1
|
Piechota M, Zieba M, Borczyk M, Golda S, Hajto J, Skupio U, Slezak M, Parkitna JR, Korostynski M. A cross-tissue transcriptomic approach decodes glucocorticoid receptor-dependent links to human metabolic phenotypes. BMC Genomics 2025; 26:462. [PMID: 40346507 PMCID: PMC12063231 DOI: 10.1186/s12864-025-11676-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025] Open
Abstract
Glucocorticoids, acting through the glucocorticoid receptor (GR), control metabolism, maintain homeostasis, and enable adaptive responses to environmental challenges. Their function has been comprehensively studied, leading to identification of numerous tissue-specific GR-dependent mechanisms. Abundant evidence shows that GR-triggered responses differ across tissues, however, the extent of this specificity was not comprehensively explored. It is also unknown how particular GR-induced molecular patterns are translated into profile of higher-level human traits. Here, we examine cross-tissue effects of GR activation on gene expression. We assessed changes induced by stimulation with GR agonist, dexamethasone in nine tissues (adrenal cortex, perigonadal adipose tissue, hypothalamus, liver, kidney, anterior thigh muscle, pituitary gland, spleen, and lungs) in adult male C57BL/6 mice, using whole-genome microarrays. Dexamethasone induced balanced transcriptional responses across all examined tissues with 585 identified dexamethasone-regulated transcripts, including 446 with significant treatment-tissue interaction effects. Clustering analysis revealed sixteen GR-dependent patterns, including those universal across tissues and tissue-specific. We leveraged existing gene annotations and created new annotation sets based on chromatin immunoprecipitation sequencing, recent large-scale genome-wide association studies, and human transcriptome collections. As expected, GR-dependent transcripts were associated with essential metabolic processes (glycolysis/gluconeogenesis, lipid-metabolism) and inflammation-related pathways. Beyond these, we found novel links between regulated gene patterns and human phenotypic traits, like reticulocyte count or blood triglyceride levels. Overall effects of GR stimulation are well coordinated and closely linked to biological roles of tissues and organs. Our findings provide novel insights into complex systemic and tissue-specific actions of glucocorticoids and their potential impacts on human physiology and pathology.
Collapse
Affiliation(s)
- Marcin Piechota
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, 31-343, Poland
| | - Mateusz Zieba
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, 31-343, Poland
| | - Małgorzata Borczyk
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, 31-343, Poland
| | - Slawomir Golda
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, 31-343, Poland
| | - Jacek Hajto
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, 31-343, Poland
| | - Urszula Skupio
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, 31-343, Poland
| | - Michal Slezak
- Lukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, 54-066, Poland
| | - Jan Rodriguez Parkitna
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, 31-343, Poland
| | - Michal Korostynski
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, 31-343, Poland.
| |
Collapse
|
2
|
Jo Y, Greene TT, Chiale C, Zhang K, Fang Z, Dallari S, Marooki N, Wang W, Zuniga EI. Genomic analysis of progenitors in viral infection implicates glucocorticoids as suppressors of plasmacytoid dendritic cell generation. Proc Natl Acad Sci U S A 2025; 122:e2410092122. [PMID: 40294270 PMCID: PMC12067256 DOI: 10.1073/pnas.2410092122] [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: 05/20/2024] [Accepted: 02/19/2025] [Indexed: 04/30/2025] Open
Abstract
Plasmacytoid Dendritic cells (pDCs) are the most potent producers of interferons, which are critical antiviral cytokines. pDC development is, however, compromised following a viral infection, and this phenomenon, as well as its relationship to conventional (c)DC development is still incompletely understood. By using lymphocytic choriomeningitis virus (LCMV) infection in mice as a model system, we observed that DC progenitors skewed away from pDC and toward cDC development during in vivo viral infection. Subsequent characterization of the transcriptional and epigenetic landscape of fms-like tyrosine kinase 3+ (Flt3+) DC progenitors and follow-up studies revealed increased apoptosis and reduced proliferation in different individual DC-progenitors as well as a profound type I interferon (IFN-I)-dependent ablation of pre-pDCs, but not pre-DC precursors, after both acute and chronic LCMV infections. In addition, integrated genomic analysis identified altered activity of 34 transcription factors in Flt3+ DC progenitors from infected mice, including two regulators of Glucocorticoid (GC) responses. Subsequent studies demonstrated that addition of GCs to DC progenitors led to downregulated pDC-primed-genes while upregulating cDC-primed-genes, and that endogenous GCs selectively decreased pDC, but not cDC, numbers upon in vivo LCMV infection. These findings demonstrate a significant ablation of pre-pDCs in infected mice and identify GCs as suppressors of pDC generation from early progenitors. This provides a potential explanation for the impaired pDC development following viral infection and links pDC numbers to the hypothalamic-pituitary-adrenal axis.
Collapse
Affiliation(s)
- Yeara Jo
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Trever T. Greene
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Carolina Chiale
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Kai Zhang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA92093
| | - Ziyan Fang
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Simone Dallari
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Nuha Marooki
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Wei Wang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA92093
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA92093
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA92093
| | - Elina I. Zuniga
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| |
Collapse
|
3
|
Chen M, He Y, Jia Y, Wu L, Zhao R. Liver transcriptome response to avian pathogenic Escherichia coli infection in broilers with corticosterone treatment. Poult Sci 2025; 104:105020. [PMID: 40088534 PMCID: PMC11937665 DOI: 10.1016/j.psj.2025.105020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
Avian pathogenic Escherichia coli (APEC) infection has high morbidity and mortality, and multiple stressors encountered during rearing place poultry in a state of stress. However, research on how poultry cope with APEC infection under stress situation is still limited. In this study, we established a broiler stress model by corticosterone (CORT) administration subcutaneously for 7 consecutive days, followed by APEC challenge intramuscularly. CORT treatment significantly reduced body weight (BW) and average daily gain (ADG) while increasing feed conversion ratio (FCR) (P < 0.01). APEC infection significantly decreased ADG (P < 0.01). CORT treatment and APEC infection elevated plasma corticosterone and heterophil to lymphocyte (H/L) ratio (P < 0.05). Additionally, plasma aspartate aminotransferase (AST), AST to alanine aminotransferase (AST/ALT) ratio, and lactate dehydrogenase (LDH) levels increased significantly (P < 0.01). Histopathological analysis revealed structural damage of liver, indicating that CORT treatment and APEC infection induced liver injury. However, CORT pretreatment broilers exhibited a milder histopathological lesions and significantly lower AST, ALT, and LDH levels (P < 0.05) compared to APEC infection alone. CORT treatment and APEC infection increased plasma levels of lysozyme (LZM), total protein (TP), and globulin (GLOB) (P < 0.05), while CORT pretreatment further elevating their concentrations compared to APEC infection alone, suggesting an enhanced innate immune response. Liver transcriptomic analysis identified 768, 335, and 567 differentially expressed genes (DEGs) following CORT, APEC, or both treatments, respectively, enriched in cytokine-cytokine receptor interaction, PPAR signaling pathway, Toll-like receptor signaling pathway, MAPK signaling pathway, steroid hormone biosynthesis pathway, arachidonic acid metabolism, and phagosome pathway, etc., indicating that CORT treatment regulates lipid metabolism and immunity, while APEC infection induces inflammation and disrupts lipid metabolism. Notably, CORT pretreatment may mitigate APEC induced liver injury by enhancing phagosome function. Moreover, glucocorticoid receptor (GR) may regulate DEGs expressions, thus affected broilers response to CORT, APEC, or both treatments. These results suggest that CORT treatment, APEC infection, or both significantly affect the growth performance, immune response and liver function of broilers, while lipid metabolism may play a crucial role.
Collapse
Affiliation(s)
- Mengru Chen
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Yifei He
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Yimin Jia
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Lei Wu
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China.
| |
Collapse
|
4
|
Knauer-Arloth J, Hryhorzhevska A, Binder EB. Multiomics Analysis of the Molecular Response to Glucocorticoids: Insights Into Shared Genetic Risk From Psychiatric to Medical Disorders. Biol Psychiatry 2025; 97:794-805. [PMID: 39393618 DOI: 10.1016/j.biopsych.2024.10.004] [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: 03/06/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Alterations in the effects of glucocorticoids have been implicated in mediating some of the negative health effects associated with chronic stress, including increased risk for psychiatric disorders and cardiovascular and metabolic diseases. In this study, we investigated how genetic variants influence gene expression and DNA methylation in response to glucocorticoid receptor (GR) activation and their association with disease risk. METHODS We measured DNA methylation (n = 199) and gene expression (n = 297) in peripheral blood before and after GR activation with dexamethasone, with matched genotype data available for all samples. A comprehensive molecular quantitative trait locus (QTL) analysis was conducted, mapping GR-response methylation (me)QTLs, GR-response expression (e)QTLs, and GR-response expression quantitative trait methylation (eQTMs). A multilevel network analysis was employed to map the complex relationships between the transcriptome, epigenome, and genetic variation. RESULTS We identified 3772 GR-response meCpGs corresponding to 104,828 local GR-response meQTLs that did not strongly overlap with baseline meQTLs. eQTM and eQTL analyses revealed distinct genetic influences on gene expression and DNA methylation. Multilevel network analysis uncovered GR-response network trio QTLs, characterized by SNP-CpG-transcript combinations where meQTLs act as both eQTLs and eQTMs. GR-response trio variants were enriched in a genome-wide association study for psychiatric, respiratory, autoimmune, and cardiovascular diseases and conferred a higher relative heritability per SNP than GR-response meQTL and baseline QTL SNPs. CONCLUSIONS Genetic variants modulating the molecular effects of glucocorticoids are associated with psychiatric as well as medical diseases and not uncovered in baseline QTL analyses.
Collapse
Affiliation(s)
- Janine Knauer-Arloth
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany; Institute of Computational Biology, Helmholtz Munich, Neuherberg, Germany.
| | | | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
5
|
Wang G, Zheng Q, Ma W, Yang E, Jing S, Zhang L, Jin Q, He Q, Li X, Wang Z. Impact of intraoperative furosemide and dexamethasone on complications following mini-percutaneous nephrolithotripsy: a retrospective propensity score-matched cohort study. BMC Urol 2025; 25:88. [PMID: 40221668 PMCID: PMC11992847 DOI: 10.1186/s12894-025-01778-8] [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: 09/24/2024] [Accepted: 04/08/2025] [Indexed: 04/14/2025] Open
Abstract
OBJECTIVE To evaluate the impact of intraoperative use of furosemide (FUR) in combination with dexamethasone (DEX) on postoperative complications following mini-percutaneous nephrolithotripsy (mini-PCNL). PATIENTS AND METHODS The study was a retrospective cohort analysis of adult patients with kidney calculi treated with mini-PCNL. Exposure was the intravenous administration of FUR and DEX during mini-PCNL. The primary outcome was postoperative fever (≥ 38°C), whereas the secondary outcomes were other complications. Propensity score matching (PSM) was performed at a 1:1 ratio. Subgroup analyses and interaction tests were used to examine differences among different demographic groups. RESULTS The pre-matched and propensity score-matched cohorts included 237 and 166 patients, respectively. In the PSM cohort, postoperative fever (≥ 38°C) occurred in 8.4% (7/83) of the FUR + DEX group and 20.5% (17/83) of the control group. The combined use of FUR and DEX was associated with a lower postoperative fever (P = 0.027). There was no statistically significant difference between the FUR + DEX group and the control group for other complications, including SIRS, urosepsis, and pain-requiring opioids. SIRS occurred in 4.8% (4/83) of the FUR + DEX group versus 8.4% (7/83) in the control group, while urosepsis rates were 2.4% (2/83) versus 3.6% (3/83), respectively. Subgroup analysis showed a significant reduction in postoperative fever in patients with an operation time of ≥ 2 h in the FUR + DEX group, as indicated by the interaction test (P = 0.05). CONCLUSION The intravenous combined use of FUR and DEX in mini-PCNL reduces postoperative fever (≥ 38°C), particularly benefiting patients with an operative time of ≥ 2 h.
Collapse
Affiliation(s)
- Guilin Wang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Qihui Zheng
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Wentao Ma
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Enguang Yang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Suoshi Jing
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Urology, The First Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Luyang Zhang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Qi Jin
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Qiqi He
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xiaoran Li
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| | - Zhiping Wang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| |
Collapse
|
6
|
Sueyoshi T, Petrillo MG, Jewell CM, Bortner CD, Perera L, Xu X, Aguayo FI, Diaz-Jimenez D, Robinson AG, Cook ME, Oakley RH, Cidlowski JA. Molecular interactions of glucocorticoid and mineralocorticoid receptors define novel transcription and biological functions. J Biol Chem 2025; 301:108488. [PMID: 40209952 DOI: 10.1016/j.jbc.2025.108488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 03/26/2025] [Accepted: 03/29/2025] [Indexed: 04/12/2025] Open
Abstract
Glucocorticoids are primary stress hormones necessary for life that function to maintain homeostasis. These hormones and their synthetic derivatives are widely used in the clinic to combat disease but are limited by development of resistance and by severe side effects. Understanding how glucocorticoids signal is crucial for developing safer and more effective glucocorticoids. Mechanistically glucocorticoid ligands induce glucocorticoid receptor (GR) homodimerization and regulation of gene expression. Here, we show that GR and mineralocorticoid receptor (MR) form molecular complexes with distinct transcriptional responses that alter the biological roles of GR. MR inhibited GR interaction with genomic DNA and diminished glucocorticoid-regulated gene expression as well as suppressed cell apoptosis induced by GR signaling. Provocatively, multiple therapeutic glucocorticoids differentially induced the GR-MR interaction revealing unknown drug effects that are exploitable for fine-tuning glucocorticoid drug treatments. Molecular modeling of the GR-MR complex predicted an interaction interface residing in the LBD of both GR and MR. Mutation of a key amino acid in the interface of GR compromised GR-MR interaction without affecting GR activity in a gene reporter assay. Overall, our findings uncovered unique crosstalk mechanisms between distinct nuclear receptors providing a novel mechanism of diversity in the action of glucocorticoids that may contribute to context-dependent GR signaling in human health and disease.
Collapse
Affiliation(s)
- Tatsuya Sueyoshi
- Molecular Endocrinology Group, Signal Transduction Laboratory, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA
| | - Maria G Petrillo
- Molecular Endocrinology Group, Signal Transduction Laboratory, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA
| | - Christine M Jewell
- Molecular Endocrinology Group, Signal Transduction Laboratory, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA
| | - Carl D Bortner
- Flow Cytometry Center, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA
| | - Lalith Perera
- Computational Chemistry & Molecular Modeling Support Group, Genomic Integrity & Structural Biology Laboratory, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA
| | - Xiaojiang Xu
- Integrative Bioinformatics Group, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA; Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Felipe I Aguayo
- Molecular Endocrinology Group, Signal Transduction Laboratory, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA
| | - David Diaz-Jimenez
- Molecular Endocrinology Group, Signal Transduction Laboratory, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA
| | - Anastasia G Robinson
- Molecular Endocrinology Group, Signal Transduction Laboratory, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA
| | - Molly E Cook
- Epigenomics and DNA Sequence Core Facility, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA
| | - Robert H Oakley
- Molecular Endocrinology Group, Signal Transduction Laboratory, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA
| | - John A Cidlowski
- Molecular Endocrinology Group, Signal Transduction Laboratory, NIEHS, NIH, DHHS, Raleigh, North Carolina, USA.
| |
Collapse
|
7
|
Sun T, Bi X, Yang N, Zhang X, Chu J, Li L, Liu H, Tang R, Lin R. Glucocorticoid receptor inhibits Th2 immune responses by down-regulating Pparg and Gata3 in schistosomiasis. Front Immunol 2025; 16:1518586. [PMID: 40196108 PMCID: PMC11973390 DOI: 10.3389/fimmu.2025.1518586] [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: 10/28/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction The Th2 immune response plays a pivotal role in the pathogenesis of schistosomiasis, contributing to the formation of hepatic granulomas and fibrosis. While the glucocorticoid receptor (GR) is a ubiquitously expressed nuclear receptor that mediates anti-inflammatory effects, its impact on Th2 responses in schistosomiasis remains underexplored. Thus, this study aimed to investigate the potential impact of GR activation on the hepatic Th2 immune response in schistosomiasis using the synthetic glucocorticoid dexamethasone. Method In vivo, Schistosoma japonicum-infected mice were treated with dexamethasone, while in vitro studies were conducted on Th2 cells. Additionally, RNA sequencing and single-cell sequencing were integrated to identify key transcription factors influenced by GR activation during Th2 cell differentiation, with gene expression levels validated both in vivo and in vitro. Results In vivo, GR activation markedly reduced the size of Schistosoma egg granulomas and substantially repressed the transcription of key Th2-related cytokines, such as IL-4, IL-5, and IL-13. In vitro, GR activation inhibited the transcription of IL-4, IL-5, and IL-13, as well as the secretion of IL-4 in Th2 cells. An integrated analysis of RNA sequencing and single-cell sequencing revealed that GR activation downregulated the expression of two major transcription factors, Gata3 and Pparg, which were elevated in infected mouse livers and Th2 cells but decreased following dexamethasone treatment. Conclusion GR activation may suppress the Th2 immune response triggered by egg antigens by downregulating the expression of the key transcription factors Gata3 and Pparg. While these findings provide insights into a potential complementary therapeutic strategy, further research is necessary to assess the feasibility and safety of targeting GR activation for the treatment of schistosomiasis.
Collapse
Affiliation(s)
- Tao Sun
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xiaojuan Bi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ning Yang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xue Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jin Chu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Liang Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hui Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Rui Tang
- Department of Tropical Infectious Diseases, Naval Medical University, Shanghai, China
| | - Renyong Lin
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
8
|
Ding X, Huang H, Chen Y, Wu J, Yan X, Ding Y, Dong J, Wang Y, Wang L, Tan Q, Yang C. Electrospun 11β-HSD1 Inhibitor-Loaded Scaffolds for Accelerating Diabetic Ulcer Healing. ACS APPLIED BIO MATERIALS 2025; 8:435-445. [PMID: 39690109 DOI: 10.1021/acsabm.4c01397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Diabetic ulcers (DUs) are a common and severe complication of diabetes, characterized by impaired wound healing due to a complex pathophysiological mechanism. Elevated levels of 11β-hydroxysteroid dehydrogenase type I (11β-HSD1) in wounds have been demonstrated to modulate glucocorticoid activity, leading to delayed skin cell proliferation and restricted angiogenesis, ultimately hindering wound healing. In this study, we propose an electrospun poly(ε-caprolactone) (PCL) nanofiber scaffold doped with the 11β-HSD1 inhibitor BVT2733 (BPs) to prevent 11β-HSD1 activity during the diabetic wound healing process. The electrospun scaffold loaded with BVT2733 is designed to achieve localized inhibition of 11β-HSD1 in DUs. This scaffold exhibited a porous morphology and desirable drug-loading capacity, meeting the requirements for wound coverage and effective delivery of BVT2733 BPs. In vitro studies demonstrated that the sustained release of BVT2733 from the scaffold promoted skin cell proliferation and migration while stimulating angiogenesis by upregulating HIF1-α/VEGF expression. The therapeutic effect of the scaffold was further confirmed in a full-thickness wound model using diabetic mice. The mice treated with the scaffolds exhibited an accelerated wound healing rate, increased neovascularization, enhanced collagen deposition, and regeneration of skin appendages within 2 weeks postinjury. The findings here provide evidence for the use of 11β-HSD1 inhibitor-integrated biomaterials in treating DUs and represent a novel biological platform for modulating dysregulated mechanisms in DUs.
Collapse
Affiliation(s)
- Xiaofeng Ding
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Heyan Huang
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yutong Chen
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Junchao Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Xin Yan
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Youjun Ding
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jie Dong
- Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yiwei Wang
- Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lili Wang
- Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210003, China
| | - Qian Tan
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chenxi Yang
- Jiangsu Provincial Engineering Research Centre of TCM External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Department of Immunology, School of Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, China
| |
Collapse
|
9
|
McSwiggen DT, Liu H, Tan R, Agramunt Puig S, Akella LB, Berman R, Bretan M, Chen H, Darzacq X, Ford K, Godbey R, Gonzalez E, Hanuka A, Heckert A, Ho JJ, Johnson SL, Kelso R, Klammer A, Krishnamurthy R, Li J, Lin K, Margolin B, McNamara P, Meyer L, Pierce SE, Sule A, Stashko C, Tang Y, Anderson DJ, Beck HP. A high-throughput platform for single-molecule tracking identifies drug interaction and cellular mechanisms. eLife 2025; 12:RP93183. [PMID: 39786807 PMCID: PMC11717362 DOI: 10.7554/elife.93183] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
The regulation of cell physiology depends largely upon interactions of functionally distinct proteins and cellular components. These interactions may be transient or long-lived, but often affect protein motion. Measurement of protein dynamics within a cellular environment, particularly while perturbing protein function with small molecules, may enable dissection of key interactions and facilitate drug discovery; however, current approaches are limited by throughput with respect to data acquisition and analysis. As a result, studies using super-resolution imaging are typically drawing conclusions from tens of cells and a few experimental conditions tested. We addressed these limitations by developing a high-throughput single-molecule tracking (htSMT) platform for pharmacologic dissection of protein dynamics in living cells at an unprecedented scale (capable of imaging >106 cells/day and screening >104 compounds). We applied htSMT to measure the cellular dynamics of fluorescently tagged estrogen receptor (ER) and screened a diverse library to identify small molecules that perturbed ER function in real time. With this one experimental modality, we determined the potency, pathway selectivity, target engagement, and mechanism of action for identified hits. Kinetic htSMT experiments were capable of distinguishing between on-target and on-pathway modulators of ER signaling. Integrated pathway analysis recapitulated the network of known ER interaction partners and suggested potentially novel, kinase-mediated regulatory mechanisms. The sensitivity of htSMT revealed a new correlation between ER dynamics and the ability of ER antagonists to suppress cancer cell growth. Therefore, measuring protein motion at scale is a powerful method to investigate dynamic interactions among proteins and may facilitate the identification and characterization of novel therapeutics.
Collapse
Affiliation(s)
| | - Helen Liu
- Eikon Therapeutics IncHaywardUnited States
| | | | | | | | | | | | | | - Xavier Darzacq
- Eikon Therapeutics IncHaywardUnited States
- University of California, BerkeleyBerkeleyUnited States
| | | | | | | | - Adi Hanuka
- Eikon Therapeutics IncHaywardUnited States
| | | | | | | | - Reed Kelso
- Eikon Therapeutics IncHaywardUnited States
| | | | | | - Jifu Li
- Eikon Therapeutics IncHaywardUnited States
| | - Kevin Lin
- Eikon Therapeutics IncHaywardUnited States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Camici M, Del Duca G, Brita AC, Antinori A. Connecting dots of long COVID-19 pathogenesis: a vagus nerve- hypothalamic-pituitary- adrenal-mitochondrial axis dysfunction. Front Cell Infect Microbiol 2024; 14:1501949. [PMID: 39735263 PMCID: PMC11671747 DOI: 10.3389/fcimb.2024.1501949] [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: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
The pathogenesis of long COVID (LC) still presents many areas of uncertainty. This leads to difficulties in finding an effective specific therapy. We hypothesize that the key to LC pathogenesis lies in the presence of chronic functional damage to the main anti-inflammatory mechanisms of our body: the three reflexes mediated by the vagus nerve, the hypothalamic-pituitary-adrenal (HPA) hormonal axis, and the mitochondrial redox status. We will illustrate that this neuro-endocrine-metabolic axis is closely interconnected and how the SARS-CoV-2 can damage it at all stages through direct, immune-inflammatory, epigenetic damage mechanisms, as well as through the reactivation of neurotropic viruses. According to our theory, the direct mitochondrial damage carried out by the virus, which replicates within these organelles, and the cellular oxidative imbalance, cannot be countered in patients who develop LC. This is because their anti-inflammatory mechanisms are inconsistent due to reduced vagal tone and direct damage to the endocrine glands of the HPA axis. We will illustrate how acetylcholine (ACh) and cortisol, with its cytoplasmatic and cellular receptors respectively, are fundamental players in the LC process. Both Ach and cortisol play multifaceted and synergistic roles in reducing inflammation. They achieve this by modulating the activity of innate and cell-mediated immunity, attenuating endothelial and platelet activation, and modulating mitochondrial function, which is crucial for cellular energy production and anti-inflammatory mechanisms. In our opinion, it is essential to study the sensitivity of the glucocorticoids receptor in people who develop LC and whether SARS-CoV-2 can cause long-term epigenetic variations in its expression and function.
Collapse
Affiliation(s)
- Marta Camici
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Giulia Del Duca
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Anna Clelia Brita
- Department of Clinical Psychology, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Andrea Antinori
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
11
|
Beck D, Cao H, Tian F, Huang Y, Jiang M, Zhao H, Tai X, Xu W, Kosasih HJ, Kealy DJ, Zhao W, Taylor SJ, Couttas TA, Song G, Chacon-Fajardo D, Walia Y, Wang M, Dowle AA, Holding AN, Bridge KS, Zhang C, Wang J, Mi JQ, Lock RB, de Bock CE, Jing D. PU.1 eviction at lymphocyte-specific chromatin domains mediates glucocorticoid response in acute lymphoblastic leukemia. Nat Commun 2024; 15:9697. [PMID: 39516193 PMCID: PMC11549222 DOI: 10.1038/s41467-024-54096-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The epigenetic landscape plays a critical role in cancer progression, yet its therapeutic potential remains underexplored. Glucocorticoids are essential components of treatments for lymphoid cancers, but resistance, driven in part by epigenetic changes at glucocorticoid-response elements, poses a major challenge to effective therapies. Here we show that glucocorticoid treatment induces distinct patterns of chromosomal organization in glucocorticoid-sensitive and resistant acute lymphoblastic leukemia xenograft models. These glucocorticoid-response elements are primed by the pioneer transcription factor PU.1, which interacts with the glucocorticoid receptor. Eviction of PU.1 promotes receptor binding, increasing the expression of genes involved in apoptosis and facilitating a stronger therapeutic response. Treatment with a PU.1 inhibitor enhances glucocorticoid sensitivity, demonstrating the clinical potential of targeting this pathway. This study uncovers a mechanism involving PU.1 and the glucocorticoid receptor, linking transcription factor activity with drug response, and suggesting potential therapeutic strategies for overcoming resistance.
Collapse
Affiliation(s)
- Dominik Beck
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Centre for Health Technologies and the School of Biomedical Engineering, University of Technology, Sydney, NSW, Australia.
| | - Honghui Cao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Tian
- Hebei Key Laboratory of Medical Data Science, Institute of Biomedical Informatics, School of Medicine, Hebei University of Engineering, Handan, Hebei Province, China
| | - Yizhou Huang
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Miao Jiang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolu Tai
- Department of Orthopedics and Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqian Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hansen J Kosasih
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - David J Kealy
- Centre for Blood Research, University of York, England, UK
| | - Weiye Zhao
- York Biomedical Research Institute, University of York, England, UK
| | - Samuel J Taylor
- Department of Cell Biology, Albert Einstein College of Medicine, Randwick, NY, USA
| | - Timothy A Couttas
- Neuroscience Research Australia, Randwick, NSW, Australia
- Brain and Mind Centre, Translational Research Collective, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Gaoxian Song
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Diego Chacon-Fajardo
- Centre for Health Technologies and the School of Biomedical Engineering, University of Technology, Sydney, NSW, Australia
| | - Yashna Walia
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Meng Wang
- Department of Orthopedics and Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Adam A Dowle
- Metabolomics & Proteomics Laboratory, Bioscience Technology Facility, Department of Biology, University of York, England, UK
| | - Andrew N Holding
- York Biomedical Research Institute, University of York, England, UK
| | | | - Chao Zhang
- Department of Orthopedics and Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Qing Mi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia.
| | - Charles E de Bock
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia.
| | - Duohui Jing
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Wang S, Ye Q. Association of cumulative methylprednisolone dosages with mortality risk from pneumonia in connective tissue disease patients. Sci Rep 2024; 14:26502. [PMID: 39489823 PMCID: PMC11532547 DOI: 10.1038/s41598-024-78233-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
Corticosteroid is essential in the treatment regimen for connective tissue disease (CTD); however, its long-term use poses significant risks, including pulmonary infections. The relationship between cumulative corticosteroid doses and adverse pneumonia outcomes requires further investigation. This study aimed to explore the association between cumulative methylprednisolone dosages (CMD) and pneumonia mortality risks among CTD patients. We conducted a retrospective analysis of data from CTD patients treated with intravenous or oral corticosteroids across six academic hospitals over approximately five years in China. We evaluated follow-up outcomes at 30 and 90 days after the onset of pneumonia. Piecewise linear regression, Cox regression analysis, and survival analysis were employed to investigate the relationship between CMD and 30-day and 90-day mortality risks. Among 335 CTD patients with pneumonia, the mean CMD was 12 g. The 30-day and 90-day mortality rates were 25.07% and 29.55%, respectively. After adjusting for potential confounders, smooth curve fitting analysis revealed a specific nonlinear relationship between CMD and 30-day and 90-day mortality risks. Cox regression analysis indicated that the lowest pneumonia mortality risk occurred when CMD ranged from 11 to 24 g (30-day adjusted hazard ratio (aHR) 0.33, 95% CI 0.14-0.77; 90-day aHR 0.37, 95% CI 0.17-0.81). Patients in the 11-24 g CMD group demonstrated significantly lower cumulative hazard and death rates compared to both the low CMD (< 11 g) and high CMD (> 24 g) groups (P < 0.05). Furthermore, interaction testing suggested that CMD's negative impact on pneumonia mortality risks was more pronounced in community-acquired pneumonia (CAP) compared to hospital-acquired pneumonia (P for interaction < 0.05). CMD shows a distinct nonlinear relationship with 30-day and 90-day pneumonia mortality risks, with potentially lower risks observed within the 11-24 g CMD range. Moreover, the varying impact of CMD on CAP mortality risk warrants further consideration in clinical management strategies.
Collapse
Affiliation(s)
- Saibin Wang
- Department of Pulmonary and Critical Care Medicine, Jinhua Municipal Central Hospital, No. 365, East Renmin Road, Jinhua, 321000, Zhejiang Province, China.
- School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China.
| | - Qian Ye
- Department of Medical Records Quality Management, Jinhua Municipal Central Hospital, Jinhua, 321000, Zhejiang Province, China
| |
Collapse
|
13
|
Ioniuc IK, Lupu A, Dragan F, Tarnita I, Alexoae MM, Streanga V, Mitrofan C, Thet AA, Nedelcu AH, Salaru DL, Burlea SL, Mitrofan EC, Lupu VV, Azoicai AN. Oxidative Stress and Antioxidants in Pediatric Asthma's Evolution and Management. Antioxidants (Basel) 2024; 13:1331. [PMID: 39594473 PMCID: PMC11590961 DOI: 10.3390/antiox13111331] [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: 09/28/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Within the pediatric population, bronchial asthma is one of the most prevalent chronic respiratory system diseases. The number of exacerbations, severity, and duration of symptoms all have a significant impact on children's life quality. In the last decades, the prevention and management strategies of this pathology have focused on maintaining or even increasing the pulmonary function to maximum levels in early childhood, as it has been demonstrated that functional deficits at this level occurring before school age cause pathological manifestations later, in adulthood. The epithelium of the airways and implicitly that of the lung is the first barrier against the lesions caused by pro-oxidative factors. Both oxidative and antioxidative factors can be of endogenous origin (produced by the body) or exogenous (from the environment or diet). Good functioning of antioxidant defense mechanisms from the molecular level to the tissue level, and a balance between pro-oxidative factors and anti- oxidative factors, influence the occurrence of compensatory mechanisms at the level of the respiratory epithelium, causing the delay of local responses to the stress induced by chronic inflammation (bronchial remodeling, thickening of airway smooth muscles, bronchoconstriction, bronchial hyper-reactivity). These mechanisms underlie the pathophysiological changes in asthma. Numerous studies carried out among the pediatric population inclusively have demonstrated the effectiveness of antioxidants in the prophylaxis, slowing down and preventing the progression of this pathology. This review complements the scientific articles, aiming at emphasizing the complexity of oxidative physio-pathological pathways and their importance in the occurrence, development, and therapeutic response in asthma, providing a good understanding of the relationship between oxidative and antioxidative factors, and being a source of future therapeutic strategies.
Collapse
Affiliation(s)
- Ileana Katerina Ioniuc
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.K.I.); (A.L.); (M.M.A.); (V.S.); (V.V.L.); (A.N.A.)
| | - Ancuta Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.K.I.); (A.L.); (M.M.A.); (V.S.); (V.V.L.); (A.N.A.)
| | - Felicia Dragan
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Irina Tarnita
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.K.I.); (A.L.); (M.M.A.); (V.S.); (V.V.L.); (A.N.A.)
| | - Monica Mihaela Alexoae
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.K.I.); (A.L.); (M.M.A.); (V.S.); (V.V.L.); (A.N.A.)
| | - Violeta Streanga
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.K.I.); (A.L.); (M.M.A.); (V.S.); (V.V.L.); (A.N.A.)
| | - Costica Mitrofan
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.); (A.A.T.); (A.H.N.); (D.L.S.)
| | - Aye Aung Thet
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.); (A.A.T.); (A.H.N.); (D.L.S.)
| | - Alin Horatiu Nedelcu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.); (A.A.T.); (A.H.N.); (D.L.S.)
| | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.M.); (A.A.T.); (A.H.N.); (D.L.S.)
| | - Stefan Lucian Burlea
- Public Health and Management Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | | | - Vasile Valeriu Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.K.I.); (A.L.); (M.M.A.); (V.S.); (V.V.L.); (A.N.A.)
| | - Alice Nicoleta Azoicai
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.K.I.); (A.L.); (M.M.A.); (V.S.); (V.V.L.); (A.N.A.)
| |
Collapse
|
14
|
Jo Y, Greene TT, Zhang K, Chiale C, Fang Z, Dallari S, Marooki N, Wang W, Zuniga EI. Genomic Analysis of Progenitors in Viral Infection Implicates Glucocorticoids as Suppressors of Plasmacytoid Dendritic Cell Generation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620771. [PMID: 39554106 PMCID: PMC11565824 DOI: 10.1101/2024.10.28.620771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Plasmacytoid Dendritic cells (pDCs) are the most potent producers of interferons, which are critical antiviral cytokines. pDC development is, however, compromised following a viral infection, and this phenomenon, as well as its relationship to conventional (c)DC development is still incompletely understood. By using lymphocytic choriomeningitis virus (LCMV) infection in mice as a model system, we observed that DC progenitors skewed away from pDC and towards cDC development during in vivo viral infection. Subsequent characterization of the transcriptional and epigenetic landscape of fms-like tyrosine kinase 3 + (Flt3 + ) DC progenitors and follow-up studies revealed increased apoptosis and reduced proliferation in different individual DC-progenitors as well as a profound IFN-I-dependent ablation of pre-pDCs, but not pre-DC precursor, after both acute and chronic LCMV infections. In addition, integrated genomic analysis identified altered activity of 34 transcription factors in Flt3 + DC progenitors from infected mice, including two regulators of Glucocorticoid (GC) responses. Subsequent studies demonstrated that addition of GCs to DC progenitors led to downregulated pDC-primed-genes while upregulating cDC-primed-genes, and that endogenous GCs selectively decreased pDC, but not cDC, numbers upon in-vivo LCMV infection. These findings demonstrate a significant ablation of pre-pDCs in infected mice and identify GCs as suppressors of pDC generation from early progenitors. This provides an explanation for the impaired pDC development following viral infection and links pDC generation to the hypothalamic-pituitary-adrenal axis. Significance Statement Plasmacytoid dendritic cells (pDCs) play critical roles in antiviral responses. However, adaptations of DC progenitors lead to compromised pDC generation after viral infection. Here, we characterized the transcriptional and epigenetic landscapes of DC progenitors after infection. We observed widespread changes in gene expression and chromatin accessibility, reflecting shifts in proliferation, apoptosis, and differentiation potential into various DC subsets. Notably, we identified alterations in the predicted activity of 34 transcription factors, including two regulators of glucocorticoid responses. Our data demonstrate that glucocorticoids inhibit pDC generation by reprogramming DC progenitors. These findings establish a molecular framework for understanding how DC progenitors adapt to infection and highlight the role of glucocorticoid signaling in this process.
Collapse
|
15
|
Bhandari S, Kyrrestad I, Simón-Santamaría J, Li R, Szafranska KJ, Dumitriu G, Sánchez Romano J, Smedsrød B, Sørensen KK. Mouse liver sinusoidal endothelial cell responses to the glucocorticoid receptor agonist dexamethasone. Front Pharmacol 2024; 15:1377136. [PMID: 39439887 PMCID: PMC11494038 DOI: 10.3389/fphar.2024.1377136] [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: 01/26/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) which make up the fenestrated wall of the hepatic sinusoids, are active scavenger cells involved in blood waste clearance and liver immune functions. Dexamethasone is a synthetic glucocorticoid commonly used in the clinic and as cell culture supplement. However, the response is dependent on tissue, cell type, and cell state. The aim of this study was to investigate the effect of dexamethasone on primary mouse LSECs (C57BL/6J); their viability (live-dead, LDH release, caspase 3/7 assays), morphology (scanning electron microscopy), release of inflammatory markers (ELISA), and scavenging functions (endocytosis assays), and associated biological processes and pathways. We have characterized and catalogued the proteome of LSECs cultured for 1, 10, or 48 h to elucidate time-dependent and dexamethasone-specific cell responses. More than 6,000 protein IDs were quantified using tandem mass tag technology and advanced mass spectrometry (synchronous precursor selection multi-notch MS3). Enrichment analysis showed a culture-induced upregulation of stress and inflammatory markers, and a significant shift in cell metabolism already at 10 h, with enhancement of glycolysis and concomitant repression of oxidative phosphorylation. At 48 h, changes in metabolic pathways were more pronounced with dexamethasone compared to time-matched controls. Dexamethasone repressed the activation of inflammatory pathways (IFN-gamma response, TNF-alpha signaling via NF-kB, Cell adhesion molecules), and culture-induced release of interleukin-6, VCAM-1, and ICAM-1, and improved cell viability partly through inhibition of apoptosis. The mouse LSECs did not proliferate in culture. Dexamethasone treated cells showed upregulation of xanthine dehydrogenase/oxidase (Xdh), and the transcription regulator Foxo1. The drug further delayed but did not block the culture-induced loss of LSEC fenestration. The LSEC capacity for endocytosis was significantly reduced at 48 h, independent of dexamethasone, which correlated with diminished expression of several scavenger receptors and C-type lectins and altered expression of proteins in the endocytic machinery. The glucocorticoid receptor (NR3C1) was suppressed by dexamethasone at 48 h, suggesting limited effect of the drug in prolonged LSEC culture. Conclusion: The study presents a detailed overview of biological processes and pathways affected by dexamethasone in mouse LSECs in vitro.
Collapse
|
16
|
Ito CNA, dos Santos Procopio E, Balsalobre NDM, Machado LL, Silva-Filho SE, Pedroso TF, de Lourenço CC, Oliveira RJ, Arena AC, Salvador MJ, Kassuya CAL. Analgesic and Anti-Arthritic Potential of Methanolic Extract and Palmatine Obtained from Annona squamosa Leaves. Pharmaceuticals (Basel) 2024; 17:1331. [PMID: 39458972 PMCID: PMC11510468 DOI: 10.3390/ph17101331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Annona squamosa is used in folk medicine to treat pain and arthritis. Palmatine is an alkaloid isolated from several plants, including A. squamosa leaves. The aim of the present study was to investigate the analgesic, anti-arthritic, and anti-inflammatory potential of the methanolic extract of A. squamosa (EMAS) and palmatine. Methods: The chemical profile of EMAS was evaluated by ultra high-performance liquid chromatography with electrospray ionization coupled to mass spectrometry (UHPLC-ESI/MS). EMAS and palmatine were evaluated in carrageenan-induced pleurisy, zymosan-induced joint inflammation, formalin-induced nociception, and tumor necrosis factor (TNF)-induced mechanical hyperalgesia in experimental models in mice. A cytotoxicity test of EMAS and palmatine was performed using a methylthiazolidiphenyl-tetrazolium (MTT) bromide assay. Results: The analysis of the chemical profile of the extract showed the presence of palmatine, liriodenine, and anonaine. Oral administration of EMAS and palmatine significantly reduced leukocyte migration and oxide nitric production in the carrageenan-induced pleurisy model. EMAS and palmatine reduced mechanical hyperalgesia, leukocyte migration, and edema formation in the joint inflammation induced by zymosan. In the formalin test, palmatine was effective against the second-phase nociceptive response, mechanical hyperalgesia, and cold allodynia. In addition, palmatine reduced mechanical hyperalgesia induced by TNF. EMAS and palmatine did not demonstrate cytotoxicity. Conclusions: The present study showed that A. squamosa and palmatine are analgesic and anti-inflammatory agents, and that the anti-hyperalgesic properties of palmatine may involve the TNF pathway. Palmatine may be one of the compounds responsible for the anti-hyperalgesic and/or anti-arthritic properties of this medicinal plant.
Collapse
Affiliation(s)
- Caren Naomi Aguero Ito
- Health Sciences College, Federal University of Grande Dourados (UFGD), Dourados 79804-970, MS, Brazil; (C.N.A.I.); (E.d.S.P.); (N.d.M.B.)
| | - Elisangela dos Santos Procopio
- Health Sciences College, Federal University of Grande Dourados (UFGD), Dourados 79804-970, MS, Brazil; (C.N.A.I.); (E.d.S.P.); (N.d.M.B.)
| | - Natália de Matos Balsalobre
- Health Sciences College, Federal University of Grande Dourados (UFGD), Dourados 79804-970, MS, Brazil; (C.N.A.I.); (E.d.S.P.); (N.d.M.B.)
| | - Lucas Luiz Machado
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil;
| | - Saulo Euclides Silva-Filho
- Pharmaceutical Sciences, Food and Nutrition College, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil;
| | - Taíse Fonseca Pedroso
- Institute of Biology, Department of Plant Biology, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil; (T.F.P.); (C.C.d.L.); (M.J.S.)
| | - Caroline Caramano de Lourenço
- Institute of Biology, Department of Plant Biology, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil; (T.F.P.); (C.C.d.L.); (M.J.S.)
| | - Rodrigo Juliano Oliveira
- Stem Cell, Cell Therapy and Toxicological Genetics Research Centre (CeTroGen), Medical School, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, MS, Brazil;
| | - Arielle Cristina Arena
- Institute of Biosciences of Botucatu, Department of Structural and Functional Biology, São Paulo State University (UNESP), Botucatu 18618-970, SP, Brazil;
| | - Marcos José Salvador
- Institute of Biology, Department of Plant Biology, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil; (T.F.P.); (C.C.d.L.); (M.J.S.)
| | - Cândida Aparecida Leite Kassuya
- Health Sciences College, Federal University of Grande Dourados (UFGD), Dourados 79804-970, MS, Brazil; (C.N.A.I.); (E.d.S.P.); (N.d.M.B.)
| |
Collapse
|
17
|
Wang S, Ye Q. The glucocorticoid dose-mortality nexus in pneumonia patients: unveiling the threshold effect. Front Pharmacol 2024; 15:1445979. [PMID: 39364057 PMCID: PMC11447404 DOI: 10.3389/fphar.2024.1445979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024] Open
Abstract
Background The impact of glucocorticoid use on mortality risk in pneumonia patients remains unclear. This study aimed to investigate the relationship between the accumulated dose of glucocorticoids (ADG) and secondary pneumonia mortality risk among patients receiving oral or intravenous glucocorticoids. Methods Data from the DRYAD database were analyzed, covering pneumonia patients from six academic hospitals over a 5-year period who had been administered oral or intravenous glucocorticoids. Piecewise linear regression and multivariate regression analysis were utilized to assess the association between ADG and mortality risk in pneumonia patients, while adjusting for potential confounders. Results Among the 628 pneumonia patients included, the 30-day mortality rate was 23.1% and the 90-day mortality rate was 26.4%. In the high-dose glucocorticoid group (≥24 mg/day of methylprednisolone or an equivalent glucocorticoid within 30 days before admission), the 30-day and 90-day mortality rates were 31.2% and 35.9%, respectively. Piecewise linear regression analysis demonstrated a non-linear relationship between ADG and mortality risk in pneumonia patients. Multivariate regression analysis revealed a significantly lower mortality risk in patients receiving an ADG of 20-39 g methylprednisolone compared to those receiving lower (<20 g) or higher doses (≥40 g), after adjusting for potential confounding factors. Additionally, in the high-dose glucocorticoid group, surpassing the inflection point of 20 g of methylprednisolone raised the 30-day and 90-day mortality risks (adjusted odds ratio, 95% confidence interval: 1.16, 1.03-1.30 and 1.23, 1.07-1.42, respectively). Notably, this threshold effect was observed exclusively in male patients. Conclusion This study provides evidence supporting a potential threshold effect between ADG and mortality risk in oral or intravenous glucocorticoid users with secondary pneumonia. Specifically, male patients receiving high-dose glucocorticoids should undergo close monitoring when the ADG of methylprednisolone exceeds 20 g, as it may be associated with an elevated risk of mortality.
Collapse
Affiliation(s)
- Saibin Wang
- Department of Pulmonary and Critical Care Medicine, Jinhua Municipal Central Hospital, Jinhua, Zhejiang Province, China
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Qian Ye
- Department of Medical Records Quality Management, Jinhua Municipal Central Hospital, Jinhua, Zhejiang Province, China
| |
Collapse
|
18
|
Szczepanska-Sadowska E, Czarzasta K, Bogacki-Rychlik W, Kowara M. The Interaction of Vasopressin with Hormones of the Hypothalamo-Pituitary-Adrenal Axis: The Significance for Therapeutic Strategies in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2024; 25:7394. [PMID: 39000501 PMCID: PMC11242374 DOI: 10.3390/ijms25137394] [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: 05/29/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
A large body of evidence indicates that vasopressin (AVP) and steroid hormones are frequently secreted together and closely cooperate in the regulation of blood pressure, metabolism, water-electrolyte balance, and behavior, thereby securing survival and the comfort of life. Vasopressin cooperates with hormones of the hypothalamo-pituitary-adrenal axis (HPA) at several levels through regulation of the release of corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), and multiple steroid hormones, as well as through interactions with steroids in the target organs. These interactions are facilitated by positive and negative feedback between specific components of the HPA. Altogether, AVP and the HPA cooperate closely as a coordinated functional AVP-HPA system. It has been shown that cooperation between AVP and steroid hormones may be affected by cellular stress combined with hypoxia, and by metabolic, cardiovascular, and respiratory disorders; neurogenic stress; and inflammation. Growing evidence indicates that central and peripheral interactions between AVP and steroid hormones are reprogrammed in cardiovascular and metabolic diseases and that these rearrangements exert either beneficial or harmful effects. The present review highlights specific mechanisms of the interactions between AVP and steroids at cellular and systemic levels and analyses the consequences of the inappropriate cooperation of various components of the AVP-HPA system for the pathogenesis of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | | | | | | |
Collapse
|
19
|
He J, Wang Y, Hou Y, Tang F, Tian J. Rapid and sensitive detection of glucocorticoids using engineered magnetosomes functionalized protein A conjugated broad-spectrum monoclonal antibody. Food Chem X 2024; 22:101523. [PMID: 38911916 PMCID: PMC11190481 DOI: 10.1016/j.fochx.2024.101523] [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: 04/01/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/25/2024] Open
Abstract
Engineered bacterial magnetic nanoparticles (BMPs) fused with protein A (BMP-PA) can bind antibodies, creating immunomagnetic beads that offer an attractive tool for targets screening. In the study, BMP-PA-IgG was formed by attaching broad-spectrum monoclonal antibodies against glucocorticoids (GCs) to BMP-PA. Immunomagnetic assay was developed for analysis of GCs, using the BMP-PA-IgG and hydrocortisone-horseradish peroxidase. The developed assay exhibited broad specificity for GCs, including hydrocortisone (HCS), betamethasone (BMS), dexamethasone (DMS), prednisolone (PNS), beclomethasone (BCMS), cortisone (CS), 6-α-methylprednisone (6-α-MPNS), and fludrocortisone acetate (HFCS), with half inhibitory concentrations (IC50) ranging from 0.88 to 6.57 ng/mL. The proposed assay showed average recoveries of HCS and DMS ranging from 75.6% to 105.2% in chicken and pork samples, which were correlated well with those obtained by LC-MS/MS. This study indicated that the integration of engineered immunomagnetic beads into immunoassay systems offer possibilities for the sensitive and selective detection of GCs.
Collapse
Affiliation(s)
- Jinxin He
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Yuan Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Yaqing Hou
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Fang Tang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Jiesheng Tian
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
20
|
Hecker M, Fitzner B, Koczan D, Klehmet J, Grothe M, Schwab M, Winkelmann A, Meister S, Dudesek A, Ludwig-Portugall I, Eulitz K, Zettl UK. Differential gene expression in B cells and T helper cells following high-dose glucocorticoid therapy for multiple sclerosis relapse. Biomed Pharmacother 2024; 175:116721. [PMID: 38749180 DOI: 10.1016/j.biopha.2024.116721] [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: 03/05/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Despite remarkable advances in the therapy of multiple sclerosis (MS), patients with MS may still experience relapses. High-dose short-term methylprednisolone (MP) remains the standard treatment in the acute management of MS relapses due to its potent anti-inflammatory and immunosuppressive properties. However, there is a lack of studies on the cell type-specific transcriptome changes that are induced by this synthetic glucocorticoid (GC). Moreover, it is not well understood why some patients do not benefit adequately from MP therapy. METHODS We collected peripheral blood from MS patients in relapse immediately before and after ∼3-5 days of therapy with MP at 4 study centers. CD19+ B cells and CD4+ T cells were then isolated for profiling the transcriptome with high-density arrays. The patients' improvement of neurological symptoms was evaluated after ∼2 weeks by the treating physicians. We finally analyzed the data to identify genes that were differentially expressed in response to the therapy and whose expression differed between clinical responders and non-responders. RESULTS After MP treatment, a total of 33 genes in B cells and 55 genes in T helper cells were significantly up- or downregulated. The gene lists overlap in 10 genes and contain genes that have already been described as GC-responsive genes in the literature on other cell types and diseases. Their differential expression points to a rapid and coordinated modulation of multiple signaling pathways that influence transcription. Genes that were previously suggested as potential prognostic biomarkers of the clinical response to MP therapy could not be confirmed in our data. However, a greater increase in the expression of genes encoding proteins with antimicrobial activity was detected in CD4+ T cells from non-responders compared to responders. CONCLUSION Our study delved into the cell type-specific effects of MP at the transcriptional level. The data suggest a therapy-induced ectopic expression of some genes (e.g., AZU1, ELANE and MPO), especially in non-responders. The biological consequences of this remain to be explored in greater depth. A better understanding of the molecular mechanisms underlying clinical recovery from relapses in patients with MS will help to optimize future treatment decisions.
Collapse
Affiliation(s)
- Michael Hecker
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany.
| | - Brit Fitzner
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Dirk Koczan
- Institute of Immunology, Rostock University Medical Center, Rostock, Germany
| | - Juliane Klehmet
- Center for Multiple Sclerosis, Department of Neurology, Jüdisches Krankenhaus Berlin, Berlin, Germany
| | - Matthias Grothe
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Schwab
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Alexander Winkelmann
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Stefanie Meister
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Ales Dudesek
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | | | | | - Uwe Klaus Zettl
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
21
|
Light JJ, Pavlesen S, Ablove RH. Hand and Upper Extremity Surgical Site Infection Rates Associated With Perioperative Corticosteroid Injection: A Review of the Literature. Hand (N Y) 2024; 19:575-586. [PMID: 36722728 PMCID: PMC11141411 DOI: 10.1177/15589447221150501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Corticosteroid injection (CSI) has a relatively high benefit-to-risk ratio and is commonly administered to treat musculoskeletal conditions. However, perioperative CSI has been associated with an increased risk of postoperative infection. The literature suggests delaying surgery after CSI to minimize the risk of postoperative infection. We review the literature to summarize the most current knowledge on the association between perioperative CSI and infection rates for different hand and upper extremity procedures. METHODS Two independent reviewers conducted a literature search using PubMed and Web of Science databases (through October 1, 2022). The database searches used were (((injection) AND (infection)) AND (risk)) AND ((hand) OR (wrist) OR (elbow) OR (shoulder)). English-language articles were screened for infection rates associated with CSI given temporally around upper extremity surgery, focusing between 6 months preoperatively and 1 month postoperatively. RESULTS Nineteen articles including database queries and retrospective case-control or cohort studies were used after screening 465 articles. Most infection rates were increased in hand, wrist, elbow, and shoulder surgery between 3 months preoperatively and 1 month postoperatively. Intraoperative injection during elbow arthroscopy demonstrated increased infection rate relative to other upper extremity surgeries. CONCLUSIONS Corticosteroid injection increased the risk of infection temporally around upper extremity surgeries; however, CSI provides benefits. The consensus regarding CSI timeline perioperatively has yet to be determined. The evidence supports an increased benefit-to-risk ratio when giving corticosteroids greater than 3 months preoperatively and greater than 1 month postoperatively for most upper extremity procedures, with relative contraindications within 1 month of upper extremity surgery.
Collapse
Affiliation(s)
| | - Sonja Pavlesen
- University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, NY, USA
| | - Robert H. Ablove
- University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, NY, USA
| |
Collapse
|
22
|
He J, Hou Y, Wu W, Li Y, Tang F. Development of a broad-spectrum one-step immunoassay for detection of glucocorticoids in milk using magnetosome-based immunomagnetic beads. Food Chem 2024; 441:138377. [PMID: 38219367 DOI: 10.1016/j.foodchem.2024.138377] [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: 08/30/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024]
Abstract
Immunomagnetic beads provide novel tools for high-throughput immunoassay techniques. In this study, protein G (PG) was immobilized onto bacterial magentic particles (BMPs) using an additional cysteine residue at the C-terminus. A broad-spectrum monoclonal antibody against glucocorticoids (GCs) was attached to BMPs through PG-Fc interaction, generating BMP-PG-mIgG immunomagentic beads. A sensitive one-step immunoassay was developed for GCs based on combination of BMP-PG-mIgG and dexamethasone-horseradish peroxidase tracer (DMS-HRP). The developed assay exhibited half inhibitory concentrations (IC50) for dexamethasone (DMS), betamethasone (BMS), prednisolone (PNS), hydrocortisone (HCS), beclomethasone (BCMS), cortisone (CS), 6-α-methylprednisone (6-α-MPNS), fludrocortisone acetate (HFCS) of 0.98, 1.49, 2.42, 9.29, 1.63, 6.13, 7.3, and 4.89 ng/mL, respectively. The method showed recoveries ranging rates from 86.5 % to 117 % with a coefficient of variation less than 12.3 % in milk sample, which showed a good correlation with LC-MS/MS. Thus, the proposed assay offers a rapid and broad-spectrum screening tool for simultaneous detection of GCs in milk.
Collapse
Affiliation(s)
- Jinxin He
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China.
| | - Yaqing Hou
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Wanqi Wu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Yujiang Li
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| | - Fang Tang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi 030801, PR China
| |
Collapse
|
23
|
Ma W, Che J, Chen W, Wang D, Zhang H, Zhao Y. Dexamethasone-Integrated Mesenchymal Stem Cells for Systemic Lupus Erythematosus Treatment via Multiple Immunomodulatory Mechanisms. ACS NANO 2024; 18:13249-13265. [PMID: 38720584 DOI: 10.1021/acsnano.4c02420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The therapeutic application of mesenchymal stem cells (MSCs) has good potential as a treatment strategy for systemic lupus erythematosus (SLE), but traditional MSC therapy still has limitations in effectively modulating immune cells. Herein, we present a promising strategy based on dexamethasone liposome-integrated MSCs (Dexlip-MSCs) for treating SLE via multiple immunomodulatory pathways. This therapeutic strategy prolonged the circulation time of dexamethasone liposomes in vivo, restrained CD4+T-cell proliferation, and inhibited the release of proinflammatory mediators (IFN-γ and TNF-α) by CD4+T cells. In addition, Dexlip-MSCs initiated cellular reprogramming by activating the glucocorticoid receptor (GR) signaling pathway to upregulate the expression of anti-inflammatory factors such as cysteine-rich secretory protein LCCL-containing domain 2 (CRISPLD2) and downregulate the expression of proinflammatory factors. In addition, Dexlip-MSCs synergistically increased the anti-inflammatory inhibitory effect of CD4+T cells through the release of dexamethasone liposomes or Dex-integrated MSC-derived exosomes (Dex-MSC-EXOs). Based on these synergistic biological effects, we demonstrated that Dexlip-MSCs alleviated disease progression in MRL/lpr mice more effectively than Dexlip or MSCs alone. These features indicate that our stem cell delivery strategy is a promising therapeutic approach for clinical SLE treatment.
Collapse
Affiliation(s)
- Wenjuan Ma
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
- Department of Pharmacy, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Junyi Che
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Weiwei Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Huayong Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
24
|
Heming N, Renault A, Kuperminc E, Brun-Buisson C, Megarbane B, Quenot JP, Siami S, Cariou A, Forceville X, Schwebel C, Leone M, Timsit JF, Misset B, Benali MA, Colin G, Souweine B, Asehnoune K, Mercier E, Chimot L, Charpentier C, François B, Boulain T, Petitpas F, Constantin JM, Dhonneur G, Baudin F, Combes A, Bohé J, Loriferne JF, Cook F, Slama M, Leroy O, Capellier G, Dargent A, Hissem T, Bounab R, Maxime V, Moine P, Bellissant E, Annane D. Hydrocortisone plus fludrocortisone for community acquired pneumonia-related septic shock: a subgroup analysis of the APROCCHSS phase 3 randomised trial. THE LANCET. RESPIRATORY MEDICINE 2024; 12:366-374. [PMID: 38310918 DOI: 10.1016/s2213-2600(23)00430-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 10/20/2023] [Accepted: 11/08/2023] [Indexed: 02/06/2024]
Abstract
BACKGROUND Glucocorticoids probably improve outcomes in patients hospitalised for community acquired pneumonia (CAP). In this a priori planned exploratory subgroup analysis of the phase 3 randomised controlled Activated Protein C and Corticosteroids for Human Septic Shock (APROCCHSS) trial, we aimed to investigate responses to hydrocortisone plus fludrocortisone between CAP and non-CAP related septic shock. METHODS APROCCHSS was a randomised controlled trial that investigated the effects of hydrocortisone plus fludrocortisone, drotrecogin-alfa (activated), or both on mortality in septic shock in a two-by-two factorial design; after drotrecogin-alfa was withdrawn on October 2011, from the market, the trial continued on two parallel groups. It was conducted in 34 centres in France. In this subgroup study, patients with CAP were a preselected subgroup for an exploratory secondary analysis of the APROCCHSS trial of hydrocortisone plus fludrocortisone in septic shock. Adults with septic shock were randomised 1:1 to receive, in a double-blind manner, a 7-day treatment with daily administration of intravenous hydrocortisone 50 mg bolus every 6h and a tablet of 50 μg of fludrocortisone via the nasogastric tube, or their placebos. The primary outcome was 90-day all-cause mortality. Secondary outcomes included all-cause mortality at intensive care unit (ICU) and hospital discharge, 28-day and 180-day mortality, the number of days alive and free of vasopressors, mechanical ventilation, or organ failure, and ICU and hospital free-days to 90-days. Analysis was done in the intention-to-treat population. The trial was registered at ClinicalTrials.gov (NCT00625209). FINDINGS Of 1241 patients included in the APROCCHSS trial, CAP could not be ruled in or out in 31 patients, 562 had a diagnosis of CAP (279 in the placebo group and 283 in the corticosteroid group), and 648 patients did not have CAP (329 in the placebo group and 319 in the corticosteroid group). In patients with CAP, there were 109 (39%) deaths of 283 patients at day 90 with hydrocortisone plus fludrocortisone and 143 (51%) of 279 patients receiving placebo (odds ratio [OR] 0·60, 95% CI 0·43-0·83). In patients without CAP, there were 148 (46%) deaths of 319 patients at day 90 in the hydrocortisone and fludrocortisone group and 157 (48%) of 329 patients in the placebo group (OR 0·95, 95% CI 0·70-1·29). There was significant heterogeneity in corticosteroid effects on 90-day mortality across subgroups with CAP and without CAP (p=0·046 for both multiplicative and additive interaction tests; moderate credibility). Of 1241 patients included in the APROCCHSS trial, 648 (52%) had ARDS (328 in the placebo group and 320 in the corticosteroid group). There were 155 (48%) deaths of 320 patients at day 90 in the corticosteroid group and 186 (57%) of 328 patients in the placebo group. The OR for death at day 90 was 0·72 (95% CI 0·53-0·98) in patients with ARDS and 0·85 (0·61-1·20) in patients without ARDS (p=0·45 for multiplicative interaction and p=0·42 for additive interaction). The OR for observing at least one serious adverse event (corticosteroid group vs placebo) within 180 days post randomisation was 0·64 (95% CI 0·46-0·89) in the CAP subgroup and 1·02 (0·75-1·39) in the non-CAP subgroup (p=0·044 for multiplicative interaction and p=0·042 for additive interaction). INTERPRETATION In a pre-specified subgroup analysis of the APROCCHSS trial of patients with CAP and septic shock, hydrocortisone plus fludrocortisone reduced mortality as compared with placebo. Although a large proportion of patients with CAP also met criteria for ARDS, the subgroup analysis was underpowered to fully discriminate between ARDS and CAP modifying effects on mortality reduction with corticosteroids. There was no evidence of a significant treatment effect of corticosteroids in the non-CAP subgroup. FUNDING Programme Hospitalier de Recherche Clinique of the French Ministry of Health, by Programme d'Investissements d'Avenir, France 2030, and IAHU-ANR-0004.
Collapse
Affiliation(s)
- Nicholas Heming
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, Garches, France; Institut Hospitalo Universitaire PROMETHEUS, Garches, France; Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France; FHU SEPSIS, Garches, France
| | - Alain Renault
- CIC 1414 INSERM, CHU Rennes, University of Rennes, Rennes, France
| | - Emmanuelle Kuperminc
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, Garches, France
| | - Christian Brun-Buisson
- Service de Réanimation Médicale, Hôpital Henri-Mondor (Assistance Publique-Hôpitaux de Paris [AP-HP]), Créteil, France
| | - Bruno Megarbane
- Réanimation Médicale et Toxicologique, Hôpital Lariboisière (AP-HP), Université Paris-Diderot, INSERM Unité Mixte de Recherche Scientifique (UMRS) 1144, Paris, France
| | - Jean-Pierre Quenot
- Service de Réanimation Médicale, Hôpital Universitaire François Mitterrand, Lipness Team, INSERM Research Center Lipids, Nutrition, Cancer-Unité Mixte de Recherche (UMR) 1231, Dijon, France; Laboratoire d'Excellence LipSTIC and CIC 1432, Epidémiologie Clinique, Université de Burgundy, Dijon, France
| | - Shidasp Siami
- Service d'Anesthésie-Réanimation, Centre Hospitalier d'Etampes, Etampes, France
| | - Alain Cariou
- Réanimation Médicale-Hôpitaux Universitaires Paris Centre-Site Cochin (AP-HP), Paris, France
| | - Xavier Forceville
- CIC INSERM 1414, Réanimation Médico-Chirurgicale, Hôpital Saint Faron, Grand Hôpital de l'Est Francilien Site de Meaux, Meaux, France
| | - Carole Schwebel
- Service de Réanimation Médicale, CHU de Grenoble, Grenoble, France
| | - Marc Leone
- Service d'Anesthésie et de Réanimation, Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Aix Marseille Université, CIC 1409 and CIC 9502, Marseille, France
| | - Jean-Francois Timsit
- Médecine Intensive et Réanimation, Pôle 2i, Infection et Immunité, Hôpital Bichat-Claude Bernard, AP-HP, Infection, Antimicrobiens, Modélisation, Evolution (IAME) Unité 1137, Université Paris Diderot, INSERM, Paris, France
| | - Benoît Misset
- Service des Soins Intensifs, CHU Liège, Liège, Belgium
| | - Mohamed Ali Benali
- Service de Réanimation Polyvalente, Centre Hospitalier de Valenciennes, Valenciennes, France
| | - Gwenhael Colin
- Service de Réanimation Médico-Chirurgicale, Centre Hospitalier Départemental de Vendée, Site de La Roche-sur-Yon, La Roche-sur-Yon, France
| | - Bertrand Souweine
- Réanimation Médicale Polyvalente, CHU Gabriel Montpied, Clermont-Ferrand, France
| | - Karim Asehnoune
- Laboratoire EA3826 Thérapeutiques et Expérimentales des Infections, Service d'Anesthésie, Réanimation Chirurgicale, Hôtel Dieu-Hôpital Mère-Enfant, CHU Nantes, Nantes, France
| | - Emmanuelle Mercier
- Réanimation Polyvalente, Centre Hospitalier Régional Universitaire Bretonneau, Tours, France
| | - Loïc Chimot
- Service d'Anesthésie-Réanimation, Centre Hospitalier de Périgueux, Périgueux, France
| | - Claire Charpentier
- Service de Réanimation Chirurgicale, Hôpital Central, CHU de Nancy, Nancy, France
| | - Bruno François
- Service de Réanimation Polyvalente, INSERM CIC 1435, CHU Dupuytren, Limoges, France
| | - Thierry Boulain
- Service Réanimation Médicale Polyvalente et Unité de Surveillance Continue, Centre Hospitalier Régional d'Orléans, Orléans, France
| | - Frank Petitpas
- Réanimation Chirurgicale, Département d'Anesthésie-Réanimations-Urgences, Service d'Assistance Médicale d'Urgence (SAMU) 86, Hôpital de la Miletrie, CHU, Poitiers, France
| | - Jean Michel Constantin
- Department of Anaesthesiology and Critical Care, Pitié-Salpêtrière Hospital, GRC 29, DMU DREAM, AP-HP, Paris, France; Sorbonne University, Paris, France
| | - Gilles Dhonneur
- Department of Anaesthesia and Intensive Care, Curie Institute, Paris, France
| | - François Baudin
- Service d'Anesthésie et Réanimations Chirurgicales, Hôpitaux Universitaires Paris Centre-Site Cochin (AP-HP), Paris, France
| | - Alain Combes
- Service de Réanimation Médicale, Hôpital Pitié-Salpêtrière (AP-HP), Paris, France; INSERM, UMRS 1166-Institute of Cardiometabolism and Nutrition, Université Paris Sorbonne, Paris, France
| | - Julien Bohé
- Service de Réanimation Médicale, Centre Hospitalier Lyon-Sud (Hospices Civils de Lyon), Pierre-Bénite, France
| | | | - Fabrice Cook
- Service d'Anesthésie et des Réanimations Chirurgicales, Hôpital Henri-Mondor (Assistance Publique-Hôpitaux de Paris [AP-HP]), Créteil, France
| | - Michel Slama
- Service de Réanimation Médicale, CHU Amiens-Picardie-Site Sud, Amiens, France
| | - Olivier Leroy
- Service de Réanimation Médicale et Maladies Infectieuses, Centre Hospitalier Tourcoing Gustave Dron, Tourcoing, France
| | - Gilles Capellier
- Service de Réanimation Médicale-SAMU 25, Hôpital Jean Minjoz-CHU de Besançon, Besançon, France
| | - Auguste Dargent
- Service de Réanimation Médicale, Hôpital Universitaire François Mitterrand, Lipness Team, INSERM Research Center Lipids, Nutrition, Cancer-Unité Mixte de Recherche (UMR) 1231, Dijon, France; Laboratoire d'Excellence LipSTIC and CIC 1432, Epidémiologie Clinique, Université de Burgundy, Dijon, France
| | - Tarik Hissem
- Service d'Anesthésie-Réanimation, Centre Hospitalier d'Etampes, Etampes, France
| | - Rania Bounab
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, Garches, France
| | - Virginie Maxime
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, Garches, France
| | - Pierre Moine
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, Garches, France; Institut Hospitalo Universitaire PROMETHEUS, Garches, France; Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France; FHU SEPSIS, Garches, France
| | - Eric Bellissant
- CIC 1414 INSERM, CHU Rennes, University of Rennes, Rennes, France
| | - Djillali Annane
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, Garches, France; Institut Hospitalo Universitaire PROMETHEUS, Garches, France; Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France; FHU SEPSIS, Garches, France.
| |
Collapse
|
25
|
Kabrita CS, Al Bitar S, Ghanem E. The temporal expression pattern of classical MHC class I in sleep-restricted mice: Generalizations and broader implications. Brain Behav Immun Health 2024; 37:100751. [PMID: 38511151 PMCID: PMC10951454 DOI: 10.1016/j.bbih.2024.100751] [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: 12/11/2023] [Revised: 02/22/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
The intricate relationship between sleep and leukocyte trafficking has garnered intense attention, particularly their homing dynamics to secondary lymphoid organs under normal and restricted sleep (SR). Considering the scarcity of information regarding circadian rhythms in major histocompatibility class I (MHC-I) expression in SR, we designed a study that assessed the temporal expression of MHC-I in murine lymph nodes and spleen and the subsequent effects of sleep recovery. Male C57BL/6, housed in 12:12 light/dark cycle, were grouped into control (C) and SR. SR was carried for one week before lymphoid tissues were sampled at selected time points and assessed for leukocyte number and MHC-I expression. SR resulted in 21% decrease in granulocyte and 24% increase in agranulocyte numbers. In C, MHC-I expression pattern in lymph nodes was bimodal and relatively higher than splenocytes during the animal's active phase (110.2 ± 1.8 vs 81.9 ± 3.8, respectively; p = 0.002). Splenocytes; however, showed a bimodal pattern upon SR, with higher protein levels during the rest than the activity period (154.6 + 36.2 vs 99.5 + 15.9, respectively; p = 0.002), suggesting preparedness for a potential infection. Furthermore, SR caused a significant drop in MHC-I expression at the onset of rest with 57% and 30% reduction in lymph nodes and splenocytes, respectively. However, the overall protein expression collectively taken from both lymphoid tissues remained stable, emphasizing its indispensable role in immunological homeostasis. This stability coincided with the restoration of protein levels to baseline after a short sleep recovery period, resembling a reset for MHC-I antigen presentation following a week of SR. Understanding the interplay between MHC-I expression and contextual factors could enhance treatment protocols, refining the efficacy and time precision of glucocorticoid-based therapies in immune modulation.
Collapse
Affiliation(s)
- Colette S. Kabrita
- Department of Sciences, Faculty of Natural and Applied Sciences, Notre Dame University-Louaize, Zouk Mosbeh, Lebanon
| | - Samar Al Bitar
- Department of Sciences, Faculty of Natural and Applied Sciences, Notre Dame University-Louaize, Zouk Mosbeh, Lebanon
| | - Esther Ghanem
- Department of Sciences, Faculty of Natural and Applied Sciences, Notre Dame University-Louaize, Zouk Mosbeh, Lebanon
| |
Collapse
|
26
|
Abbasciano RG, Olivieri GM, Chubsey R, Gatta F, Tyson N, Easwarakumar K, Fudulu DP, Marsico R, Kofler M, Elshafie G, Lai F, Loubani M, Kendall S, Zakkar M, Murphy GJ. Prophylactic corticosteroids for cardiopulmonary bypass in adult cardiac surgery. Cochrane Database Syst Rev 2024; 3:CD005566. [PMID: 38506343 PMCID: PMC10952358 DOI: 10.1002/14651858.cd005566.pub4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
BACKGROUND Cardiac surgery triggers a strong inflammatory reaction, which carries significant clinical consequences. Corticosteroids have been suggested as a potential perioperative strategy to reduce inflammation and help prevent postoperative complications. However, the safety and effectiveness of perioperative corticosteroid use in adult cardiac surgery is uncertain. This is an update of the 2011 review with 18 studies added. OBJECTIVES Primary objective: to estimate the effects of prophylactic corticosteroid use in adults undergoing cardiac surgery with cardiopulmonary bypass on the: - co-primary endpoints of mortality, myocardial complications, and pulmonary complications; and - secondary outcomes including atrial fibrillation, infection, organ injury, known complications of steroid therapy, prolonged mechanical ventilation, prolonged postoperative stay, and cost-effectiveness. SECONDARY OBJECTIVE to explore the role of characteristics of the study cohort and specific features of the intervention in determining the treatment effects via a series of prespecified subgroup analyses. SEARCH METHODS We used standard, extensive Cochrane search methods to identify randomised studies assessing the effect of corticosteroids in adult cardiac surgery. The latest searches were performed on 14 October 2022. SELECTION CRITERIA We included randomised controlled trials in adults (over 18 years, either with a diagnosis of coronary artery disease or cardiac valve disease, or who were candidates for cardiac surgery with the use of cardiopulmonary bypass), comparing corticosteroids with no treatments. There were no restrictions with respect to length of the follow-up period. All selected studies qualified for pooling of results for one or more endpoints. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Our primary outcomes were all-cause mortality, and cardiac and pulmonary complications. Secondary outcomes were infectious complications, gastrointestinal bleeding, occurrence of new post-surgery atrial fibrillation, re-thoracotomy for bleeding, neurological complications, renal failure, inotropic support, postoperative bleeding, mechanical ventilation time, length of stays in the intensive care unit (ICU) and hospital, patient quality of life, and cost-effectiveness. We used GRADE to assess the certainty of evidence for each outcome. MAIN RESULTS This updated review includes 72 randomised trials with 17,282 participants (all 72 trials with 16,962 participants were included in data synthesis). Four trials (6%) were considered at low risk of bias in all the domains. The median age of participants included in the studies was 62.9 years. Study populations consisted mainly (89%) of low-risk, first-time coronary artery bypass grafting (CABG) or valve surgery. The use of perioperative corticosteroids may result in little to no difference in all-cause mortality (risk with corticosteroids: 25 to 36 per 1000 versus 33 per 1000 with placebo or no treatment; risk ratio (RR) 0.90, 95% confidence interval (CI) 0.75 to 1.07; 25 studies, 14,940 participants; low-certainty evidence). Corticosteroids may increase the risk of myocardial complications (68 to 86 per 1000) compared with placebo or no treatment (66 per 1000; RR 1.16, 95% CI 1.04 to 1.31; 25 studies, 14,766 participants; low-certainty evidence), and may reduce the risk of pulmonary complications (risk with corticosteroids: 61 to 77 per 1000 versus 78 per 1000 with placebo/no treatment; RR 0.88, 0.78 to 0.99; 18 studies, 13,549 participants; low-certainty evidence). Analyses of secondary endpoints showed that corticosteroids may reduce the incidence of infectious complications (risk with corticosteroids: 94 to 113 per 1000 versus 123 per 1000 with placebo/no treatment; RR 0.84, 95% CI 0.76 to 0.92; 28 studies, 14,771 participants; low-certainty evidence). Corticosteroids may result in little to no difference in incidence of gastrointestinal bleeding (risk with corticosteroids: 9 to 17 per 1000 versus 10 per 1000 with placebo/no treatment; RR 1.21, 95% CI 0.87 to 1.67; 6 studies, 12,533 participants; low-certainty evidence) and renal failure (risk with corticosteroids: 23 to 35 per 1000 versus 34 per 1000 with placebo/no treatment; RR 0.84, 95% CI 0.69 to 1.02; 13 studies, 12,799; low-certainty evidence). Corticosteroids may reduce the length of hospital stay, but the evidence is very uncertain (-0.5 days, 0.97 to 0.04 fewer days of length of hospital stay compared with placebo/no treatment; 25 studies, 1841 participants; very low-certainty evidence). The results from the two largest trials included in the review possibly skew the overall findings from the meta-analysis. AUTHORS' CONCLUSIONS A systematic review of trials evaluating the organ protective effects of corticosteroids in cardiac surgery demonstrated little or no treatment effect on mortality, gastrointestinal bleeding, and renal failure. There were opposing treatment effects on cardiac and pulmonary complications, with evidence that corticosteroids may increase cardiac complications but reduce pulmonary complications; however, the level of certainty for these estimates was low. There were minor benefits from corticosteroid therapy for infectious complications, but the evidence on hospital length of stay was very uncertain. The inconsistent treatment effects across different outcomes and the limited data on high-risk groups reduced the applicability of the findings. Further research should explore the role of these drugs in specific, vulnerable cohorts.
Collapse
Affiliation(s)
| | | | - Rachel Chubsey
- University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Francesca Gatta
- Department of Cardiothoracic Surgery, Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Nathan Tyson
- Department of Cardiac Surgery, University Hospitals of Leicester, Leicester, UK
| | | | - Daniel P Fudulu
- Department of Cardiac Surgery, University Hospital Bristol NHS Trust, Bristol, UK
| | | | | | - Ghazi Elshafie
- Department of Cardiothoracic Surgery, Hull and East Yorkshire Hospitals NHS Trust, Hull, UK
| | - Florence Lai
- Leicester Clinical Trials Unit, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Mahmoud Loubani
- Department of Cardiothoracic Surgery, Hull and East Yorkshire Hospitals NHS Trust, Hull, UK
| | | | - Mustafa Zakkar
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Gavin J Murphy
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
27
|
Athanasiou N, Diamantopoulos A, Keskinidou C, Katsaounou P, Angelousi A, Jahaj E, Mourelatos P, Vrettou CS, Botoula E, Vassiliou AG, Kotanidou A, Tsagarakis S, Dimopoulou I, Vassiliadi DA. Adrenal function in relation to cytokines and outcome in non-critically ill patients with COVID-19. J Endocrinol Invest 2024; 47:721-728. [PMID: 37702927 DOI: 10.1007/s40618-023-02189-y] [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: 06/06/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023]
Abstract
PURPOSE We aimed to identify whether hypothalamic-pituitary-adrenal (HPA) axis dysfunction is related to deterioration in a percentage of patients who progress to severe COVID-19. METHODS In this cohort observational study, we evaluated HPA axis activation by measuring cortisol, adrenocorticotropic hormone (ACTH), dehydroepiandrosterone sulfate (DHEA-S) levels, whole blood expression levels of the key glucocorticoid receptor, GCR-α, and the glucocorticoid-induced leucine zipper (GILZ), and cytokines, as markers of the inflammatory phase, in 149 patients with respiratory infection admitted in the ward, without known adrenal disease and/or confounding medications (glucocorticoids). One hundred and four (104) patients were SARS-CoV-2 positive (C +) and controls consisted of 45 SARS-CoV-2-negative patients (NC). RESULTS No differences in cortisol levels were observed between the C + and the NC patients. Cortisol levels correlated with ACTH (r = 0.284, p = 0.001) and IL-6 (r = 0.289, p = 0.04). In C + patients, cortisol levels mainly correlated with IL-6 levels (r = 0.28; p = 0.017). GCR-α expression was significantly higher in C + patients compared to NC. Patients with higher cortisol levels were more likely to progress to respiratory function deterioration or die. Both GCR-α and GILZ expression were significantly higher in C + non-survivors. CONCLUSION Our findings indicate that cortisol serves as an indicator of disease severity. GILZ expression appears to be a more effective marker of mortality prediction in moderate COVID-19 cases. However, routine measurement of GILZ levels is currently unavailable. Elevated levels of cortisol may be indicative of patients with moderate COVID-19 who are at a higher risk of deterioration. This information can aid in identifying individuals who require early medical attention.
Collapse
Affiliation(s)
- N Athanasiou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
| | - A Diamantopoulos
- Department of Endocrinology Diabetes and Metabolism, National and European Expertise Center for Rare Endocrine Diseases, Evangelismos Hospital, Athens, Greece
| | - C Keskinidou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
| | - P Katsaounou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
| | - A Angelousi
- First Department of Internal Medicine, Unit of Endocrinology, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - E Jahaj
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
| | - P Mourelatos
- Department of Endocrinology Diabetes and Metabolism, National and European Expertise Center for Rare Endocrine Diseases, Evangelismos Hospital, Athens, Greece
| | - C S Vrettou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
| | - E Botoula
- Department of Endocrinology Diabetes and Metabolism, National and European Expertise Center for Rare Endocrine Diseases, Evangelismos Hospital, Athens, Greece
| | - A G Vassiliou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
| | - A Kotanidou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
| | - S Tsagarakis
- Department of Endocrinology Diabetes and Metabolism, National and European Expertise Center for Rare Endocrine Diseases, Evangelismos Hospital, Athens, Greece
| | - I Dimopoulou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens, Greece
| | - D A Vassiliadi
- Department of Endocrinology Diabetes and Metabolism, National and European Expertise Center for Rare Endocrine Diseases, Evangelismos Hospital, Athens, Greece.
| |
Collapse
|
28
|
Zhang Y, Du C, Wang W, Qiao W, Li Y, Zhang Y, Sheng S, Zhou X, Zhang L, Fan H, Yu Y, Chen Y, Liao Y, Chen S, Chang Y. Glucocorticoids increase adiposity by stimulating Krüppel-like factor 9 expression in macrophages. Nat Commun 2024; 15:1190. [PMID: 38331933 PMCID: PMC10853261 DOI: 10.1038/s41467-024-45477-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
The mechanisms underlying glucocorticoid (GC)-induced obesity are poorly understood. Macrophages are the primary targets by which GCs exert pharmacological effects and perform critical functions in adipose tissue homeostasis. Here, we show that macrophages are essential for GC-induced obesity. Dexamethasone (Dex) strongly induced Krüppel-like factor 9 (Klf9) expression in macrophages. Similar to Dex, lentivirus-mediated Klf9 overexpression inhibits M1 and M2a markers expression, causing macrophage deactivation. Furthermore, the myeloid-specific Klf9 transgene promotes obesity. Conversely, myeloid-specific Klf9-knockout (mKlf9KO) mice are lean. Moreover, myeloid Klf9 knockout largely blocks obesity induced by chronic GC treatment. Mechanistically, GC-inducible KLF9 recruits the SIN3A/HDAC complex to the promoter regions of Il6, Ptgs2, Il10, Arg1, and Chil3 to inhibit their expression, subsequently reducing thermogenesis and increasing lipid accumulation by inhibiting STAT3 signaling in adipocytes. Thus, KLF9 in macrophages integrates the beneficial anti-inflammatory and adverse metabolic effects of GCs and represents a potential target for therapeutic interventions.
Collapse
Affiliation(s)
- Yinliang Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Chunyuan Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Wei Wang
- Key Laboratory of Biotechnology of Hubei Province, Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei University, Wuhan, China
| | - Wei Qiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Yuhui Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Yujie Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Sufang Sheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Xuenan Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Lei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China
| | - Heng Fan
- Ningxia Key Laboratory of Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, Ningxia, China
| | - Ying Yu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yong Chen
- Key Laboratory of Biotechnology of Hubei Province, Key Laboratory of Biotechnology of Chinese Traditional Medicine, National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, Hubei University, Wuhan, China
| | - Yunfei Liao
- Department of Endocrinology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shihong Chen
- Department of Endocrinology, The Second Hospital of Shandong University, Jinan, China.
| | - Yongsheng Chang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Cellular Homeostasis and Disease, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
29
|
Duan S, Li Q, Wang F, Kuang W, Dong Y, Liu D, Wang J, Li W, Chen Q, Zeng X, Li T. Single-Cell Transcriptomes and Immune Repertoires Reveal the Cell State and Molecular Changes in Pemphigus Vulgaris. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:375-388. [PMID: 38117802 DOI: 10.4049/jimmunol.2300312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 10/05/2023] [Indexed: 12/22/2023]
Abstract
The etiology and pathogenesis of pemphigus vulgaris (PV) entail intricate interactions between immune cells and epithelial cells. However, the specific subtypes of immune cells involved in PV, along with their respective roles, remain elusive. Likewise, the precise functions and mechanisms by which glucocorticoids affect cell types within the disease context require further elucidation. To address these knowledge gaps, we performed 5' single-cell RNA sequencing, combined with V(D)J enrichment on buccal mucosal lesions and peripheral blood samples from treatment-naive patients with PV, in conjunction with post-treatment peripheral blood samples obtained after oral prednisone treatment. Our findings suggest that the IL-1α signaling pathway, myeloid APCs, inflammatory CD8+ resident memory T cells, and dysfunctional CD4+ regulatory T cells are involved in the pathogenesis of PV. Part of these findings were validated by immunohistochemical assays and multiplex immunofluorescence assays. Furthermore, our results highlight the significant impact of prednisone treatment on monocytes and mucosal-associated invariant T cells while revealing a limited effect on CD4+ regulatory T cells. Additionally, we present the CDR3 amino acid sequence of BCR related to PV disease and investigate the characteristics of TCR/BCR clonotypes. In conclusion, our study provides a comprehensive understanding of PV, particularly focusing on the mucosal-dominant type, and sheds light on the effects of glucocorticoids within the PV context. These insights hold promise for the development of new therapeutic strategies in this autoimmune disorder.
Collapse
Affiliation(s)
- Shumin Duan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Qionghua Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Fei Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Wenjing Kuang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yunmei Dong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Dan Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jiongke Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Wei Li
- Department of Dermatology, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Taiwen Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
30
|
Zhao X, Hu C, Chen X, Ren S, Gao F. Drug Repositioning of Inflammatory Bowel Disease Based on Co-Target Gene Expression Signature of Glucocorticoid Receptor and TET2. BIOLOGY 2024; 13:82. [PMID: 38392301 PMCID: PMC10886832 DOI: 10.3390/biology13020082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/16/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024]
Abstract
The glucocorticoid receptor (GR) and ten-eleven translocation 2 (TET2), respectively, play a crucial role in regulating immunity and inflammation, and GR interacts with TET2. However, their synergetic roles in inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), remain unclear. This study aimed to investigate the co-target gene signatures of GR and TET2 in IBD and provide potential therapeutic interventions for IBD. By integrating public data, we identified 179 GR- and TET2-targeted differentially expressed genes (DEGs) in CD and 401 in UC. These genes were found to be closely associated with immunometabolism, inflammatory responses, and cell stress pathways. In vitro inflammatory cellular models were constructed using LPS-treated HT29 and HCT116 cells, respectively. Drug repositioning based on the co-target gene signatures of GR and TET2 derived from transcriptomic data of UC, CD, and the in vitro model was performed using the Connectivity Map (CMap). BMS-536924 emerged as a top therapeutic candidate, and its validation experiment within the in vitro inflammatory model confirmed its efficacy in mitigating the LPS-induced inflammatory response. This study sheds light on the pathogenesis of IBD from a new perspective and may accelerate the development of novel therapeutic agents for inflammatory diseases including IBD.
Collapse
Affiliation(s)
- Xianglin Zhao
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
- School of Life Sciences, Henan University, Kaifeng 475004, China
- Shenzhen Research Institute of Henan University, Henan University, Shenzhen 518000, China
| | - Chenghao Hu
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Xinyu Chen
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Shuqiang Ren
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Fei Gao
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou 310022, China
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
31
|
Ono E, Lenief V, Lefevre MA, Cuzin R, Guironnet-Paquet A, Mosnier A, Nosbaum A, Nicolas JF, Vocanson M. Topical corticosteroids inhibit allergic skin inflammation but are ineffective in impeding the formation and expansion of resident memory T cells. Allergy 2024; 79:52-64. [PMID: 37539746 DOI: 10.1111/all.15819] [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: 11/28/2022] [Revised: 04/28/2023] [Accepted: 05/23/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND Tissue-resident memory T (TRM ) cells are detrimental in allergic contact dermatitis (ACD), in which they contribute to the chronicity and severity of the disease. METHODS We assessed the impact of a standard topical corticosteroid (TCS) treatment, triamcinolone acetonide (TA), on the formation, maintenance and reactivation of epidermal TRM cells in a preclinical model of ACD to 2,4-dinitrofluorobenzene. TA 0.01% was applied at different time points of ACD response and we monitored skin inflammation and tracked CD8+ CD69+ CD103+ TRM by flow cytometry and RNA sequencing. RESULTS The impact of TA on TRM formation depended on treatment regimen: (i) in a preventive mode, that is, in sensitized mice before challenge, TA transiently inhibited the infiltration of effector T cells and the accumulation of TRM upon hapten challenge. In contrast, (ii) in a curative mode, that is, at the peak of the ACD response, TA blocked skin inflammation but failed to prevent the formation of TRM . Finally, (iii) in a proactive mode, that is, on previous eczema lesions, TA had no effect on the survival of skin TRM , but transiently inhibited their reactivation program upon allergen reexposure. Indeed, specific TRM progressively regained proliferative functions upon TA discontinuation and expanded in the tissue, leading to exaggerated iterative responses. Interestingly, TRM re-expansion correlated with the decreased clearance of hapten moieties from the skin induced by repeated TA applications. CONCLUSIONS Our results demonstrate that TCS successfully treat ACD inflammation, but are mostly ineffective in impeding the formation and expansion of allergen-specific TRM , which certainly restricts the induction of lasting tolerance in patients with chronic dermatitis.
Collapse
Affiliation(s)
- Emi Ono
- CIRI-Centre International de Recherche en Infectiologie, INSERM, U1111, Université Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, CNRS UMR 5308, Lyon, France
| | - Vanina Lenief
- CIRI-Centre International de Recherche en Infectiologie, INSERM, U1111, Université Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, CNRS UMR 5308, Lyon, France
| | - Marine-Alexia Lefevre
- CIRI-Centre International de Recherche en Infectiologie, INSERM, U1111, Université Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, CNRS UMR 5308, Lyon, France
| | - Roxane Cuzin
- CIRI-Centre International de Recherche en Infectiologie, INSERM, U1111, Université Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, CNRS UMR 5308, Lyon, France
| | - Aurélie Guironnet-Paquet
- CIRI-Centre International de Recherche en Infectiologie, INSERM, U1111, Université Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, CNRS UMR 5308, Lyon, France
- Etablissement Français du Sang (EFS) Auvergne Rhône-Alpes, Apheresis Unit, Hôpital Lyon Sud, Pierre Bénite, France
| | - Amandine Mosnier
- CIRI-Centre International de Recherche en Infectiologie, INSERM, U1111, Université Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, CNRS UMR 5308, Lyon, France
| | - Audrey Nosbaum
- CIRI-Centre International de Recherche en Infectiologie, INSERM, U1111, Université Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, CNRS UMR 5308, Lyon, France
- Allergology and Clinical Immunology Department, Lyon Sud University Hospital, Pierre Bénite, France
| | - Jean-Francois Nicolas
- CIRI-Centre International de Recherche en Infectiologie, INSERM, U1111, Université Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, CNRS UMR 5308, Lyon, France
- Allergology and Clinical Immunology Department, Lyon Sud University Hospital, Pierre Bénite, France
| | - Marc Vocanson
- CIRI-Centre International de Recherche en Infectiologie, INSERM, U1111, Université Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, CNRS UMR 5308, Lyon, France
| |
Collapse
|
32
|
Chan KL, Poller WC, Swirski FK, Russo SJ. Central regulation of stress-evoked peripheral immune responses. Nat Rev Neurosci 2023; 24:591-604. [PMID: 37626176 PMCID: PMC10848316 DOI: 10.1038/s41583-023-00729-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2023] [Indexed: 08/27/2023]
Abstract
Stress-linked psychiatric disorders, including anxiety and major depressive disorder, are associated with systemic inflammation. Recent studies have reported stress-induced alterations in haematopoiesis that result in monocytosis, neutrophilia, lymphocytopenia and, consequently, in the upregulation of pro-inflammatory processes in immunologically relevant peripheral tissues. There is now evidence that this peripheral inflammation contributes to the development of psychiatric symptoms as well as to common co-morbidities of psychiatric disorders such as metabolic syndrome and immunosuppression. Here, we review the specific brain and spinal regions, and the neuronal populations within them, that respond to stress and transmit signals to peripheral tissues via the autonomic nervous system or neuroendocrine pathways to influence immunological function. We comprehensively summarize studies that have employed retrograde tracing to define neurocircuits linking the brain to the bone marrow, spleen, gut, adipose tissue and liver. Moreover, we highlight studies that have used chemogenetic or optogenetic manipulation or intracerebroventricular administration of peptide hormones to control somatic immune responses. Collectively, this growing body of literature illustrates potential mechanisms through which stress signals are conveyed from the CNS to immune cells to regulate stress-relevant behaviours and comorbid pathophysiology.
Collapse
Affiliation(s)
- Kenny L Chan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Wolfram C Poller
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Filip K Swirski
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
33
|
Adam L, Stanifer M, Springer F, Mathony J, Brune M, Di Ponzio C, Eils R, Boulant S, Niopek D, Kallenberger SM. Transcriptomics-inferred dynamics of SARS-CoV-2 interactions with host epithelial cells. Sci Signal 2023; 16:eabl8266. [PMID: 37751479 DOI: 10.1126/scisignal.abl8266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
Virus-host interactions can reveal potentially effective and selective therapeutic targets for treating infection. Here, we performed an integrated analysis of the dynamics of virus replication and the host cell transcriptional response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection using human Caco-2 colon cancer cells as a model. Time-resolved RNA sequencing revealed that, upon infection, cells immediately transcriptionally activated genes associated with inflammatory pathways that mediate the antiviral response, which was followed by an increase in the expression of genes involved in ribosome and mitochondria function, thus suggesting rapid alterations in protein production and cellular energy supply. At later stages, between 24 and 48 hours after infection, the expression of genes involved in metabolic processes-in particular, those related to xenobiotic metabolism-was decreased. Mathematical modeling incorporating SARS-CoV-2 replication suggested that SARS-CoV-2 proteins inhibited the host antiviral response and that virus transcripts exceeded the translation capacity of the host cells. Targeting kinase-dependent pathways that exhibited increases in transcription in host cells was as effective as a virus-targeted inhibitor at repressing viral replication. Our findings in this model system delineate a sequence of SARS-CoV-2 virus-host interactions that may facilitate the identification of druggable host pathways to suppress infection.
Collapse
Affiliation(s)
- Lukas Adam
- Health Data Science Unit, University Hospital Heidelberg and Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), University of Heidelberg, Heidelberg 69120, Germany
| | - Megan Stanifer
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg 69120, Germany
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
| | - Fabian Springer
- Health Data Science Unit, University Hospital Heidelberg and Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), University of Heidelberg, Heidelberg 69120, Germany
| | - Jan Mathony
- Department of Biology, Technical University of Darmstadt, Darmstadt 64287, Germany
- Center for Synthetic Biology, Technical University of Darmstadt, Darmstadt 64287, Germany
- BZH Graduate School, Heidelberg University, Heidelberg 69120, Germany
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Faculty of Engineering Sciences, Heidelberg University, Heidelberg 69120, Germany
| | - Maik Brune
- Clinic of Endocrinology, Diabetology, Metabolism, and Clinical Chemistry, Central Laboratory, Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Chiara Di Ponzio
- Health Data Science Unit, University Hospital Heidelberg and Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), University of Heidelberg, Heidelberg 69120, Germany
- Digital Health Center, Berlin Institute of Health (BIH) and Charité, Berlin 10178, Germany
| | - Roland Eils
- Health Data Science Unit, University Hospital Heidelberg and Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), University of Heidelberg, Heidelberg 69120, Germany
- Digital Health Center, Berlin Institute of Health (BIH) and Charité, Berlin 10178, Germany
| | - Steeve Boulant
- Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg 69120, Germany
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL 32603, USA
- Research Group "Cellular polarity and viral infection" (F140), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Dominik Niopek
- Department of Biology, Technical University of Darmstadt, Darmstadt 64287, Germany
- Center for Synthetic Biology, Technical University of Darmstadt, Darmstadt 64287, Germany
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Faculty of Engineering Sciences, Heidelberg University, Heidelberg 69120, Germany
| | - Stefan M Kallenberger
- Health Data Science Unit, University Hospital Heidelberg and Center for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), University of Heidelberg, Heidelberg 69120, Germany
- Division of Applied Bioinformatics (G200), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- National Center for Tumor Diseases, Department of Medical Oncology, Heidelberg University Hospital, Heidelberg 69120, Germany
| |
Collapse
|
34
|
Amatya S, Tietje-Mckinney D, Mueller S, Petrillo MG, Woolard MD, Bharrhan S, Orr AW, Kevil CG, Cidlowski JA, Cruz-Topete D. Adipocyte Glucocorticoid Receptor Inhibits Immune Regulatory Genes to Maintain Immune Cell Homeostasis in Adipose Tissue. Endocrinology 2023; 164:bqad143. [PMID: 37738419 PMCID: PMC10558062 DOI: 10.1210/endocr/bqad143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023]
Abstract
Glucocorticoids acting via the glucocorticoid receptors (GR) are key regulators of metabolism and the stress response. However, uncontrolled or excessive GR signaling adversely affects adipose tissue, including endocrine, immune, and metabolic functions. Inflammation of the adipose tissue promotes systemic metabolic dysfunction; however, the molecular mechanisms underlying the role of adipocyte GR in regulating genes associated with adipose tissue inflammation are poorly understood. We performed in vivo studies using adipocyte-specific GR knockout mice in conjunction with in vitro studies to understand the contribution of adipocyte GR in regulating adipose tissue immune homeostasis. Our findings show that adipocyte-specific GR signaling regulates adipokines at both mRNA and plasma levels and immune regulatory (Coch, Pdcd1, Cemip, and Cxcr2) mRNA gene expression, which affects myeloid immune cell presence in white adipose tissue. We found that, in adipocytes, GR directly influences Cxcr2. This chemokine receptor promotes immune cell migration, indirectly affecting Pdcd1 and Cemip gene expression in nonadipocyte or stromal cells. Our findings suggest that GR adipocyte signaling suppresses inflammatory signals, maintaining immune homeostasis. We also found that GR signaling in adipose tissue in response to stress is sexually dimorphic. Understanding the molecular relationship between GR signaling and adipose tissue inflammation could help develop potential targets to improve local and systemic inflammation, insulin sensitivity, and metabolic health.
Collapse
Affiliation(s)
- Shripa Amatya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
| | - Dylan Tietje-Mckinney
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
| | - Schaefer Mueller
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
| | - Maria G Petrillo
- Department of Health and Human Services, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Matthew D Woolard
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
| | - Sushma Bharrhan
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
| | - Anthony Wayne Orr
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
| | - Christopher G Kevil
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
| | - John A Cidlowski
- Department of Health and Human Services, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Diana Cruz-Topete
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, Louisiana State University Health Sciences Center—Shreveport, Shreveport, LA 71103, USA
| |
Collapse
|
35
|
Swatler J, Ju YJ, Anderson AC, Lugli E. Tumors recycle glucocorticoids to drive Treg-mediated immunosuppression. J Clin Invest 2023; 133:e173141. [PMID: 37712416 PMCID: PMC10503790 DOI: 10.1172/jci173141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023] Open
Abstract
Suppression of antitumor immunity is a prominent feature of the tumor microenvironment. In this issue of the JCI, Taves, Otsuka, and authors show that glucocorticoids (GCs), which are potent immunosuppressive hormones mainly produced by the adrenals, can be reconverted from their inactive form to active metabolites via the 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) enzyme expressed by murine tumor cell lines. In the tumor microenvironment, GCs acted on CD4+ regulatory T cells to enhance their immunosuppressive function and promote tumor growth. The findings suggest that targeting GC recycling as a strategy for modulating tumor immunosuppression has the potential to improve therapeutic efficacy of immune checkpoint blockade.
Collapse
Affiliation(s)
- Julian Swatler
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan
| | - Young-Jun Ju
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ana C. Anderson
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Enrico Lugli
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan
| |
Collapse
|
36
|
Zare F, Solhjoo A, Sadeghpour H, Sakhteman A, Dehshahri A. Structure-based virtual screening, molecular docking, molecular dynamics simulation and MM/PBSA calculations towards identification of steroidal and non-steroidal selective glucocorticoid receptor modulators. J Biomol Struct Dyn 2023; 41:7640-7650. [PMID: 36134594 DOI: 10.1080/07391102.2022.2123392] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/05/2022] [Indexed: 10/14/2022]
Abstract
Glucocorticoids have been used in the treatment of many diseases including inflammatory and autoimmune diseases. Despite the wide therapeutic effects of synthetic glucocorticoids, the use of these compounds has been limited due to side effects such as osteoporosis, immunodeficiency, and hyperglycaemia. To this end, extensive studies have been performed to discover new glucocorticoid modulators with the aim of increasing affinity for the receptor and thus less side effects. In the present work, structure-based virtual screening was used for the identification of novel potent compounds with glucocorticoid effects. The molecules derived from ZINC database were screened on account of structural similarity with some glucocorticoid agonists as the template. Subsequently, molecular docking was performed on 200 selected compounds to obtain the best steroidal and non-steroidal conformations. Three compounds, namely ZINC_000002083318, ZINC_000253697499 and ZINC_000003845653, were selected with the binding energies of -11.5, -10.5, and -9.5 kcal/mol, respectively. Molecular dynamic simulations on superior structures were accomplished with the glucocorticoid receptor. Additionally, root mean square deviations, root mean square fluctuation, radius of gyration, hydrogen bonds, and binding-free energy analysis showed the binding stability of the proposed compounds compared to budesonide as an approved drug. The results demonstrated that all the compounds had suitable binding stability compared to budesonide, while ZINC_000002083318 showed a tighter binding energy compared to the other compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fateme Zare
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Solhjoo
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Sadeghpour
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Sakhteman
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Dehshahri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
37
|
Liu X, Xie Y, Gao W, Zhan L, Hu L, Zuo L, Li Y. Experimental study of dexamethasone-loaded hollow hydroxyapatite microspheres applied to direct pulp capping of rat molars. Front Endocrinol (Lausanne) 2023; 14:1192420. [PMID: 37600685 PMCID: PMC10435764 DOI: 10.3389/fendo.2023.1192420] [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: 03/23/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Background Dexamethasone (DEX) exerts anti-inflammatory and osteogenic effects. Hydroxyapatite is commonly used in bone repair due to its osteoconductivity, osseointegration, and osteogenesis induction. Hollow hydroxyapatite (HHAM) is often used as a drug carrier. Objective This study aimed to investigate the histological responses of exposed dental pulp when dexamethasone-loaded nanohydroxyapatite microspheres (DHHAM) were used as a direct capping agent. Methods Cavities were created in the left maxillary first molar of Wistar rats and filled with Dycal, HHAM, and DHHAM. No drug was administered to the control group. The rats were sacrificed at 1, 2, and 4 weeks after the procedure. The molars were extracted for fixation, demineralization, dehydration, embedding, and sectioning. H&E staining was performed to detect the formation of reparative dentin. H&E and CD45 immunohistochemical staining were performed to detect pulp inflammation. Immunohistochemical staining was performed to assess the expressions of dentin matrix protein 1 (DMP-1), interleukin (IL)-6, tumor necrosis factor (TNF)-α, and IL-1β. Results The results of H&E and CD45 immunohistochemical staining showed that the degree of inflammation in the DHHAM group was less than that in the Control and HHAM groups at 1, 2, and 4 weeks after capping of the rat molar teeth (p<0.01). The H&E staining showed that the percentage of reparative dentin formed in the DHHAM group was higher than that in the Control, HHAM (p<0.001), and Dycal groups (p<0.01) at 1 and 2 weeks, and was significantly higher than that in the Control group (p<0.001) and the HHAM group (p<0.01) at 4 weeks. The immunohistochemical staining showed a lower range and intensity of expression of IL-1β, IL-6, and TNF-α and high expression levels of DMP-1 in the DHHAM group at 1, 2, and 4 weeks after pulp capping relative to the Control group. Conclusions DHHAM significantly inhibited the progression of inflammation and promoted reparative dentin formation.
Collapse
Affiliation(s)
- Xiaoli Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Yuandong Xie
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Weijia Gao
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Luoning Zhan
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Ling Hu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Linjing Zuo
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Yi Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| |
Collapse
|
38
|
Mazur O, Bondar M. Studying the effectiveness of varieties of anticoagulant therapy programs in patients with a severe course of COVID-19 (analysis of own clinical observations). EMERGENCY MEDICINE 2023; 19:162-168. [DOI: 10.22141/2224-0586.19.3.2023.1576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Background. Currently, there is no internationally approved protocol for anticoagulant therapy (ACT) in patients with COVID-19 that describes the different types of ACT programs depending on the severity of COVID-19. The objective: to determine optimal ACT programs. Materials and methods. A retrospective analysis of ACT programs was conducted in 268 patients with extremely severe COVID-19 who were treated in the intensive care units. Results. The article presents the results of retrospective analysis of ACT programs in 268 patients with a very severe course of COVID-19 treated in the general intensive care unit in 2021. ACT programs in 162 patients who died because of COVID-19 and in 106 survived patients were studied separately. The total duration of hospital stay was from 9 to 55 days. ACT programs were mostly in line with European guidelines/instructions. ACT included a stepwise combination of unfractionated heparin, low-molecular-weight heparins, and oral anticoagulants depending on the clinical course, concomitant pathology, and laboratory parameters. The systemic inflammatory response was characterized by the concentration of C-reactive protein in the blood plasma within the range of 54.7–331.4 mg/l (on average 95.4 ± 73.4 mg/ml), ferritin of 208–2094 μg/l (on average 947.16 ± 872.00 μg/l), interleukin-6 of 33.9–507.4 pg/ml (on average 152.60 ± 201.15 pg/ml). The level of D-dimer in blood plasma ranged from 3.74 to 82.0 μg/ml (on average 8.200 ± 3.525 μg/ml). Platelet content in peripheral blood ranged from 372 × 109/l to 72 × 109/l. Conclusions. The optimal ACT program in patients with severe and extremely severe COVID-19 is a program of sequential change of anticoagulant drugs to enhance their anticoagulant effect, which can be controlled by laboratory tests.
Collapse
|
39
|
Lee ZY, Tran T. Genomic and non-genomic effects of glucocorticoids in respiratory diseases. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 98:1-30. [PMID: 37524484 DOI: 10.1016/bs.apha.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Cortisol is an endogenous steroid hormone essential for the natural resolution of inflammation. Synthetic glucocorticoids (GCs) were developed and are currently amongst the most widely prescribed anti-inflammatory drugs in our modern clinical landscape owing to their potent anti-inflammatory activity. However, the extent of GC's effects has yet to be fully elucidated. Indeed, GCs modulate a broad spectrum of cellular activity, from their classical regulation of gene expression to acute non-genomic mechanisms of action. Furthermore, tissue specific effects, disease specific conditions, and dose-dependent responses complicate their use, with side-effects potentially plaguing their use. It is thus vital to outline and consolidate the effects of GCs, to demystify and maximize their therapeutic potential while avoiding pitfalls that would otherwise render them obsolete.
Collapse
Affiliation(s)
- Zhao-Yong Lee
- Infectious Disease Translational Research Program, National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Thai Tran
- Infectious Disease Translational Research Program, National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
40
|
Bain CR, Myles PS, Corcoran T, Dieleman JM. Postoperative systemic inflammatory dysregulation and corticosteroids: a narrative review. Anaesthesia 2023; 78:356-370. [PMID: 36308338 PMCID: PMC10092416 DOI: 10.1111/anae.15896] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2022] [Indexed: 12/15/2022]
Abstract
In some patients, the inflammatory-immune response to surgical injury progresses to a harmful, dysregulated state. We posit that postoperative systemic inflammatory dysregulation forms part of a pathophysiological response to surgical injury that places patients at increased risk of complications and subsequently prolongs hospital stay. In this narrative review, we have outlined the evolution, measurement and prediction of postoperative systemic inflammatory dysregulation, distinguishing it from a healthy and self-limiting host response. We reviewed the actions of glucocorticoids and the potential for heterogeneous responses to peri-operative corticosteroid supplementation. We have then appraised the evidence highlighting the safety of corticosteroid supplementation, and the potential benefits of high/repeated doses to reduce the risks of major complications and death. Finally, we addressed how clinical trials in the future should target patients at higher risk of peri-operative inflammatory complications, whereby corticosteroid regimes should be tailored to modify not only the a priori risk, but also further adjusted in response to markers of an evolving pathophysiological response.
Collapse
Affiliation(s)
- C R Bain
- Department of Anaesthesiology and Peri-operative Medicine, Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - P S Myles
- Department of Anaesthesiology and Peri-operative Medicine, Alfred Hospital and Monash University, Melbourne, VIC, Australia
| | - T Corcoran
- Department of Anaesthesia and Pain Medicine, Royal Perth Hospital, Perth, WA, Australia
| | - J M Dieleman
- Department of Anaesthesia and Peri-operative Medicine, Westmead Hospital, Sydney and Western Sydney University, Sydney, NSW, Australia
| |
Collapse
|
41
|
Zhang Q, Sun W, Li T, Liu F. Polarization Behavior of Bone Macrophage as Well as Associated Osteoimmunity in Glucocorticoid-Induced Osteonecrosis of the Femoral Head. J Inflamm Res 2023; 16:879-894. [PMID: 36891172 PMCID: PMC9986469 DOI: 10.2147/jir.s401968] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
Glucocorticoid-induced osteonecrosis of the femoral head (GIONFH) is a disabling disease with high mortality in China but the detailed molecular and cellular mechanisms remain to be investigated. Macrophages are considered the key cells in osteoimmunology, and the cross-talk between bone macrophages and other cells in the microenvironment is involved in maintaining bone homeostasis. M1 polarized macrophages launch a chronic inflammatory response and secrete a broad spectrum of cytokines (eg, TNF-α, IL-6 and IL-1β) and chemokines to initiate a chronic inflammatory state in GIONFH. M2 macrophage is the alternatively activated anti-inflammatory type distributed mainly in the perivascular area of the necrotic femoral head. In the development of GIONFH, injured bone vascular endothelial cells and necrotic bone activate the TLR4/NF-κB signal pathway, promote dimerization of PKM2 and subsequently enhance the production of HIF-1, inducing metabolic transformation of macrophage to the M1 phenotype. Considering these findings, putative interventions by local chemokine regulation to correct the imbalance between M1/M2 polarized macrophages by switching macrophages to an M2 phenotype, or inhibiting the adoption of an M1 phenotype appear to be plausible regimens for preventing or intervening GIONFH in the early stage. However, these results were mainly obtained by in vitro tissue or experimental animal model. Further studies to completely elucidate the alterations of the M1/M2 macrophage polarization and functions of macrophages in glucocorticoid-induced osteonecrosis of the femoral head are imperative.
Collapse
Affiliation(s)
- Qingyu Zhang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| | - Wei Sun
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Centre for Osteonecrosis and Joint-Preserving & Reconstruction, Orthopaedic Department, China Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Tengqi Li
- Department of Orthopedics, Peking University Shougang Hospital, Beijing, People’s Republic of China
- Department of Orthopedics, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, People’s Republic of China
| | - Fanxiao Liu
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
42
|
Influence of Perioperative Anesthesia on Cancer Recurrence: from Basic Science to Clinical Practice. Curr Oncol Rep 2023; 25:63-81. [PMID: 36512273 PMCID: PMC9745294 DOI: 10.1007/s11912-022-01342-9] [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] [Accepted: 08/02/2022] [Indexed: 12/15/2022]
Abstract
PURPOSEOF REVIEW In this review, we will summarize the effects of these perioperative anesthetics and anesthetic interventions on the immune system and tumorigenesis as well as address the related clinical evidence on cancer-related mortality and recurrence. RECENT FINDINGS Cancer remains a leading cause of morbidity and mortality worldwide. For many solid tumors, surgery is one of the major therapies. Unfortunately, surgery promotes angiogenesis, shedding of circulating cancer cells, and suppresses immunity. Hence, the perioperative period has a close relationship with cancer metastases or recurrence. In the perioperative period, patients require multiple anesthetic management including anesthetics, anesthetic techniques, and body temperature control. Preclinical and retrospective studies have found that these anesthetic agents and interventions have complex effects on cancer outcomes. Therefore, well-planned, prospective, randomized controlled trials are required to explore the effects of different anesthetics and techniques on long-term outcomes after cancer surgery. Due to the conflicting effects of anesthetic management on cancer recurrence, further preclinical and clinical trials are required and beneficial to the development of systemic cancer therapies.
Collapse
|
43
|
Batule S, Soldevila B, Figueredo C, Julián MT, Egea-Cortés L, Reyes-Ureña J, Casabona J, Mateu L, Paredes R, Clotet B, López R, Puig-Domingo M, Alonso N. Factors associated with critical care requirements in diabetic patients treated with dexamethasone for COVID-19 infection in the first wave of the pandemia. Front Endocrinol (Lausanne) 2022; 13:1009028. [PMID: 36619546 PMCID: PMC9815103 DOI: 10.3389/fendo.2022.1009028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Diabetes mellitus (DM) and hyperglycemia are important risk factors for poor outcomes in hospitalized patients with coronavirus disease 2019 (COVID-19). The aim of the present study was to analyze the factors associated with the composite outcome of the necessity of invasive mechanical ventilation (IMV) or admission to the intensive care unit (ICU) in subjects with severe COVID-19 infection treated with dexamethasone comparing patients with DM vs. patients without DM. Research design and methods An observational retrospective cohort study was performed, including hospitalized subjects with a diagnosis of SARS-CoV-2 pneumonia. Inclusion criteria were: age ≥18 years old with severe COVID-19 disease requiring daily intravenous 6 mg dexamethasone treatment for 10 days. Exclusion criteria were: <18 years old, non-severe illness and/or patients in charge of ICU. Variables related to clinical and analytical parameters, glycemic control, acquired-hospital superinfections, mortality, IMV requirement, ICU admission and length of stay were included. Results Two hundred and nine individuals with COVID-19 disease treated with dexamethasone were included. One hundred twenty-five out of these subjects (59.8%) were patients with DM. Overall, from the 209 subjects, 66 (31.6%) required IMV or were admitted to the ICU, with significant differences between patients with DM (n=50) vs. patients without DM (n=16) (76% vs. 24%, p=0.002). Among the group of subjects with DM (n=125), those who required IMV or were admitted to the ICU showed higher serum concentrations of C-reactive protein, interleukin-6, D-dimer, ferritin and pro-calcitonin and significantly lower serum concentrations of albumin compared to those who did not require IMV or were not admitted to the ICU. Besides, between these two groups of patients with DM, we observed no differences in glycemic parameters, including median capillary blood glucose values, glycosylated hemoglobin, coefficient of variability and hypoglycemic episodes. In the multinomial analysis, factors independently associated with the composite outcome of IMV or admission to the ICU in the insulin-treated group were the National Early Warning Score (NEWS) 2 score (OR 1.55 [1.17-2.17], p=0.005) and the presence of hospital-acquired superinfections (OR 35.21 [5.11-386.99], p=0.001). Conclusions In our study, parameters related to glycemic control were not associated with IMV requirement nor admission to the ICU in patients with DM and severe COVID-19 disease receiving daily 6 mg of dexamethasone for 10 days. However, hospital-acquired superinfections and disease severity at admission were independent factors associated with this composite outcome.
Collapse
Affiliation(s)
- Sol Batule
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute and Hospital, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Berta Soldevila
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute and Hospital, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carme Figueredo
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute and Hospital, Badalona, Spain
| | - María Teresa Julián
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute and Hospital, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laia Egea-Cortés
- Centre d'Estudis Epidemiològics sobre les Infeccions de Transmissió Sexual i Sida de Catalunya (CEEISCAT), Badalona, Spain
| | - Juliana Reyes-Ureña
- Centre d'Estudis Epidemiològics sobre les Infeccions de Transmissió Sexual i Sida de Catalunya (CEEISCAT), Badalona, Spain
| | - Jordi Casabona
- Centre d'Estudis Epidemiològics sobre les Infeccions de Transmissió Sexual i Sida de Catalunya (CEEISCAT), Badalona, Spain
| | - Lourdes Mateu
- Infectious Disease Service, Germans Trias i Pujol Research Institute and Hospital, Badalona, Spain
| | - Roger Paredes
- Infectious Disease Service, Germans Trias i Pujol Research Institute and Hospital, Badalona, Spain
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Bonaventura Clotet
- Infectious Disease Service, Germans Trias i Pujol Research Institute and Hospital, Badalona, Spain
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Rosa López
- Direcció d'Organització i Sistemes Gerència Territorial Metropolitana Nord, Institut Català de la Salut, Badalona, Spain
| | - Manel Puig-Domingo
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute and Hospital, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Núria Alonso
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute and Hospital, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
44
|
Kim D, Witt EE, Schubert S, Sotirchos E, Bhargava P, Mowry EM, Sachs K, Bilen B, Steinman L, Awani A, He Z, Calabresi PA, Van Haren K. Peripheral T-Cells, B-Cells, and Monocytes from Multiple Sclerosis Patients Supplemented with High-Dose Vitamin D Show Distinct Changes in Gene Expression Profiles. Nutrients 2022; 14:4737. [PMID: 36432424 PMCID: PMC9694020 DOI: 10.3390/nu14224737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Vitamin D is a steroid hormone that has been widely studied as a potential therapy for multiple sclerosis and other inflammatory disorders. Pre-clinical studies have implicated vitamin D in the transcription of thousands of genes, but its influence may vary by cell type. A handful of clinical studies have failed to identify an in vivo gene expression signature when using bulk analysis of all peripheral immune cells. We hypothesized that vitamin D's gene signature would vary by immune cell type, requiring the analysis of distinct cell types. Multiple sclerosis patients (n = 18) were given high-dose vitamin D (10,400 IU/day) for six months as part of a prospective clinical trial (NCT01024777). We collected peripheral blood mononuclear cells from participants at baseline and again after six months of treatment. We used flow cytometry to isolate three immune cell types (CD4+ T-cells, CD19+ B-cells, CD14+ monocytes) for RNA microarray analysis and compared the expression profiles between baseline and six months. We identified distinct sets of differentially expressed genes and enriched pathways between baseline and six months for each cell type. Vitamin D's in vivo gene expression profile in the immune system likely differs by cell type. Future clinical studies should consider techniques that allow for a similar cell-type resolution.
Collapse
Affiliation(s)
- Dohyup Kim
- Neurology and Neurological Sciences Department, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Simone Schubert
- Department of Environmental Health and Safety, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elias Sotirchos
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ellen M. Mowry
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Karen Sachs
- Next Generation Analytics, Palo Alto, CA 94301, USA
| | - Biter Bilen
- Data Science and Engineering Consultant, Mountain View, CA 94041, USA
| | - Lawrence Steinman
- Neurology and Neurological Sciences Department, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Avni Awani
- Neurology and Neurological Sciences Department, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zihuai He
- Neurology and Neurological Sciences Department, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Keith Van Haren
- Neurology and Neurological Sciences Department, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
45
|
Stepanov YK, Speidel JD, Herrmann C, Schmid N, Behr R, Köhn FM, Stöckl JB, Pickl U, Trottmann M, Fröhlich T, Mayerhofer A, Welter H. Profound Effects of Dexamethasone on the Immunological State, Synthesis and Secretion Capacity of Human Testicular Peritubular Cells. Cells 2022; 11:cells11193164. [PMID: 36231125 PMCID: PMC9562650 DOI: 10.3390/cells11193164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/21/2022] [Accepted: 10/04/2022] [Indexed: 11/21/2022] Open
Abstract
The functions of human testicular peritubular cells (HTPCs), forming a small compartment located between the seminiferous epithelium and the interstitial areas of the testis, are not fully known but go beyond intratesticular sperm transport and include immunological roles. The expression of the glucocorticoid receptor (GR) indicates that they may be regulated by glucocorticoids (GCs). Herein, we studied the consequences of the GC dexamethasone (Dex) in cultured HTPCs, which serves as a unique window into the human testis. We examined changes in cytokines, mainly by qPCR and ELISA. A holistic mass-spectrometry-based proteome analysis of cellular and secreted proteins was also performed. Dex, used in a therapeutic concentration, decreased the transcript level of proinflammatory cytokines, e.g., IL6, IL8 and MCP1. An siRNA-mediated knockdown of GR reduced the actions on IL6. Changes in IL6 were confirmed by ELISA measurements. Of note, Dex also lowered GR levels. The proteomic results revealed strong responses after 24 h (31 significantly altered cellular proteins) and more pronounced ones after 72 h of Dex exposure (30 less abundant and 42 more abundant cellular proteins). Dex also altered the composition of the secretome (33 proteins decreased, 13 increased) after 72 h. Among the regulated proteins were extracellular matrix (ECM) and basement membrane components (e.g., FBLN2, COL1A2 and COL3A1), as well as PTX3 and StAR. These results pinpoint novel, profound effects of Dex in HTPCs. If transferrable to the human testis, changes specifically in ECM and the immunological state of the testis may occur in men upon treatment with Dex for medical reasons.
Collapse
Affiliation(s)
| | - Jan Dominik Speidel
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University Munich, 82152 Planegg-Martinsried, Germany
| | - Carola Herrmann
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University Munich, 82152 Planegg-Martinsried, Germany
| | - Nina Schmid
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University Munich, 82152 Planegg-Martinsried, Germany
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | | | - Jan Bernd Stöckl
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 München, Germany
| | | | | | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, LMU München, 81377 München, Germany
| | - Artur Mayerhofer
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University Munich, 82152 Planegg-Martinsried, Germany
- Correspondence: (A.M.); (H.W.); Tel.: +49-89218075859 (A.M.); +49-89218071882 (H.W.)
| | - Harald Welter
- Biomedical Center, Cell Biology, Anatomy III, Faculty of Medicine, Ludwig Maximilian University Munich, 82152 Planegg-Martinsried, Germany
- Correspondence: (A.M.); (H.W.); Tel.: +49-89218075859 (A.M.); +49-89218071882 (H.W.)
| |
Collapse
|
46
|
Di Cola I, Cipriani P, Ruscitti P. Perspectives on the use of non-biological pharmacotherapy for adult-onset Still's disease. Expert Opin Pharmacother 2022; 23:1577-1587. [PMID: 36124816 DOI: 10.1080/14656566.2022.2126764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The treatment of the patients with adult-onset Still's disease (AOSD) remains largely empirical and it is based on the administration of immunosuppressive drugs. In this work, we described the use of non-biological pharmacotherapies for AOSD. AREA COVERED Although nonsteroidal anti-inflammatory drugs (NSAIDs) are employed during the diagnostic phase, glucocorticoids (GCs) are the first-line therapy, administered at the beginning of the disease. As second-line therapy, conventional synthetic disease modifying anti-rheumatic drugs (csDMARDs) are used when GCs do not fully control the disease and/or to reduce the dosage of concomitant GCs. Methotrexate (MTX) is the most commonly administered csDMARDs whereas calcineurin inhibitors (CNIs) are used in severe patients. The lack of achievement of clinical response may lead to the administration of biologic DMARDs, with or without csDMARDs. EXPERT OPINION The management of AOSD may benefit from the administration of non-biological pharmacotherapies, including GCs, MTX, and CNIs. These therapies showed efficacy in inducing a clinical response, in managing life-threatening complications, and may be well tolerated in combination with biologic DMARDs. However, further specific studies are needed to fully clarify the specific role of such drugs in clinical practice to improve the management of AOSD and to provide a more tailored treatment.
Collapse
Affiliation(s)
- Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
47
|
Denans N, Tran NTT, Swall ME, Diaz DC, Blanck J, Piotrowski T. An anti-inflammatory activation sequence governs macrophage transcriptional dynamics during tissue injury in zebrafish. Nat Commun 2022; 13:5356. [PMID: 36127326 PMCID: PMC9489698 DOI: 10.1038/s41467-022-33015-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 08/26/2022] [Indexed: 11/23/2022] Open
Abstract
Macrophages are essential for tissue repair and regeneration. Yet, the molecular programs, as well as the timing of their activation during and after tissue injury are poorly defined. Using a high spatio-temporal resolution single cell analysis of macrophages coupled with live imaging after sensory hair cell death in zebrafish, we find that the same population of macrophages transitions through a sequence of three major anti-inflammatory activation states. Macrophages first show a signature of glucocorticoid activation, then IL-10 signaling and finally the induction of oxidative phosphorylation by IL-4/Polyamine signaling. Importantly, loss-of-function of glucocorticoid and IL-10 signaling shows that each step of the sequence is independently activated. Lastly, we show that IL-10 and IL-4 signaling act synergistically to promote synaptogenesis between hair cells and efferent neurons during regeneration. Our results show that macrophages, in addition to a switch from M1 to M2, sequentially and independently transition though three anti-inflammatory pathways in vivo during tissue injury in a regenerating organ.
Collapse
Affiliation(s)
- Nicolas Denans
- Stowers Institute for Medical Research, 1000 east 50th street, Kansas City, MO, 64110, USA.
| | - Nhung T T Tran
- Stowers Institute for Medical Research, 1000 east 50th street, Kansas City, MO, 64110, USA
| | - Madeleine E Swall
- Stowers Institute for Medical Research, 1000 east 50th street, Kansas City, MO, 64110, USA
| | - Daniel C Diaz
- Stowers Institute for Medical Research, 1000 east 50th street, Kansas City, MO, 64110, USA
- Parse Biosciences, 201 Elliott Ave W, Suite 290, Seattle, WA, 98119, USA
| | - Jillian Blanck
- Stowers Institute for Medical Research, 1000 east 50th street, Kansas City, MO, 64110, USA
| | - Tatjana Piotrowski
- Stowers Institute for Medical Research, 1000 east 50th street, Kansas City, MO, 64110, USA.
| |
Collapse
|
48
|
de Souza MLM, Borçoi AR, Dutra BAB, Dos Santos Vieira T, Mendes SO, Nascimento IAA, Quaioto BR, Olinda AS, Cunha ER, Freitas FV, Pinheiro JA, Dos Santos JG, Sorroche BP, Arantes LMRB, Sartório CL, da Silva AMA. Lifestyle and NR3C1 exon 1F gene methylation is associated with changes in glucose levels and insulin resistance. Life Sci 2022; 309:120940. [PMID: 36108769 DOI: 10.1016/j.lfs.2022.120940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/03/2022] [Accepted: 09/05/2022] [Indexed: 10/31/2022]
Abstract
AIMS the present study aimed to investigate how glucose and insulin levels may be associated with changes in NR3C1 gene methylation levels in adults. MAIN METHODS 375 volunteers users of the Brazilian Public Unified Health System (SUS) were recruited to assess socioeconomic status, lifestyle, anthropometric data, blood glucose and serum cortisol levels, insulin resistance, and NR3C1 gene methylation assessment. Factors associated with glucose levels and insulin resistance were investigated using multivariate analysis GLzM at 5 % significance (p < 0.05). KEY FINDINGS our results verified that glucose levels and insulin resistance were directly related to NR3C1 gene methylation and age, while not being overweight and obese and no tobacco consumption were indirectly related to glucose levels and insulin resistance. SIGNIFICANCE habits and lifestyle may influence NR3C1 gene regulation, revealing the complexity of environmental impacts on NR3C1 methylation. Furthermore, associated risk factors must be taken into account in epigenetic studies as they directly interfere with blood glucose levels and insulin resistance.
Collapse
Affiliation(s)
| | - Aline Ribeiro Borçoi
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | | | - Tamires Dos Santos Vieira
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | | | | | - Barbara Risse Quaioto
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Amanda Sgrancio Olinda
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Ester Ribeiro Cunha
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Flávia Vitorino Freitas
- Department of Pharmacy and Nutrition, Universidade Federal do Espírito Santo, Alegre, ES, Brazil
| | - Julia Assis Pinheiro
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | | | - Bruna Pereira Sorroche
- Molecular Oncology Research Center, Hospital do Câncer de Barretos, Barretos, São Paulo, Brazil
| | | | - Carmem Luíza Sartório
- Biotechnology/Renorbio Postgraduate Program, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | | |
Collapse
|
49
|
Li R, Bhandari S, Martinez-Zubiaurre I, Bruun JA, Urbarova I, Smedsrød B, Simón-Santamaría J, Sørensen KK. Changes in the proteome and secretome of rat liver sinusoidal endothelial cells during early primary culture and effects of dexamethasone. PLoS One 2022; 17:e0273843. [PMID: 36054185 PMCID: PMC9439253 DOI: 10.1371/journal.pone.0273843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 08/16/2022] [Indexed: 11/28/2022] Open
Abstract
Introduction Liver sinusoidal endothelial cells (LSECs) are specialized fenestrated scavenger endothelial cells involved in the elimination of modified plasma proteins and tissue turnover waste macromolecules from blood. LSECs also participate in liver immune responses. A challenge when studying LSEC biology is the rapid loss of the in vivo phenotype in culture. In this study, we have examined biological processes and pathways affected during early-stage primary culture of rat LSECs and checked for cell responses to the pro-inflammatory cytokine interleukin (IL)-1β and the anti-inflammatory drug dexamethasone. Methods LSECs from male Sprague Dawley rats were cultured on type I collagen in 5% oxygen atmosphere in DMEM with serum-free supplements for 2 and 24 h. Quantitative proteomics using tandem mass tag technology was used to examine proteins in cells and supernatants. Validation was done with qPCR, ELISA, multiplex immunoassay, and caspase 3/7 assay. Cell ultrastructure was examined by scanning electron microscopy, and scavenger function by quantitative endocytosis assays. Results LSECs cultured for 24 h showed a characteristic pro-inflammatory phenotype both in the presence and absence of IL-1β, with upregulation of cellular responses to cytokines and interferon-γ, cell-cell adhesion, and glycolysis, increased expression of fatty acid binding proteins (FABP4, FABP5), and downregulation of several membrane receptors (STAB1, STAB2, LYVE1, CLEC4G) and proteins in pyruvate metabolism, citric acid cycle, fatty acid elongation, amino acid metabolism, and oxidation-reduction processes. Dexamethasone inhibited apoptosis and improved LSEC viability in culture, repressed inflammatory and immune regulatory pathways and secretion of IL-1β and IL-6, and further upregulated FABP4 and FABP5 compared to time-matched controls. The LSEC porosity and endocytic activity were reduced at 24 h both with and without dexamethasone but the dexamethasone-treated cells showed a less stressed phenotype. Conclusion Rat LSECs become activated towards a pro-inflammatory phenotype during early culture. Dexamethasone represses LSEC activation, inhibits apoptosis, and improves cell viability.
Collapse
Affiliation(s)
- Ruomei Li
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Sabin Bhandari
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | | | - Jack-Ansgar Bruun
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Ilona Urbarova
- Department of Community Medicine, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Bård Smedsrød
- Department of Medical Biology, UiT–The Arctic University of Norway, Tromsø, Norway
| | | | | |
Collapse
|
50
|
Rothenberg ME, Roufosse F, Faguer S, Gleich GJ, Steinfeld J, Yancey SW, Mavropoulou E, Kwon N. Mepolizumab Reduces Hypereosinophilic Syndrome Flares Irrespective of Blood Eosinophil Count and Interleukin-5. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:2367-2374.e3. [PMID: 35568330 DOI: 10.1016/j.jaip.2022.04.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/14/2022] [Accepted: 04/29/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Mepolizumab, an anti-interleukin-5 (IL-5) antibody, reduces disease flares in patients with hypereosinophilic syndrome (HES). Factors predicting treatment response are unknown. OBJECTIVE To assess mepolizumab efficacy by baseline blood eosinophil count (BEC) and serum IL-5 level in patients with HES. METHODS This post hoc analysis used data from the phase III study assessing mepolizumab in patients with HES (NCT02836496). Patients 12 years old or older, with HES for 6 or more months, 2 or more flares in the previous year, and BEC ≥1,000 cells/μL at screening were randomized (1:1) to 4-weekly subcutaneous mepolizumab (300 mg) or placebo, plus baseline HES therapy, for 32 weeks. The proportion of patients experiencing 1 or more flares (wk 32), annualized flare rate, and proportion of patients with change from baseline in Brief Fatigue Inventory (BFI) item 3 (wk 32), were analyzed by baseline BEC (<1500/≥1500 to <2500/≥2500 cells/μL). Flare outcomes were assessed by baseline serum IL-5 (<7.81/≥7.81 pg/mL). RESULTS Across baseline BEC subgroups, mepolizumab reduced the proportion of patients experiencing 1 or more flares by 63% to 90% and flare rate by 58% to 84% (treatment-by-eosinophil interaction P = .76 and P = .90, respectively); patients had improved BFI item 3 score with mepolizumab versus placebo (cells/μL: <1,500: 54% vs 37%; ≥1,500 to <2,500: 47% vs 31%; ≥2,500: 61% vs 0%; treatment-by-eosinophil interaction P = .42). Most patients had undetectable baseline serum IL-5 levels; among these, mepolizumab versus placebo reduced the proportion of patients with 1 or more flares (77%) and flare rate (67%). CONCLUSIONS Mepolizumab was efficacious in the patients with HES studied, irrespective of baseline BEC. Undetectable IL-5 levels should not preclude mepolizumab treatment.
Collapse
Affiliation(s)
- Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, and the Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Florence Roufosse
- Department of Internal Medicine, Erasmus Hospital, Free University of Brussels, Brussels, Belgium
| | - Stanislas Faguer
- Department of Nephrology and Organ Transplantation, Reference Center for Rare Kidney Diseases, Rangueil Hospital, Toulouse University Hospital, Toulouse, France
| | - Gerald J Gleich
- Departments of Dermatology and Medicine, School of Medicine, University of Utah, Salt Lake City, Utah
| | | | - Steven W Yancey
- Respiratory Therapeutic Area, GSK, Research Triangle Park, NC
| | | | - Namhee Kwon
- Respiratory Research & Development, GSK, Brentford, Middlesex, UK.
| | | |
Collapse
|