effects on non-target organs. One strategy to reduce these side effects is to develop targeted systems with the controlled release by conjugation to polymeric carriers. This review describes the methods available for the synthesis of DEX conjugates (carbodiimide chemistry, solid-phase synthesis, reversible addition fragmentation-chain transfer [RAFT] polymerization, click reactions, and 2-iminothiolane chemistry) and perspectives for their medical application as GC drug or gene delivery systems for anti-tumor therapy. Additionally, the review focuses on the development of DEX conjugates with different physical-chemical properties as successful delivery systems in the target organs such as eye, joint, kidney, and others. Finally, polymer conjugates with improved transfection activity in which DEX is used as a vector for gene delivery in the cell nucleus have been described.
Collapse
Affiliation(s)
| | | | - Yury A. Skorik
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, 199004 St. Petersburg, Russia; (N.V.D.); (A.N.B.)
| |
Collapse
|
49
|
Martínez-Botía P, Bernardo Á, Acebes-Huerta A, Caro A, Leoz B, Martínez-Carballeira D, Palomo-Antequera C, Soto I, Gutiérrez L. Clinical Management of Hypertension, Inflammation and Thrombosis in Hospitalized COVID-19 Patients: Impact on Survival and Concerns. J Clin Med 2021; 10:jcm10051073. [PMID: 33806709 PMCID: PMC7961450 DOI: 10.3390/jcm10051073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/13/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
The most severe clinical manifestations of the Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), are due to an unbalanced immune response and a pro-thrombotic hemostatic disturbance, with arterial hypertension or diabetes as acknowledged risk factors. While waiting for a specific treatment, the clinical management of hospitalized patients is still a matter of debate, and the effectiveness of treatments to manage clinical manifestations and comorbidities has been questioned. In this study, we aim to assess the impact of the clinical management of arterial hypertension, inflammation and thrombosis on the survival of COVID-19 patients. The Spanish cohorts included in this observational retrospective study are from HM Hospitales (2035 patients) and from Hospital Universitario Central de Asturias (72 patients). Kaplan Meier survival curves, Cox regression and propensity score matching analyses were employed, considering demographic variables, comorbidities and treatment arms (when opportune) as covariates. The management of arterial hypertension with angiotensin-converting enzyme 2 (ACE2) inhibitors or angiotensin receptor blockers is not detrimental, as was initially reported, and neither was the use of non-steroidal anti-inflammatory drugs (NSAIDs). On the contrary, our analysis shows that the use on itself of corticosteroids is not beneficial. Importantly, the management of COVID-19 patients with low molecular weight heparin (LMWH) as an anticoagulant significantly improves the survival of hospitalized patients. These results delineate the current treatment options under debate, supporting the effectiveness of thrombosis prophylaxis on COVID-19 patients as a first-line treatment without the need for compromising the treatment of comorbidities, while suggesting cautiousness when administering corticosteroids.
Collapse
Affiliation(s)
- Patricia Martínez-Botía
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
| | - Ángel Bernardo
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
- Clinical Diagnosis Laboratory, Department of Hematology, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Andrea Acebes-Huerta
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
| | - Alberto Caro
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
- Department of Hematology, Hemostasis Section, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Blanca Leoz
- Department of Intensive Medicine, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain;
- Translational Microbiology Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Daniel Martínez-Carballeira
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
- Department of Hematology, Hemostasis Section, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Carmen Palomo-Antequera
- Department of Internal Medicine, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain;
- Bone Metabolism, Vascular Metabolism and Chronic Inflammatory Diseases Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Inmaculada Soto
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
- Department of Hematology, Hemostasis Section, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
- Correspondence: (I.S.); (L.G.)
| | - Laura Gutiérrez
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (P.M.-B.); (Á.B.); (A.A.-H.); (A.C.); (D.M.-C.)
- Department of Medicine, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Correspondence: (I.S.); (L.G.)
| |
Collapse
|
50
|
Ronchetti S, Ayroldi E, Ricci E, Gentili M, Migliorati G, Riccardi C. A Glance at the Use of Glucocorticoids in Rare Inflammatory and Autoimmune Diseases: Still an Indispensable Pharmacological Tool? Front Immunol 2021; 11:613435. [PMID: 33584696 PMCID: PMC7874096 DOI: 10.3389/fimmu.2020.613435] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022] Open
Abstract
Since their discovery, glucocorticoids (GCs) have been used to treat almost all autoimmune and chronic inflammatory diseases, as well as allergies and some forms of malignancies, because of their immunosuppressive and anti-inflammatory effects. Although GCs provide only symptomatic relief and do not eliminate the cause of the pathology, in the majority of treatments, GCs frequently cannot be replaced by other classes of drugs. Consequently, long-term treatments cause adverse effects that may, in turn, lead to new pathologies that sometimes require the withdrawal of GC therapy. Therefore, thus far, researchers have focused their efforts on molecules that have the same efficacy as that of GCs but cause fewer adverse effects. To this end, some GC-induced proteins, such as glucocorticoid-induced leucine zipper (GILZ), have been used as drugs in mouse models of inflammatory pathologies. In this review, we focus on some important but rare autoimmune and chronic inflammatory diseases for which the biomedical research investment in new therapies is less likely. Additionally, we critically evaluate the possibility of treating such diseases with other drugs, either GC-related or unrelated.
Collapse
Affiliation(s)
- Simona Ronchetti
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emira Ayroldi
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Erika Ricci
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marco Gentili
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Graziella Migliorati
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Carlo Riccardi
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|