1
|
Peixoto DO, Bittencourt RR, Gasparotto J, Kessler FGC, Brum PO, Somensi N, Girardi CS, Dos Santos da Silva L, Outeiro TF, Moreira JCF, Gelain DP. Increased alpha-synuclein and neuroinflammation in the substantia nigra triggered by systemic inflammation are reversed by targeted inhibition of the receptor for advanced glycation end products (RAGE). J Neurochem 2024; 168:1587-1607. [PMID: 37661637 DOI: 10.1111/jnc.15956] [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: 03/16/2023] [Revised: 07/18/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023]
Abstract
The receptor for advanced glycation end products (RAGE) is a protein of the immunoglobulin superfamily capable of regulating inflammation. Considering the role of this receptor in the initiation and establishment of neuroinflammation, and the limited understanding of the function of RAGE in the maintenance of this condition, this study describes the effects of RAGE inhibition in the brain, through an intranasal treatment with the antagonist FPS-ZM1, in an animal model of chronic neuroinflammation induced by acute intraperitoneal injection of lipopolysaccharide (LPS). Seventy days after LPS administration (2 mg/kg, i.p.), Wistar rats received, intranasally, 1.2 mg of FPS-ZM1 over 14 days. On days 88 and 89, the animals were submitted to the open-field test and were killed on day 90 after the intraperitoneal injection of LPS. Our results indicate that blockade of encephalic RAGE attenuates LPS-induced chronic neuroinflammation in different brain regions. Furthermore, we found that intranasal FPS-ZM1 administration reduced levels of gliosis markers, RAGE ligands, and α-synuclein in the substantia nigra pars compacta. Additionally, the treatment also reversed the increase in S100 calcium-binding protein B (RAGE ligand) in the cerebrospinal fluid and the cognitive-behavioral deficits promoted by LPS-less time spent in the central zone of the open-field arena (more time in the lateral zones), decreased total distance traveled, and increased number of freezing episodes. In summary, our study demonstrates the prominent role of RAGE in the maintenance of a chronic neuroinflammatory state triggered by a single episode of systemic inflammation and also points to possible future RAGE-based therapeutic approaches to treat conditions in which chronic neuroinflammation and increased α-synuclein levels could play a relevant role, such as in Parkinson's disease.
Collapse
Affiliation(s)
- Daniel Oppermann Peixoto
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Alicante, Spain
| | - Reykla Ramon Bittencourt
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Juciano Gasparotto
- Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (ICB-UNIFAL), Alfenas, Brazil
| | - Flávio Gabriel Carazza Kessler
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | | | - Nauana Somensi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Carolina Saibro Girardi
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Lucas Dos Santos da Silva
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Natural Sciences, Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle Upon Tyne, UK
- Scientific Employee with an Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - José Cláudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| | - Daniel Pens Gelain
- Centro de Estudos em Estresse Oxidativo, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (ICBS-UFRGS), Porto Alegre, Brazil
| |
Collapse
|
2
|
Wegener AJ, Hyer MM, Targett I, Kloster A, Shaw GA, Rodriguez AMM, Dyer SK, Neigh GN. Behavior, synaptic mitochondria, and microglia are differentially impacted by chronic adolescent stress and repeated endotoxin exposure in male and female rats. Stress 2024; 27:2299971. [PMID: 38179979 PMCID: PMC11064104 DOI: 10.1080/10253890.2023.2299971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/16/2023] [Indexed: 01/06/2024] Open
Abstract
Early life adversity and chronic inflammation have both been associated with cognitive impairment and neural compromise. In this study, we investigated the interactions between a history of chronic adolescent stress (CAS) and repeated endotoxin exposure on behavior, synaptic mitochondria, and microglia in adult male and female Wistar rats. Adult rats from chronic stress and control conditions were exposed to either repeated endotoxin (lipopolysaccharide; LPS) or saline injections every 3 days for 9 weeks. In both sexes, repeated LPS, regardless of stress history, impaired working memory in the Y maze. Regarding spatial memory, LPS impaired function for females; whereas, CAS altered function in males. Although males had an increase in anxiety-like behavior shortly after CAS, there were no long-term effects on anxiety-like behavior or social interaction observed in males or females. Stress did not alter synaptic mitochondrial function in either sex. Repeated LPS altered synaptic mitochondrial function such that ATP production was increased in females only. There were no observed increases in IBA-1 positive cells within the hippocampus for either sex. However, LPS and CAS altered microglia morphology in females. Impact of repeated LPS was evident at the terminal endpoint with increased spleen weight in both sexes and decreased adrenal weight in males only. Circulating cytokines were not impacted by repeated LPS at the terminal endpoint, but evidence of CAS effects on cytokines in females were evident. These data suggest a long-term impact of chronic stress and an impact of repeated endotoxin challenge in adulthood; however, not all physiological and behavioral metrics examined were impacted by the paradigm employed in this study and the two environmental challenges rarely interacted.
Collapse
Affiliation(s)
- A J Wegener
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - M M Hyer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - I Targett
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - A Kloster
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - G A Shaw
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - A M M Rodriguez
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - S K Dyer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - G N Neigh
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
3
|
Affiliation(s)
- Vasileios Chortis
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Mateska I, Witt A, Hagag E, Sinha A, Yilmaz C, Thanou E, Sun N, Kolliniati O, Patschin M, Abdelmegeed H, Henneicke H, Kanczkowski W, Wielockx B, Tsatsanis C, Dahl A, Walch AK, Li KW, Peitzsch M, Chavakis T, Alexaki VI. Succinate mediates inflammation-induced adrenocortical dysfunction. eLife 2023; 12:e83064. [PMID: 37449973 PMCID: PMC10374281 DOI: 10.7554/elife.83064] [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: 08/30/2022] [Accepted: 07/13/2023] [Indexed: 07/18/2023] Open
Abstract
The hypothalamus-pituitary-adrenal (HPA) axis is activated in response to inflammation leading to increased production of anti-inflammatory glucocorticoids by the adrenal cortex, thereby representing an endogenous feedback loop. However, severe inflammation reduces the responsiveness of the adrenal gland to adrenocorticotropic hormone (ACTH), although the underlying mechanisms are poorly understood. Here, we show by transcriptomic, proteomic, and metabolomic analyses that LPS-induced systemic inflammation triggers profound metabolic changes in steroidogenic adrenocortical cells, including downregulation of the TCA cycle and oxidative phosphorylation, in mice. Inflammation disrupts the TCA cycle at the level of succinate dehydrogenase (SDH), leading to succinate accumulation and disturbed steroidogenesis. Mechanistically, IL-1β reduces SDHB expression through upregulation of DNA methyltransferase 1 (DNMT1) and methylation of the SDHB promoter. Consequently, increased succinate levels impair oxidative phosphorylation and ATP synthesis and enhance ROS production, leading to reduced steroidogenesis. Together, we demonstrate that the IL-1β-DNMT1-SDHB-succinate axis disrupts steroidogenesis. Our findings not only provide a mechanistic explanation for adrenal dysfunction in severe inflammation, but also offer a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Ivona Mateska
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Anke Witt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Eman Hagag
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Anupam Sinha
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Canelif Yilmaz
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Evangelia Thanou
- Center of Neurogenomics and Cognitive Research (CNCR), Department of Molecular and 10 Cellular Neurobiology, Vrije UniversiteitAmsterdamNetherlands
| | - Na Sun
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum MünchenMunichGermany
| | - Ourania Kolliniati
- Department of Clinical Chemistry, Medical School, University of CreteHeraklionGreece
| | - Maria Patschin
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Heba Abdelmegeed
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Holger Henneicke
- Department of Medicine III & Center for Healthy Ageing, Technische Universität DresdenDresdenGermany
- Center for Regenerative Therapies, TU Dresden, Technische Universität DresdenDresdenGermany
| | - Waldemar Kanczkowski
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Christos Tsatsanis
- Department of Clinical Chemistry, Medical School, University of CreteHeraklionGreece
| | - Andreas Dahl
- DRESDEN-concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität DresdenDresdenGermany
| | - Axel Karl Walch
- Research Unit Analytical Pathology, German Research Center for Environmental Health, Helmholtz Zentrum MünchenMunichGermany
| | - Ka Wan Li
- Center of Neurogenomics and Cognitive Research (CNCR), Department of Molecular and 10 Cellular Neurobiology, Vrije UniversiteitAmsterdamNetherlands
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität DresdenDresdenGermany
| |
Collapse
|
5
|
Zhang K, Hu Y, Li R, Li T. Single-cell atlas of murine adrenal glands reveals immune-adrenal crosstalk during systemic <i>Candida albicans</i> infection. Front Immunol 2022; 13:966814. [PMID: 36389688 PMCID: PMC9664004 DOI: 10.3389/fimmu.2022.966814] [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: 06/11/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Fungal sepsis remains a major health threat with high mortality, where the adrenal gland stress response has been rarely reported. <i>Candida albicans</i> (<i>C.albicans</i>) is the most common opportunistic fungal pathogen of life-threatening disseminated candidiasis and fungal sepsis. In the present study, we performed single-cell RNA sequencing (scRNA-Seq) using the 10x Genomics platform to analyze the changes in murine adrenal transcriptome following systemic <i>C.albicans</i> infection. A total of 16 021 cells were categorized into 18 transcriptionally distinct clusters, representing adrenocortical cells, endothelial cells, various immune cells, mesenchymal cells, smooth muscle cells, adrenal capsule, chromaffin cells, neurons and glials. As the main cell component in the adrenal gland responsible for steroidogenesis, the adrenocortical cells dramatically diminished and were further grouped into 10 subclusters, which differently distributed in the infected and uninfected samples. Pseudo-time analysis revealed transitions of the adrenocortical cells from the initial normal states to active or dysfunctional states following systemic <i>C.albicans</i> infection <i>via</i> two trajectory paths. Endothelial cells in the highly vascularized organ of adrenal gland further proliferated following infection, with the upregulation of genes positively regulating angiogenesis and downregulation of protective genes of endothelial cells. Immune cells were also excessively infiltrated in adrenal glands of <i>C.albicans</i>-infected mice. Macrophages dominated the immune microenvironments in murine adrenal glands both before and after <i>C.albicans</i> infection, mediating the crosstalk among the steroid-producing cells, endothelial cells and immune cells within the adrenal gland. NLR family, pyrin domain containing 3 (NLRP3, encoded by <i>Nlrp3</i>) and complement receptor 3 (CR3, encoded by <i>Itgam</i>) were found to be significantly upregulated on the adrenal macrophages upon systemic <i>C.albicans</i> infection and might play critical roles in mediating the myeloid response. Meanwhile, the number and strength of the interactions between the infiltrating immune cells and adrenal resident cells were unveiled by cell-cell communication analysis to be dramatically increased after systemic <i>C.albicans</i> infection, indicating that the immune-adrenal crosstalk might contribute to the compromised functions of adrenal cells. Overall, our comprehensive picture of the murine adrenal gland microenvironment in systemic <i>C.albicans</i> infection provides deeper insights into the immune-adrenal cell communications during fungal sepsis.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China,National Clinical Research Center for Skin and Immune Diseases, Beijing, China,Research Center for Medical Mycology, Peking University, Beijing, China,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
| | - Yuzhe Hu
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China,Key Laboratory of Medical Immunology, National Health Commission of the People's Republic of China, Beijing, China,Peking University Center for Human Disease Genomics, Beijing, China
| | - Ruoyu Li
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China,National Clinical Research Center for Skin and Immune Diseases, Beijing, China,Research Center for Medical Mycology, Peking University, Beijing, China,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China,*Correspondence: Ting Li, ; Ruoyu Li,
| | - Ting Li
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China,Key Laboratory of Medical Immunology, National Health Commission of the People's Republic of China, Beijing, China,Peking University Center for Human Disease Genomics, Beijing, China,*Correspondence: Ting Li, ; Ruoyu Li,
| |
Collapse
|
6
|
Liu YX, Yu Y, Liu JP, Liu WJ, Cao Y, Yan RM, Yao YM. Neuroimmune Regulation in Sepsis-Associated Encephalopathy: The Interaction Between the Brain and Peripheral Immunity. Front Neurol 2022; 13:892480. [PMID: 35832175 PMCID: PMC9271799 DOI: 10.3389/fneur.2022.892480] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/27/2022] [Indexed: 11/15/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE), the most popular cause of coma in the intensive care unit (ICU), is the diffuse cerebral damage caused by the septic challenge. SAE is closely related to high mortality and extended cognitive impairment in patients in septic shock. At present, many studies have demonstrated that SAE might be mainly associated with blood–brain barrier damage, abnormal neurotransmitter secretion, oxidative stress, and neuroimmune dysfunction. Nevertheless, the precise mechanism which initiates SAE and contributes to the long-term cognitive impairment remains largely unknown. Recently, a growing body of evidence has indicated that there is close crosstalk between SAE and peripheral immunity. The excessive migration of peripheral immune cells to the brain, the activation of glia, and resulting dysfunction of the central immune system are the main causes of septic nerve damage. This study reviews the update on the pathogenesis of septic encephalopathy, focusing on the over-activation of immune cells in the central nervous system (CNS) and the “neurocentral–endocrine–immune” networks in the development of SAE, aiming to further understand the potential mechanism of SAE and provide new targets for diagnosis and management of septic complications.
Collapse
Affiliation(s)
- Yu-xiao Liu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
- Department of Neurosurgery, The Chinese PLA General Hospital, Beijing, China
| | - Yang Yu
- Department of Traditional Chinese Medical Science, Sixth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Jing-peng Liu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
- Department of Traditional Chinese Medical Science, Sixth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Wen-jia Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Yang Cao
- Department of Neurosurgery, The Chinese PLA General Hospital, Beijing, China
| | - Run-min Yan
- Department of Neurosurgery, The Chinese PLA General Hospital, Beijing, China
- *Correspondence: Yong-ming Yao
| | - Yong-ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
- Run-min Yan
| |
Collapse
|
7
|
Cherkasova MN, Borovaya TG, Zhukhovitskii VG, Pukhalskaia VG. Results of Structural and Bacteriological Analysis of the Mouse Adrenal Glands in a Sepsis Model. Bull Exp Biol Med 2022; 173:67-71. [PMID: 35622248 DOI: 10.1007/s10517-022-05495-2] [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: 09/03/2021] [Indexed: 11/29/2022]
Abstract
The article presents the results of histological and bacteriological analysis of the adrenal glands in two models of sepsis caused by intraperitoneal administration of Pseudomonas aeruginosa strains 1623 and 5266 to sexually mature male C57BL/6 mice. In both models, histological changes in the adrenal glands in the dynamics of sepsis consist in cell destruction and decrease in the absolute areas of the zona glomerulosa, columnar part of the zona fasciculata, and medulla, the development of venous congestion in the absence of pronounced signs of leukocytic infiltration. Most adrenocortical cells of the zona glomerulosa and chromaffin cells of the medulla show signs of destruction. The columnar part of the zona fasciculata loses normal architectonics, the cells undergo degeneration and apoptosis, a significant part of cells in the deep layers of the zona fasciculata remain intact, but do not show tinctorial and ultrastructural signs of steroidogenesis. The active growth of P. aeruginosa colonies from adrenal homogenates in both models already in the first hours after infection of animals makes it possible to associate the revealed structural changes in the adrenal glands with the direct negative effect of P. aeruginosa, and high levels of mRNA of proinflammatory cytokines in the adrenal tissues raise the question of the possible synthesis of these modulators of inflammation in the adrenal parenchyma of septic animals.
Collapse
Affiliation(s)
- M N Cherkasova
- Laboratory of Indication and Ultrastructural Analysis of Microorganisms, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - T G Borovaya
- Laboratory of Indication and Ultrastructural Analysis of Microorganisms, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V G Zhukhovitskii
- Laboratory of Indication and Ultrastructural Analysis of Microorganisms, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia.,Department of Medical Chemistry and Immunopathology, Russian Medical Academy of Continuous Professional Education, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V G Pukhalskaia
- Department of Biological Chemistry, I. M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
8
|
Huang L, Liao J, Chen Y, Zou C, Zhang H, Yang X, Zhang Q, Li T, Mo L, Zeng Y, Bao M, Zhang F, Ye Y, Yang Z, Cheng J, Mo Z. Single-cell transcriptomes reveal characteristic features of cell types within the human adrenal microenvironment. J Cell Physiol 2021; 236:7308-7321. [PMID: 33934358 DOI: 10.1002/jcp.30398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 01/24/2023]
Abstract
Various cells within the adrenal microenvironment are important in maintaining the body homeostasis. However, our understanding of adrenal disease pathogenesis is limited by an incomplete molecular characterization of the cell types responsible for the organ's multiple homeostatic functions. We report a cellular landscape of the human adrenal gland using single-cell RNA sequencing. We reveal characteristic features of cell types within the human adrenal microenvironment and found immune activation of nonimmune cells in the adrenal endothelial cells. We also reveal that abundant immune cells occupied a lot of space in adrenal gland. Additionally, Sex-related diversity in the adrenocortical cells and different gene expression profiles between the left and right adrenal gland are also observed at single-cell resolution. Together, at single-cell resolution, the transcriptomic map presents a comprehensive view of the human adrenal gland, which serves as a fundamental baseline description of this organ and paves a way for the further studies of adrenal diseases.
Collapse
Affiliation(s)
- Lin Huang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Institute of Urology and Nephrology, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Jinling Liao
- Institute of Urology and Nephrology, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Yang Chen
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Institute of Urology and Nephrology, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Chunlin Zou
- Key Laboratory of Longevity and Ageing-related Diseases, Ministry of Education, Nanning, Guangxi Zhuang, China
| | - Haiying Zhang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Xiaobo Yang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Qinyun Zhang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Tianyu Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Institute of Urology and Nephrology, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China
| | - Linjian Mo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Institute of Urology and Nephrology, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China
| | - Yanyu Zeng
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Mengying Bao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Fangxing Zhang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Yu Ye
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Zhanbin Yang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Institute of Urology and Nephrology, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China
| | - Jiwen Cheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Institute of Urology and Nephrology, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| | - Zengnan Mo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Institute of Urology and Nephrology, The First Afliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang, China.,Guangxi key Laboratory for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning, Guangxi Zhuang, China
| |
Collapse
|
9
|
Fudulu DP, Horn G, Hazell G, Lefrançois-Martinez AM, Martinez A, Angelini GD, Lightman SL, Spiga F. Co-culture of monocytes and zona fasciculata adrenal cells: An in vitro model to study the immune-adrenal cross-talk. Mol Cell Endocrinol 2021; 526:111195. [PMID: 33571577 PMCID: PMC8024787 DOI: 10.1016/j.mce.2021.111195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/05/2021] [Accepted: 01/31/2021] [Indexed: 12/13/2022]
Abstract
The hypothalamic-pituitary-adrenal axis is the primary neuroendocrine system activated to re-establish homeostasis during periods of stress, including critical illness and major surgery. During critical illness, evidence suggests that locally induced inflammation of the adrenal gland could facilitate immune-adrenal cross-talk and, in turn, modulate cortisol secretion. It has been hypothesized that immune cells are necessary to mediate the effect of inflammatory stimuli on the steroidogenic pathway that has been observed in vivo. To test this hypothesis, we developed and characterized a trans-well co-culture model of THP1 (human monocytic cell)-derived macrophages and ATC7 murine zona fasciculata adrenocortical cells. We found that co-culture of ATC7 and THP1 cells results in a significant increase in the basal levels of IL-6 mRNA in ATC7 cells, and this effect was potentiated by treatment with LPS. Addition of LPS to co-cultures of ATC7 and THP1 significantly decreased the expression of key adrenal steroidogenic enzymes (including StAR and DAX-1), and this was also found in ATC7 cells treated with pro-inflammatory cytokines. Moreover, 24-h treatment with the synthetic glucocorticoid dexamethasone prevented the effects of LPS stimulation on IL-6, StAR and DAX-1 mRNA in ATC7 cells co-cultured with THP1 cells. Our data suggest that the expression of IL-6 and steroidogenic genes in response to LPS depends on the activation of intra-adrenal immune cells. Moreover, we also show that the effects of LPS can be modulated by glucocorticoids in a time- and dose-dependent manner with potential implications for clinical practice.
Collapse
Affiliation(s)
- Daniel P Fudulu
- Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, BS1 3NY, United Kingdom; Bristol Heart Institute, University of Bristol, 68 Horfield Rd, Bristol, BS2 8ED, United Kingdom.
| | - George Horn
- Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, BS1 3NY, United Kingdom
| | - Georgina Hazell
- Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, BS1 3NY, United Kingdom
| | - Anne-Marie Lefrançois-Martinez
- Génétique Reproduction & Développement, CNRS UMR 6293, Inserm U1103, Université Clermont Auvergne, 63001, Clermont-Ferrand, France
| | - Antoine Martinez
- Génétique Reproduction & Développement, CNRS UMR 6293, Inserm U1103, Université Clermont Auvergne, 63001, Clermont-Ferrand, France
| | - Gianni D Angelini
- Bristol Heart Institute, University of Bristol, 68 Horfield Rd, Bristol, BS2 8ED, United Kingdom
| | - Stafford L Lightman
- Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, BS1 3NY, United Kingdom
| | - Francesca Spiga
- Bristol Medical School: Translational Health Sciences, University of Bristol, Bristol, BS1 3NY, United Kingdom.
| |
Collapse
|
10
|
MHC/class-II-positive cells inhibit corticosterone of adrenal gland cells in experimental arthritis: a role for IL-1β, IL-18, and the inflammasome. Sci Rep 2020; 10:17071. [PMID: 33051554 PMCID: PMC7554037 DOI: 10.1038/s41598-020-74309-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/30/2020] [Indexed: 12/29/2022] Open
Abstract
In experimental arthritis, glucocorticoid secretion is inadequate relative to inflammation. We hypothesized that IL-1 is a key factor for inadequate glucocorticoid secretion in arthritic rats. Collagen type II—induced arthritis (CIA) in DA rats was the model to study effects of IL-1 on adrenal function. In the CIA model, an increase of intraadrenal MHCII-positive cells was observed. MHCII-positive cells or bone marrow-derived dendritic cells inhibited glucocorticoid secretion of adrenal gland cells. IL-1, but also IL-18 and the inflammasome were critical in glucocorticoid inhibition. Arthritic compared to control adrenal gland cells produced higher amounts of CXC chemokines from MHCII+ adrenal cells, particularly CINC-2, which is strongly dependent on presence of IL-1. In CIA, macrophages and/or dendritic cells inhibit glucocorticoid secretion via IL-1 in adrenal glands. These findings show that activated macrophages and/or dendritic cells inhibit glucocorticoid secretion in experimental arthritis and that IL-1β is a decisive factor.
Collapse
|
11
|
Ziogas A, Maekawa T, Wiessner JR, Le TT, Sprott D, Troullinaki M, Neuwirth A, Anastasopoulou V, Grossklaus S, Chung KJ, Sperandio M, Chavakis T, Hajishengallis G, Alexaki VI. DHEA Inhibits Leukocyte Recruitment through Regulation of the Integrin Antagonist DEL-1. THE JOURNAL OF IMMUNOLOGY 2020; 204:1214-1224. [PMID: 31980574 DOI: 10.4049/jimmunol.1900746] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
Leukocytes are rapidly recruited to sites of inflammation via interactions with the vascular endothelium. The steroid hormone dehydroepiandrosterone (DHEA) exerts anti-inflammatory properties; however, the underlying mechanisms are poorly understood. In this study, we show that an anti-inflammatory mechanism of DHEA involves the regulation of developmental endothelial locus 1 (DEL-1) expression. DEL-1 is a secreted homeostatic factor that inhibits β2-integrin-dependent leukocyte adhesion, and the subsequent leukocyte recruitment and its expression is downregulated upon inflammation. Similarly, DHEA inhibited leukocyte adhesion to the endothelium in venules of the inflamed mouse cremaster muscle. Importantly, in a model of lung inflammation, DHEA limited neutrophil recruitment in a DEL-1-dependent manner. Mechanistically, DHEA counteracted the inhibitory effect of inflammation on DEL-1 expression. Indeed, whereas TNF reduced DEL-1 expression and secretion in endothelial cells by diminishing C/EBPβ binding to the DEL-1 gene promoter, DHEA counteracted the inhibitory effect of TNF via activation of tropomyosin receptor kinase A (TRKA) and downstream PI3K/AKT signaling that restored C/EBPβ binding to the DEL-1 promoter. In conclusion, DHEA restrains neutrophil recruitment by reversing inflammation-induced downregulation of DEL-1 expression. Therefore, the anti-inflammatory DHEA/DEL-1 axis could be harnessed therapeutically in the context of inflammatory diseases.
Collapse
Affiliation(s)
- Athanasios Ziogas
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany;
| | - Tomoki Maekawa
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104.,Research Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, 951-8514 Niigata, Japan
| | - Johannes R Wiessner
- Walter Brendel Centre of Experimental Medicine and Institute of Cardiovascular Physiology and Pathophysiology, BioMedical Centre, Ludwig Maximilians University of Munich, 81377 Planegg-Martinsried, Germany; and
| | - Thi Trang Le
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - David Sprott
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Maria Troullinaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ales Neuwirth
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Vasiliki Anastasopoulou
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Sylvia Grossklaus
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Kyoung-Jin Chung
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Markus Sperandio
- Walter Brendel Centre of Experimental Medicine and Institute of Cardiovascular Physiology and Pathophysiology, BioMedical Centre, Ludwig Maximilians University of Munich, 81377 Planegg-Martinsried, Germany; and
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, United Kingdom
| | - George Hajishengallis
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Vasileia Ismini Alexaki
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany;
| |
Collapse
|
12
|
Vagnerová K, Vodička M, Hermanová P, Ergang P, Šrůtková D, Klusoňová P, Balounová K, Hudcovic T, Pácha J. Interactions Between Gut Microbiota and Acute Restraint Stress in Peripheral Structures of the Hypothalamic-Pituitary-Adrenal Axis and the Intestine of Male Mice. Front Immunol 2019; 10:2655. [PMID: 31798585 PMCID: PMC6878942 DOI: 10.3389/fimmu.2019.02655] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022] Open
Abstract
The gut microbiota play an important role in shaping brain functions and behavior, including the activity of the hypothalamus-pituitary-adrenocortical (HPA) axis. However, little is known about the effect of the microbiota on the distinct structures (hypothalamus, pituitary, and adrenals) of the HPA axis. In the present study, we analyzed the influence of the microbiota on acute restraint stress (ARS) response in the pituitary, adrenal gland, and intestine, an organ of extra-adrenal glucocorticoid synthesis. Using specific pathogen-free (SPF) and germ-free (GF) male BALB/c mice, we showed that the plasma corticosterone response to ARS was higher in GF than in SPF mice. In the pituitary, stress downregulated the expression of the gene encoding CRH receptor type 1 (Crhr1), upregulated the expression of the Fkbp5 gene regulating glucocorticoid receptor sensitivity and did not affect the expression of the proopiomelanocortin (Pomc) and glucocorticoid receptor (Gr) genes. In contrast, the microbiota downregulated the expression of pituitary Pomc and Crhr1 but had no effect on Fkbp5 and Gr. In the adrenals, the steroidogenic pathway was strongly stimulated by ARS at the level of the steroidogenic transcriptional regulator Sf-1, cholesterol transporter Star and Cyp11a1, the first enzyme of steroidogenic pathway. In contrast, the effect of the microbiota was significantly detected at the level of genes encoding steroidogenic enzymes but not at the level of Sf-1 and Star. Unlike adrenal Sf-1, the expression of the gene Lrh-1, which encodes the crucial transcriptional regulator of intestinal steroidogenesis, was modulated by the microbiota and ARS and this effect differed between the ileum and colon. The findings demonstrate that gut microbiota have an impact on the response of the pituitary, adrenals and intestine to ARS and that the interaction between stress and the microbiota during activation of glucocorticoid steroidogenesis differs between organs. The results suggest that downregulated expression of pituitary Pomc and Crhr1 in SPF animals might be an important factor in the exaggerated HPA response of GF mice to stress.
Collapse
Affiliation(s)
- Karla Vagnerová
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Vodička
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Petra Hermanová
- Institute of Microbiology of the Czech Academy of Sciences, Nový Hrádek, Czechia
| | - Peter Ergang
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Dagmar Šrůtková
- Institute of Microbiology of the Czech Academy of Sciences, Nový Hrádek, Czechia
| | - Petra Klusoňová
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Kateřina Balounová
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia.,Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Tomáš Hudcovic
- Institute of Microbiology of the Czech Academy of Sciences, Nový Hrádek, Czechia
| | - Jiří Pácha
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia.,Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| |
Collapse
|
13
|
BCL6 inhibitor FX1 attenuates inflammatory responses in murine sepsis through strengthening BCL6 binding affinity to downstream target gene promoters. Int Immunopharmacol 2019; 75:105789. [PMID: 31401377 DOI: 10.1016/j.intimp.2019.105789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Sepsis occurs when an infection triggers deranged inflammatory responses. There exists no efficacious treatment for this condition. The transcriptional repressor B-cell Lymphoma 6 (BCL6) is known to act as an inhibitor of macrophage-mediated inflammatory responses. FX1, a novel specific BCL6 BTB inhibitor, is able to attenuate activity of B cell-like diffuse large B cell lymphoma (ABC-DLBCL). Nevertheless, the effect of FX1 in inflammatory responses and sepsis remains unknown. OBJECTIVES Here, we explored the effect and potential mechanisms of FX1 on the regulation of LPS-induced inflammatory responses in murine sepsis. METHOD Mice models of LPS-induced sepsis were monitored for survival rate following FX1 administration. ELISA was used to assess how FX1 administration affected pro-inflammatory cytokines present in macrophages exposed to LPS and in the serum of mice sepsis models. Flow cytometric analysis, Western blot and qRT-PCR were performed to evaluate differences in macrophages immune responses after FX1 pre-treatment. Finally, the affinity of BCL6 binding to downstream target genes was checked by ChIP. RESULTS The survival rate of mice models of LPS-induced sepsis was improved in following FX1 administration. FX1 decreased the production of inflammatory cytokines, attenuated macrophage infiltration activities and reduced monocytes chemotaxis activities, all of which suggest that FX1 exert anti-inflammatory effects. Mechanistically, FX1 may enhance the affinity of BCL6 binding to downstream target pro-inflammatory genes. CONCLUSIONS These findings illustrated the anti-inflammatory properties and potential mechanisms of FX1 in sepsis caused by LPS. FX1 could potentially become a new immunosuppressive and anti-inflammatory drug candidate in sepsis therapy.
Collapse
|
14
|
Chen LS, Singh SP, Schuster M, Grinenko T, Bornstein SR, Kanczkowski W. RNA-seq analysis of LPS-induced transcriptional changes and its possible implications for the adrenal gland dysregulation during sepsis. J Steroid Biochem Mol Biol 2019; 191:105360. [PMID: 31028792 DOI: 10.1016/j.jsbmb.2019.04.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/27/2019] [Accepted: 04/07/2019] [Indexed: 12/22/2022]
Abstract
Activation of the adrenal gland stress response is of utmost importance to survive sepsis. Experimental and clinical evidence exists demonstrating that adrenal gland often develops functional and structural damage due to sepsis with mechanisms remaining largely unknown. In the present study, we have used RNA Sequencing (RNA-Seq) technology to analyze changes in adrenal transcriptome elucidated by bacterial LPS. We aimed to find particularly alterations in genes that were previously not reported to be involved in the adrenal gland dysregulation in contexts of sepsis. Our results demonstrate that systemic administration of LPS significantly altered expression of 8458 genes as compared to saline injected animals. The subsequent quality and functional analysis of these gene signatures revealed that LPS-induced highly homogenous transcriptional response in total upregulating 4312 and downregulating 4146 genes. Furthermore, functional annotation analysis together with gene enrichment set analysis (GSEA) clearly demonstrated that adrenal response to LPS involved alterations in multiple pathways related to the inflammatory response along with previously unexplored activation of the hypoxia pathway. In addition, LPS strongly downregulated genes involved in the adrenal homeostasis, development, and regeneration. Those alterations were subsequently verified in clinically relevant cecal ligation and puncture (CLP)-induced sepsis model. Collectively, our study demonstrates that RNA-seq is a very useful method that can be applied to search for new unexplored pathways potentially involved in adrenal gland dysregulation during sepsis.
Collapse
Affiliation(s)
- Lan-Sun Chen
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Sumeet Pal Singh
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Technische Universität Dresden, Dresden, 01307, Germany
| | - Maria Schuster
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Tatyana Grinenko
- Department of Clinical Pathobiochemistry, Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Waldemar Kanczkowski
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
15
|
Varodayan FP, Khom S, Patel RR, Steinman MQ, Hedges DM, Oleata CS, Homanics GE, Roberto M, Bajo M. Role of TLR4 in the Modulation of Central Amygdala GABA Transmission by CRF Following Restraint Stress. Alcohol Alcohol 2019; 53:642-649. [PMID: 29309503 PMCID: PMC6203127 DOI: 10.1093/alcalc/agx114] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/08/2017] [Indexed: 12/20/2022] Open
Abstract
Aims Stress induces neuroimmune responses via Toll-like receptor 4 (TLR4) activation. Here, we investigated the role of TLR4 in the effects of the stress peptide corticotropin-releasing factor (CRF) on GABAergic transmission in the central nucleus of the amygdala (CeA) following restraint stress. Methods Tlr4 knock out (KO) and wild-type rats were exposed to no stress (naïve), a single restraint stress (1 h) or repeated restraint stress (1 h per day for 3 consecutive days). After 1 h recovery from the final stress session, whole-cell patch-clamp electrophysiology was used to investigate the effects of CRF (200 nM) on CeA GABAA-mediated spontaneous inhibitory postsynaptic currents (sIPSCs). Results TLR4 does not regulate baseline GABAergic transmission in the CeA of naive and stress-treated animals. However, CRF significantly increased the mean sIPSC frequencies (indicating enhanced GABA release) across all genotypes and stress treatments, except for the Tlr4 KO rats that experienced repeated restraint stress. Conclusions Overall, our results suggest a limited role for TLR4 in CRF's modulation of CeA GABAergic synapses in naïve and single stress rats, though TLR4-deficient rats that experienced repeated psychological stress exhibit a blunted CRF cellular response. Short Summary TLR4 has a limited role in CRF's activation of the CeA under basal conditions, but interacts with the CRF system to regulate GABAergic synapse function in animals that experience repeated psychological stress.
Collapse
Affiliation(s)
- F P Varodayan
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - S Khom
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - R R Patel
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - M Q Steinman
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - D M Hedges
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - C S Oleata
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - G E Homanics
- Departments of Anesthesiology, Pharmacology & Chemical Biology, and Neurobiology, University of 6060 Biomedical Science Tower 3, Pittsburgh, Pittsburgh, PA, USA
| | - M Roberto
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - M Bajo
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| |
Collapse
|
16
|
Sominsky L, Ong LK, Ziko I, Dickson PW, Spencer SJ. Neonatal overfeeding increases capacity for catecholamine biosynthesis from the adrenal gland acutely and long-term in the male rat. Mol Cell Endocrinol 2018; 470:295-303. [PMID: 29183807 DOI: 10.1016/j.mce.2017.11.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 12/22/2022]
Abstract
A poor nutritional environment during early development has long been known to increase disease susceptibility later in life. We have previously shown that rats that are overfed as neonates (i.e. suckled in small litters (4 pups) relative to control conditions (12 pups)) show dysregulated hypothalamic-pituitary-adrenal axis responses to immune stress in adulthood, particularly due to an altered capacity of the adrenal to respond to an immune challenge. Here we hypothesised that neonatal overfeeding similarly affects the sympathomedullary system, testing this by investigating the biochemical function of tyrosine hydroxylase (TH), the first rate-limiting enzyme in the catecholamine synthesis. We also examined changes in adrenal expression of the leptin receptor and in mitogen-activated protein kinase (MAPK) signalling. During the neonatal period, we saw age-dependent changes in TH activity and phosphorylation, with neonatal overfeeding stimulating increased adrenal TH specific activity at postnatal days 7 and 14, along with a compensatory reduction in total TH protein levels. This increased TH activity was maintained into adulthood where neonatally overfed rats exhibited increased adrenal responsiveness 30 min after an immune challenge with lipopolysaccharide, evident in a concomitant increase in TH protein levels and specific activity. Neonatal overfeeding significantly reduced the expression of the leptin receptor in neonatal adrenals at postnatal day 7 and in adult adrenals, but did not affect MAPK signalling. These data suggest neonatal overfeeding alters the capacity of the adrenal to synthesise catecholamines, both acutely and long term, and these effects may be independent of leptin signalling.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia.
| | - Lin Kooi Ong
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, NSW, Australia
| | - Ilvana Ziko
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia
| | - Phillip W Dickson
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, NSW, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., Australia
| |
Collapse
|
17
|
Death of adrenocortical cells during murine acute T. cruzi infection is not associated with TNF-R1 signaling but mostly with the type II pathway of Fas-mediated apoptosis. Brain Behav Immun 2017; 65:284-295. [PMID: 28666938 DOI: 10.1016/j.bbi.2017.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/22/2017] [Accepted: 05/26/2017] [Indexed: 01/28/2023] Open
Abstract
Earlier studies from our laboratory demonstrated that acute experimental Trypanosoma cruzi infection promotes an intense inflammation along with a sepsis-like dysregulated adrenal response characterized by normal levels of ACTH with raised glucocorticoid secretion. Inflammation was also known to result in adrenal cell apoptosis, which in turn may influence HPA axis uncoupling. To explore factors and pathways which may be involved in the apoptosis of adrenal cells, together with its impact on the functionality of the gland, we carried out a series of studies in mice lacking death receptors, such as TNF-R1 (C57BL/6-Tnfrsf1a tm1Imx or TNF-R1-/-) or Fas ligand (C57BL/6 Fas-deficient lpr mice), undergoing acute T. cruzi infection. Here we demonstrate that the late hypercorticosterolism seen in C57BL/6 mice during acute T. cruzi infection coexists with and hyperplasia and hypertrophy of zona fasciculata, paralleled by increased number of apoptotic cells. Apoptosis seems to be mediated mainly by the type II pathway of Fas-mediated apoptosis, which engages the mitochondrial pathway of apoptosis triggering the cytochrome c release to increase caspase-3 activation. Fas-induced apoptosis of adrenocortical cells is also related with an exacerbated production of intra-adrenal cytokines that probably maintain the late supply of adrenal hormones during host response. Present results shed light on the molecular mechanisms dealing with these phenomena which are crucial not only for the development of interventions attempting to avoid adrenal dysfunction, but also for its wide occurrence in other infectious-based critical illnesses.
Collapse
|
18
|
|
19
|
Merlo E, Podratz PL, Sena GC, de Araújo JFP, Lima LCF, Alves ISS, Gama-de-Souza LN, Pelição R, Rodrigues LCM, Brandão PAA, Carneiro MTWD, Pires RGW, Martins-Silva C, Alarcon TA, Miranda-Alves L, Silva IV, Graceli JB. The Environmental Pollutant Tributyltin Chloride Disrupts the Hypothalamic-Pituitary-Adrenal Axis at Different Levels in Female Rats. Endocrinology 2016; 157:2978-95. [PMID: 27267847 DOI: 10.1210/en.2015-1896] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tributyltin chloride (TBT) is an environmental contaminant that is used as a biocide in antifouling paints. TBT has been shown to induce endocrine-disrupting effects. However, studies evaluating the effects of TBT on the hypothalamus-pituitary-adrenal (HPA) axis are especially rare. The current study demonstrates that exposure to TBT is critically responsible for the improper function of the mammalian HPA axis as well as the development of abnormal morphophysiology in the pituitary and adrenal glands. Female rats were treated with TBT, and their HPA axis morphophysiology was assessed. High CRH and low ACTH expression and high plasma corticosterone levels were detected in TBT rats. In addition, TBT leads to an increased in the inducible nitric oxide synthase protein expression in the hypothalamus of TBT rats. Morphophysiological abnormalities, including increases in inflammation, a disrupted cellular redox balance, apoptosis, and collagen deposition in the pituitary and adrenal glands, were observed in TBT rats. Increases in adiposity and peroxisome proliferator-activated receptor-γ protein expression in the adrenal gland were observed in TBT rats. Together, these data provide in vivo evidence that TBT leads to functional dissociation between CRH, ACTH, and costicosterone, which could be associated an inflammation and increased of inducible nitric oxide synthase expression in hypothalamus. Thus, TBT exerts toxic effects at different levels on the HPA axis function.
Collapse
Affiliation(s)
- Eduardo Merlo
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Priscila L Podratz
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Gabriela C Sena
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Julia F P de Araújo
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Leandro C F Lima
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Izabela S S Alves
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Letícia N Gama-de-Souza
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Renan Pelição
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Lívia C M Rodrigues
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Poliane A A Brandão
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Maria T W D Carneiro
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Rita G W Pires
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Cristina Martins-Silva
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Tamara A Alarcon
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Leandro Miranda-Alves
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Ian V Silva
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Jones B Graceli
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| |
Collapse
|
20
|
Kanczkowski W, Sue M, Bornstein SR. Adrenal Gland Microenvironment and Its Involvement in the Regulation of Stress-Induced Hormone Secretion during Sepsis. Front Endocrinol (Lausanne) 2016; 7:156. [PMID: 28018291 PMCID: PMC5155014 DOI: 10.3389/fendo.2016.00156] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/29/2016] [Indexed: 01/11/2023] Open
Abstract
Survival of all living organisms depends on maintenance of a steady state of homeostasis, which process relies on its ability to react and adapt to various physical and emotional threats. The defense against stress is executed by the hypothalamic-pituitary-adrenal axis and the sympathetic-adrenal medullary system. Adrenal gland is a major effector organ of stress system. During stress, adrenal gland rapidly responds with increased secretion of glucocorticoids (GCs) and catecholamines into circulation, which hormones, in turn, affect metabolism, to provide acutely energy, vasculature to increase blood pressure, and the immune system to prevent it from extensive activation. Sepsis resulting from microbial infections is a sustained and extreme example of stress situation. In many critical ill patients, levels of both corticotropin-releasing hormone and adrenocorticotropin, the two major regulators of adrenal hormone production, are suppressed. Levels of GCs, however, remain normal or are elevated in these patients, suggesting a shift from central to local intra-adrenal regulation of adrenal stress response. Among many mechanisms potentially involved in this process, reduced GC metabolism and activation of intra-adrenal cellular systems composed of adrenocortical and adrenomedullary cells, endothelial cells, and resident and recruited immune cells play a key role. Hence, dysregulated function of any of these cells and cellular compartments can ultimately affect adrenal stress response. The purpose of this mini review is to highlight recent insights into our understanding of the adrenal gland microenvironment and its role in coordination of stress-induced hormone secretion.
Collapse
Affiliation(s)
- Waldemar Kanczkowski
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
- *Correspondence: Waldemar Kanczkowski,
| | - Mariko Sue
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Stefan R. Bornstein
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
- Department of Endocrinology and Diabetes, King’s College London, London, UK
| |
Collapse
|
21
|
Kanczkowski W, Sue M, Zacharowski K, Reincke M, Bornstein SR. The role of adrenal gland microenvironment in the HPA axis function and dysfunction during sepsis. Mol Cell Endocrinol 2015; 408:241-8. [PMID: 25543020 DOI: 10.1016/j.mce.2014.12.019] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/21/2014] [Accepted: 12/21/2014] [Indexed: 12/21/2022]
Abstract
Sepsis and septic shock in response to bacterial or viral infections remain the major health problem worldwide. Despite decades of intensive research and improvements in medical care, severe sepsis is associated with high mortality. Rapid activation of the adrenal gland glucocorticoid and catecholamine production is a fundamental component of the stress response and is essential for survival of the host. However, in many critically ill patients this homeostatic function of the adrenal gland is often impaired. In these patients, plasma levels of adrenocorticotropic hormone (ACTH) and cortisol are often dissociated. This has been attributed to the stimulatory action of non-ACTH factors within the adrenal gland such as cytokines, and recently with decreased cortisol metabolism and suppressed ACTH synthesis. Regulation of the hypothalamus-pituitary-adrenal (HPA) axis function during sepsis is a complex process which involves various immune and neuroendocrine interactions occurring at the levels of the central nervous system (CNS) and the adrenal gland. A coordinated interaction of numerous cell types and systems within the adrenal gland is involved in the sustained adrenal glucocorticoid production. This review article describes and discusses recent experimental findings regarding the role of adrenal gland microenvironment including the adrenal vasculature and the immune-adrenal crosstalk in the disregulated HPA axis during sepsis conditions. In summary, in addition to the reduced cortisol breakdown and related ACTH suppression, sepsis-mediated chronic activation of the immune-adrenal crosstalk and vascular dysfunction may contribute to the HPA axis dysregulation found in septic patients.
Collapse
Affiliation(s)
- Waldemar Kanczkowski
- Department of Medicine III, Faculty of Medicine of the Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany.
| | - Mariko Sue
- Department of Medicine III, Faculty of Medicine of the Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Kai Zacharowski
- Clinic of Anesthesiology, Intensive Care Medicine and Pain Therapy, 60595 Frankfurt am Main, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, München, Germany
| | - Stefan R Bornstein
- Department of Medicine III, Faculty of Medicine of the Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| |
Collapse
|
22
|
Wang CN, Duan GL, Liu YJ, Yu Q, Tang XL, Zhao W, Li XH, Zhu XY, Ni X. Overproduction of nitric oxide by endothelial cells and macrophages contributes to mitochondrial oxidative stress in adrenocortical cells and adrenal insufficiency during endotoxemia. Free Radic Biol Med 2015; 83:31-40. [PMID: 25744413 DOI: 10.1016/j.freeradbiomed.2015.02.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 02/08/2015] [Accepted: 02/21/2015] [Indexed: 01/20/2023]
Abstract
We have recently demonstrated that lipopolysaccharide (LPS) causes mitochondrial oxidative stress and dysfunction in adrenal glands, thereby leading to adrenocortical insufficiency. Since nitric oxide (NO) produced by inducible nitric oxide synthase (iNOS) leads to mitochondrial damage in various tissues, the present study aims to investigate whether NO contributes to mitochondrial oxidative stress in adrenal cortex and adrenocortical insufficiency during endotoxemia. Systemic administration of LPS increased iNOS expression and NO production in adrenal glands of mice. The specific iNOS inhibitor 1400 W significantly attenuated the LPS-induced mitochondrial superoxide production and dysfunction in adrenal glands, and reversed the LPS-induced adrenocortical hyporesponsiveness to adrenocorticotropic hormone (ACTH). In contrast, administration of the NO donor sodium nitroprusside (SNP) led to mitochondrial oxidative stress and dysfunction in adrenal glands, which resulted in a blunted corticosterone response to ACTH. Using double immunofluorescence staining for iNOS with the vascular endothelial cell marker CD31 or the macrophage marker CD68, we found that increased iNOS expression was found in vascular endothelial cells and macrophages, but not adrenocortical cells in the adrenal gland during endotoxemia. Administration of the hydrogen sulfide (H2S) donor GYY4137 inhibited NO production and reversed LPS-induced adrenocortical hyporesponsiveness. Our data suggest that overproduction of NO, which is mainly generated by endothelial cells and macrophages during endotoxemia, contributes to mitochondrial oxidative stress in adrenocortical cells and subsequently leads to adrenal insufficiency.
Collapse
Affiliation(s)
- Chang-Nan Wang
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai, 200433, China
| | - Guo-Li Duan
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai, 200433, China; The Eight-year Program on Clinical Medicine, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yu-Jian Liu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, 200438, China
| | - Qing Yu
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai, 200433, China
| | - Xiao-Lu Tang
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai, 200433, China
| | - Wei Zhao
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai, 200433, China
| | - Xiao-Han Li
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai, 200433, China
| | - Xiao-Yan Zhu
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai, 200433, China.
| | - Xin Ni
- Department of Physiology and The Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai, 200433, China.
| |
Collapse
|
23
|
Hua YL, Ji P, Xue ZY, Wei YM. Construction and analysis of correlation networks based on gas chromatography-mass spectrometry metabonomics data for lipopolysaccharide-induced inflammation and intervention with volatile oil from Angelica sinensis in rats. MOLECULAR BIOSYSTEMS 2015; 11:3174-87. [DOI: 10.1039/c5mb00405e] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Angelica sinensis (AS) is a well-known important traditional Chinese medicine that yields a volatile oil with anti-inflammatory effects.
Collapse
Affiliation(s)
- Yong-li Hua
- College of Veterinary Medicine
- Gansu Agricultural University
- Lanzhou
- People's Republic of China
| | - Peng Ji
- College of Veterinary Medicine
- Gansu Agricultural University
- Lanzhou
- People's Republic of China
| | - Zi-yu Xue
- College of Veterinary Medicine
- Gansu Agricultural University
- Lanzhou
- People's Republic of China
| | - Yan-ming Wei
- College of Veterinary Medicine
- Gansu Agricultural University
- Lanzhou
- People's Republic of China
| |
Collapse
|
24
|
Chen Y, Liu H, Xu S, Wang T, Li W. Targeting microsomal prostaglandin E2synthase-1 (mPGES-1): the development of inhibitors as an alternative to non-steroidal anti-inflammatory drugs (NSAIDs). MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00278h] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AA cascade and several key residues in the 3D structure of mPGES-1.
Collapse
Affiliation(s)
- Yuqing Chen
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | | | - Shuang Xu
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | - Tianlin Wang
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| |
Collapse
|
25
|
Wang CN, Liu YJ, Duan GL, Zhao W, Li XH, Zhu XY, Ni X. CBS and CSE are critical for maintenance of mitochondrial function and glucocorticoid production in adrenal cortex. Antioxid Redox Signal 2014; 21:2192-207. [PMID: 24702258 DOI: 10.1089/ars.2013.5682] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIMS Mitochondria are known to play a central role in adrenocortical steroidogenesis. Recently, hydrogen sulfide (H2S), a gaseous transmitter endogenously produced by cystathionine-β-synthase (CBS) and cystathionine-γ-lyase (CSE), has been found to improve mitochondrial function. The present study aimed at examining whether CBS and CSE are expressed in adrenal glands, and investigated the role of these enzymes in the maintenance of mitochondrial function and the production of glucocorticoids in adrenocortical cells. RESULTS Both CBS and CSE are present in murine adrenocortical cells and account for H2S generation in adrenal glands. Using a combination of both in vivo and in vitro approaches, we demonstrated that either CBS/CSE inhibitors or small interfering RNAs led to mitochondrial oxidative stress and dysfunction, which meanwhile resulted in blunted corticosterone responses to adrenocorticotropic hormone (ACTH). These effects were significantly attenuated by the treatment of H2S donor GYY4137. Lipopolysaccharide (LPS) also caused mitochondrial damage, thereby resulting in adrenal insufficiency. Moreover, LPS inhibited CBS/CSE expression and H2S production in adrenal glands, while H₂S donor GYY4137 protected against LPS-induced mitochondrial damage and hyporesponsiveness to ACTH. Local suppression of CBS or CSE in adrenal glands significantly increased the mortality in endotoxemic mice, which was also improved by GYY4137. INNOVATION The identification of endogenous H2S generation as critical regulators of adrenocortical responsiveness might result in the development of new therapeutic approaches for the treatment of relative adrenal insufficiency during sepsis. CONCLUSIONS Endogenous H₂S plays a critical role in the maintenance of mitochondrial function in the adrenal cortex, thereby resulting in an adequate adrenocortical response to ACTH.
Collapse
Affiliation(s)
- Chang-Nan Wang
- 1 The Key Laboratory of Molecular Neurobiology of Ministry of Education, Department of Physiology, Second Military Medical University , Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Ge Y, Hu S, Zhang Y, Wang W, Xu Q, Zhou L, Mao H. Levobupivacaine inhibits lipopolysaccharide-induced high mobility group box 1 release in vitro and in vivo. J Surg Res 2014; 192:582-91. [DOI: 10.1016/j.jss.2014.05.087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 05/28/2014] [Accepted: 05/29/2014] [Indexed: 12/11/2022]
|
27
|
Zhang Z, Zhang L, Zhou C, Wu H. Ketamine inhibits LPS-induced HGMB1 release in vitro and in vivo. Int Immunopharmacol 2014; 23:14-26. [DOI: 10.1016/j.intimp.2014.08.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 07/31/2014] [Accepted: 08/04/2014] [Indexed: 11/26/2022]
|
28
|
Fan X, Zhang Y, Dong H, Wang B, Ji H, Liu X. Trilobatin attenuates the LPS-mediated inflammatory response by suppressing the NF-κB signaling pathway. Food Chem 2014; 166:609-615. [PMID: 25053100 DOI: 10.1016/j.foodchem.2014.06.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 04/29/2014] [Accepted: 06/05/2014] [Indexed: 12/12/2022]
Abstract
We investigated the anti-inflammatory effect of trilobatin, the flavonoid isolated from the leaves of Lithocarpus polystachyus Rehd, as well as the underlying molecular mechanisms. Treatment with trilobatin (0.005-5 μM) dose-dependently inhibited the lipopolysaccharide (LPS)-induced mRNA expression and secretion of pro-inflammatory cytokines, including tumor necrosis factor α (TNFα), interleukin-1β (IL-1β) and interleukin-6 (IL-6), in RAW 264.7 macrophages. However, no further inhibition was detected when the concentration of trilobatin was increased to 50 μM. Western blot analysis confirmed that the mechanism of the anti-inflammatory effect was correlated with the inhibition of LPS-induced inhibitor of nuclear factor-kappa B α (IκBα) degradation and nuclear factor-kappa B (NF-κB) p65 phosphorylation. In addition, trilobatin also showed a significant inhibition of LPS-induced TNFα and IL-6 at both the mRNA and protein levels in a mouse model. Our results suggest that trilobatin potentially inhibits the LPS-induced inflammatory response by suppressing the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiaolong Fan
- Department of Food Science, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yinghui Zhang
- Department of Food Safety, College of Food and Horticultural Sciences, Foshan University (Northern Campus), Foshan 528231, China.
| | - Huaqiang Dong
- Department of Food Safety, College of Food and Horticultural Sciences, Foshan University (Northern Campus), Foshan 528231, China
| | - Bingyun Wang
- Department of Veterinary Medicine, College of Life Science, Foshan University (Northern Campus), Foshan 528231, China
| | - Huiqin Ji
- Department of Veterinary Medicine, College of Life Science, Foshan University (Northern Campus), Foshan 528231, China
| | - Xin Liu
- Department of Food Science, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
29
|
Hypothalamo-pituitary and immune-dependent adrenal regulation during systemic inflammation. Proc Natl Acad Sci U S A 2013; 110:14801-6. [PMID: 23959899 DOI: 10.1073/pnas.1313945110] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Inflammation-related dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis is central to the course of systemic inflammatory response syndrome or sepsis. The underlying mechanisms, however, are not well understood. Initial activation of adrenocortical hormone production during early sepsis depends on the stimulation of hypothalamus and pituitary mediated by cytokines; in late sepsis, there is a shift from neuroendocrine to local immune-adrenal regulation of glucocorticoid production. Therefore, the modulation of the local immune-adrenal cross talk, and not of the neuroendocrine circuits involved in adrenocorticotropic hormone production, may be more promising in the prevention of the adrenal insufficiency associated with prolonged sepsis. In the present work, we investigated the function of the crucial Toll-like receptor (TLR) adaptor protein myeloid differentiation factor 88 (MyD88) in systemic and local activation of adrenal gland inflammation and glucocorticoid production mediated by lipopolysachharides (LPSs). To this end, we used mice with a conditional MyD88 allele. These mice either were interbred with Mx1 Cre mice, resulting in systemic MyD88 deletion, predominantly in the liver and hematopoietic system, or were crossed with Akr1b7 Cre transgenic mice, resulting thereby in deletion of MyD88, which was adrenocortical-specific. Although reduced adrenal inflammation and HPA-axis activation mediated by LPS were found in Mx1(Cre+)-MyD88(fl/fl) mice, adrenocortical-specific MyD88 deletion did not alter the adrenal inflammation or HPA-axis activity under systemic inflammatory response syndrome conditions. Thus, our data suggest an important role of immune cell rather than adrenocortical MyD88 for adrenal inflammation and HPA-axis activation mediated by LPS.
Collapse
|