1
|
Qu L, Cao Y, Wang M, Song D, Huang G. Effects of laughter therapy on improving physical and psychological symptoms among cancer patients: a systematic review and meta-analysis. Support Care Cancer 2025; 33:230. [PMID: 40014214 DOI: 10.1007/s00520-025-09276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
INTRODUCTION Cancer is a major cause of death worldwide and laughter therapy is used as a complementary therapy for cancer treatment. This study aimed to investigate the effects of laughter therapy on psychological symptoms (stress, anxiety, and depression) and physical symptoms (pain, fatigue, and sleep quality) among cancer patients. METHODS Eight databases were searched, with the search period was limited to April 2024. Quality assessment of the included randomized controlled trials was performed according to the criteria for evaluating randomized controlled trials in the Cochrane Handbook for Systematic Reviews of Interventions. The RevMan 5.4 software was used for the data analysis. RESULTS Nine randomized controlled trials were included in this systematic review and meta-analysis. The quality of the included studies was relatively high. The results showed that laughter therapy can effectively improve cancer patients' stress, anxiety, depression, pain, and fatigue, but has no effect on sleep quality. CONCLUSIONS Laughter therapy is a low-cost, easy-to-implement intervention that may have potential benefits in improving psychological symptoms (stress, anxiety, and depression) and physical symptoms (pain and fatigue) among cancer patients. Laughter therapy has the characteristics of high safety, strong interaction, and strong feasibility. It may promote communication between patients and between patients and medical healthcare, enhance the trust relationship between medical staff, and potentially improve the quality of life for cancer patients. Healthcare professionals should consider laughter therapy as a potential adjunctive therapy, but its implementation should be tailored to individual patient needs and supported by robust evidence.
Collapse
Affiliation(s)
- Liying Qu
- Department of Breast Surgery, The Third Bethune Hospital of Jilin University, Changchun, 130033, Jilin, China
| | - Yang Cao
- Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Meng Wang
- Department of Nursing, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, UPM Serdang, 43400, Selangor, Malaysia
| | - Daoqun Song
- Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Guoyun Huang
- Department of Breast Surgery, The Third Bethune Hospital of Jilin University, Changchun, 130033, Jilin, China.
| |
Collapse
|
2
|
Kamsrijai U, Charoensup R, Jaidee W, Hawiset T, Thaweethee-Sukjai B, Praman S. Cannabidiol/cannabidiolic acid-rich hemp (Cannabis sativa L.) extract attenuates cognitive impairments and glial activations in rats exposed to chronic stress. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119113. [PMID: 39551282 DOI: 10.1016/j.jep.2024.119113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hemp (Cannabis sativa L.) is increasingly being recognized for its medicinal properties beside utilizing it for food, oil, and textile fibers. The high level of cannabidiol (CBD) content in hemp's flowers shows promising neuroprotective properties without causing psychotomimetic or addictive effects. Recently, products containing CBD and its precursor, cannabidiolic acid (CBDA), have been used to treat stress-related cognitive impairment. However, the therapeutic potential of hemp extract remains inadequately explored. AIM OF THE STUDY To investigate the effect of CBD/CBDA-rich hemp extract on learning and memory, neuroendocrine alterations, and hippocampal neuropathological changes in the chronic restraint stress model. MATERIALS AND METHODS Chronic restraint stress (CRS) was induced in male Wistar rats by immobilizing them in a restrainer for 6 h per day for 21 consecutive days. CBD/CBDA-rich hemp extract (10 and 30 mg/kg, intraperitoneal injection) was administered daily, 1 h before restraint. After the last day of CRS, behavioral tests for cognition were conducted using the Y-maze and object recognition tests. Serum corticosterone (CORT) levels were measured by ELISA. Histopathological changes, neuronal density, and the activation of microglia and astrocytes were visualized using cresyl violet and immunohistochemical staining. RESULTS A high dose of CBD/CBDA-rich hemp extract effectively ameliorated CRS-induced cognitive impairment and reversed HPA axis hyperactivity in CRS rats by reducing CORT levels and adrenal gland weight. Additionally, CBD/CBDA-rich hemp extract protected CRS-induced damage to hippocampal neurons. Further analysis showed that CBD/CBDA-rich hemp extract reduced specific markers of microglial activation (ionized calcium-binding adaptor molecule-1, Iba-1) and astrocytic structural protein (glial fibrillary acidic protein, GFAP) in CRS rats. CONCLUSION CBD/CBDA-rich hemp extracts remarkably reversed the stress-induced behavioral perturbations and hippocampal damage, suggesting its ameliorative effect on stress response.
Collapse
Affiliation(s)
| | - Rawiwan Charoensup
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai, 57100, Thailand; School of Integrative Medicine, Major of Applied Thai Traditional Medicine, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Wuttichai Jaidee
- Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Thaneeya Hawiset
- School of Medicine, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | | | - Siwaporn Praman
- School of Medicine, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| |
Collapse
|
3
|
Guadagnin AR, Peñagaricano F, Dahl GE, Laporta J. Programming effects of intrauterine hyperthermia on adrenal gland development. J Dairy Sci 2024; 107:6308-6321. [PMID: 38580145 DOI: 10.3168/jds.2023-24606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/27/2024] [Indexed: 04/07/2024]
Abstract
Maternal heat stress during late pregnancy can lead to intrauterine hyperthermia and affect fetal hypothalamic-pituitary-adrenal axis development and function. Herein, we investigated the effects of chronic environmental heat stress exposure of Holstein cows in the last 2 mo of gestation on their offspring's adrenal gland histomorphology and transcriptome. Cows in their last 54 ± 5 d of gestation were either heat stressed (housed under the shade of a freestall barn) or provided heat stress abatement via active cooling (via water soakers and fans) during a subtropical summer (temperature-humidity index >68). Respiration rate (RR) and skin temperature (ST) were elevated in heat-stressed dams relative to the cows with access to heat abatement (23 breaths/min and 2°C higher for RR and ST, respectively). Heifers born to heat-stressed cows experienced heat stress in utero (HS), whereas heifers born to actively cooled cows did not (CL). The adrenal gland was harvested from 6 heifers per group that were euthanized at birth (d 0; n = 12) or 1 wk after weaning (d 63; n = 12). Circulating cortisol was measured from blood samples collected weekly throughout the preweaning period. At d 63, heifers that experienced HS while developing in utero had heavier adrenal glands, with a greater total tissue surface area and thickness of the zona glomerulosa (ZG), fasciculata (ZF), and reticularis (ZR), compared with CL heifers. In addition, the adrenal gland of HS heifers had fewer cells in the ZG, more and larger cells in the ZF, and larger cells in the ZR, relative to CL heifers. Although no changes in circulating cortisol were observed through the preweaning period, the transcriptomic profile of the adrenal tissue was altered by fetal exposure to hyperthermia. Both at birth and on d 63, approximately 30 pathways were differentially expressed in the adrenal glands of HS heifers relative to CL. These pathways were associated with immune function, inflammation, prolactin signaling, cell function, and calcium transport. Upstream regulators significantly activated or inhibited in the adrenal glands of heifers exposed to intrauterine hyperthermia were identified. Maternal exposure to heat stress during late gestation caused an enlargement of their offspring's adrenal glands by inducing ZG and ZF cell hypertrophy, and caused gene expression changes. These phenotypic, histological, and molecular changes in the adrenal gland might lead to alterations in stress, immune, and metabolic responses later in life.
Collapse
Affiliation(s)
- Anne R Guadagnin
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706
| | - Francisco Peñagaricano
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706
| | - Geoffrey E Dahl
- Department of Animal Sciences, University of Florida, Gainesville, FL 32608
| | - Jimena Laporta
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706.
| |
Collapse
|
4
|
Nour SA, Foda DS, Elsehemy IA, Hassan ME. Co-administration of xylo-oligosaccharides produced by immobilized Aspergillus terreus xylanase with carbimazole to mitigate its adverse effects on the adrenal gland. Sci Rep 2024; 14:17481. [PMID: 39080323 PMCID: PMC11289116 DOI: 10.1038/s41598-024-67310-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Carbimazole has disadvantages on different body organs, especially the thyroid gland and, rarely, the adrenal glands. Most studies have not suggested any solution or medication for ameliorating the noxious effects of drugs on the glands. Our study focused on the production of xylooligosaccharide (XOS), which, when coadministered with carbimazole, relieves the toxic effects of the drug on the adrenal glands. In addition to accelerating the regeneration of adrenal gland cells, XOS significantly decreases the oxidative stress caused by obesity. This XOS produced by Aspergillus terreus xylanase was covalently immobilized using microbial Scleroglucan gel beads, which improved the immobilization yield, efficiency, and operational stability. Over a wide pH range (6-7.5), the covalent immobilization of xylanase on scleroglucan increased xylanase activity compared to that of its free form. Additionally, the reaction temperature was increased to 65 °C. However, the immobilized enzyme demonstrated superior thermal stability, sustaining 80.22% of its original activity at 60 °C for 120 min. Additionally, the full activity of the immobilized enzyme was sustained after 12 consecutive cycles, and the activity reached 78.33% after 18 cycles. After 41 days of storage at 4 °C, the immobilized enzyme was still active at approximately 98%. The immobilized enzyme has the capability to produce xylo-oligosaccharides (XOSs). Subsequently, these XOSs can be coadministered alongside carbimazole to mitigate the adverse effects of the drug on the adrenal glands. In addition to accelerating the regeneration of adrenal gland cells, XOS significantly decreases the oxidative stress caused by obesity.
Collapse
Affiliation(s)
- Shaimaa A Nour
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Behouth Street, Cairo, 12622, Egypt
| | - Doaa S Foda
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Behouth Street, Cairo, 12622, Egypt
| | - Islam A Elsehemy
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Behouth Street, Cairo, 12622, Egypt
| | - Mohamed E Hassan
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Behouth Street, Cairo, 12622, Egypt.
- Centre of Excellence, Encapsulation and Nano Biotechnology Group, Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Behouth Street, Cairo, 12622, Egypt.
| |
Collapse
|
5
|
Morsi AA, Mersal EA, Abdelmoneim AM, Hussein G, Sofii MM, Ibrahim KE, Salim MS. Interrogating the estrogen-mediated regulation of adrenocortical Klotho expression using ovariectomized albino rat model exposed to repeated restraint stress. Hum Cell 2024; 37:1008-1023. [PMID: 38753278 DOI: 10.1007/s13577-024-01069-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/19/2024] [Indexed: 06/24/2024]
Abstract
Reproductive aging is associated with altered stress response and many other menopausal symptoms. Little is known about the adrenal expression of the anti-aging protein Klotho or how it is modulated by estrogen in ovariectomized stressed rats. Fifty-six Wistar female rats were assigned into seven equal groups. Sham-operated (Sham), sham stressed (Sham/STS), ovariectomized (OVR), ovariectomized stressed (OVR/STS), ovariectomized stressed rosiglitazone-treated (OVR/STS/R), ovariectomized stressed estrogen-treated (OVR/STS/E), and ovariectomized stressed estrogen/GW9662 co-treated (OVR/STS/E/GW) groups. All stressed rats were subjected daily to a one-hour restraint stress test for 19 days. At the end of the experiment, blood was collected for serum corticosterone (CORT) analysis. Adrenal tissues were obtained and prepared for polymerase chain reaction (PCR) assay, hematoxylin and eosin (H&E), immunohistochemistry-based identification of Klotho and PPAR-γ, and Oil Red O (ORO) staining. The rise in serum CORT was negligible in the OVR/STS group, in contrast to the Sham/STS group. The limited CORT response in the former group was restored by estrogen and rosiglitazone and blocked by estrogen/GW9226 co-administration. ORO-staining revealed a more evident reduction in the adrenal fat in the OVR/STS group, which was reversed by estrogen and counteracted by GW. Also, there was a comparable expression pattern of Klotho and PPAR-γ in the adrenals. The adrenal Klotho decreased in the OVR/STS group, but was reversed by estrogen treatment. GW9226/estrogen co-treatment interfered with the regulatory effect of estrogen on Klotho. The study suggested modulation of the adrenal Kotho expression by estrogen, in the ovariectomized rats subjected to a restraint stress test. This estrogen-provided adrenal protection might be mediated by PPAR-γ activation.
Collapse
Affiliation(s)
- Ahmed A Morsi
- Department of Histology and Cell Biology, Faculty of Medicine, Fayoum University, Fayoum, 63511, Egypt.
| | - Ezat A Mersal
- Biochemistry Department, Faculty of Science, Assiut University, 71515, Assiut, Egypt
| | - Ahmed M Abdelmoneim
- Physiology Department, Faculty of Medicine, Fayoum University, Fayoum, 63511, Egypt
| | - Ghaiath Hussein
- Medical Ethics and Law, Department of Medical Education, School of Medicine, Trinity College Dublin, 152-160 Pearse St, Dublin, D02 R590, Ireland
| | - Mohamed M Sofii
- Department of Anatomy and Embryology, Faculty of Medicine, Fayoum University, Fayoum, 63511, Egypt
| | - Khalid Elfaki Ibrahim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Mohamed S Salim
- Medical Laboratory Technology Department, Higher Technological Institute of Applied Health Sciences, Beni-Suef, Egypt
| |
Collapse
|
6
|
Reimondo G, Solitro F, Puglisi S, Balbi M, Tiranti GM, Perini AME, Cultrera A, Brero D, Botto C, Perotti P, Caramello V, Boccuzzi A, Pia A, Veltri A, Terzolo M. Serendipitous Adrenal Hyperplasia in Patients Admitted to the Emergency Department for Suspected SARS-CoV-2 Infection is Linked to Increased Mortality. Arch Med Res 2024; 55:103010. [PMID: 38805767 DOI: 10.1016/j.arcmed.2024.103010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Few data are available on adrenal morphology in patients with acute diseases, although it is known that endogenous glucocorticoids are essential for survival under stress conditions and that an adequate response is driven by activation of the hypothalamic-pituitary-adrenal (HPA) axis. AIMS The aim of this study was to assess adrenal morphology in patients with acute disease compared with patients with non-acute disease. METHODS This cross-sectional study included: 402 patients admitted to the emergency department (ED) for suspected SARS-CoV-2 infection (March-May, 2020) [main cohort]; 200 patients admitted to the ED for acute conditions (December 2018-February 2019) [control group A]; 200 outpatients who underwent radiological evaluation of non-acute conditions (January-February 2019) [control group B]. Chest and/or abdominal CT scans were reviewed to identify adrenal nodules or hyperplasia. RESULTS In the main cohort, altered adrenal morphology was found in 24.9% of the patients (15.4% adrenal hyperplasia; 9.5% adrenal nodules). The frequency of adrenal hyperplasia was higher both in the main cohort (15.4%) and control group A (15.5%) compared to control group B (8.5%; p = 0.02 and p = 0.03, respectively). In the main cohort, 14.9% patients died within 30 d. According to a multivariate analysis, adrenal hyperplasia was an independent risk factor for mortality (p = 0.04), as were older age (p <0.001) and active cancer (p = 0.01). CONCLUSIONS The notable frequency of adrenal hyperplasia in patients with acute diseases suggests an exaggerated activation of the HPA axis due to stressful conditions. The increased risk of short-term mortality found in patients with adrenal hyperplasia suggests that it may be a possible hallmark of worse prognosis.
Collapse
Affiliation(s)
- Giuseppe Reimondo
- Internal Medicine 1, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Federica Solitro
- Radiology Unit, Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Soraya Puglisi
- Internal Medicine 1, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy.
| | - Maurizio Balbi
- Radiology Unit, Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Giorgio Maria Tiranti
- Radiology Unit, Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Anna Maria Elena Perini
- Internal Medicine 1, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Alessandra Cultrera
- Internal Medicine 1, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Dalila Brero
- Internal Medicine 1, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Cristina Botto
- Internal Medicine 1, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Paola Perotti
- Internal Medicine 1, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | | | - Adriana Boccuzzi
- Emergency Medicine, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Anna Pia
- Internal Medicine 1, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Andrea Veltri
- Radiology Unit, Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Massimo Terzolo
- Internal Medicine 1, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| |
Collapse
|
7
|
Wen TZ, Fu WJ, Xiao SQ, Wang S, Li TR, Chen XY, Chen HY, Luo J, Bian XW, Yao XH. Disorganized adrenocortical zonational structure in COVID-19 patients: Implications of critical illness duration. Pathol Res Pract 2024; 256:155251. [PMID: 38490097 DOI: 10.1016/j.prp.2024.155251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/16/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
Aberrant adrenal function has been frequently reported in COVID-19 patients, but histopathological evidence remains limited. This retrospective autopsy study aims to scrutinize the impact of COVID-19 duration on adrenocortical zonational architecture and peripheral corticosteroid reactivity. The adrenal glands procured from 15 long intensive care unit (ICU)-stay COVID-19 patients, 9 short ICU-stay COVID-19 patients, and 20 matched controls. Subjects who had received glucocorticoid treatment prior to sampling were excluded. Applying hematoxylin and eosin (H&E) and immunohistochemical (IHC) staining, we disclosed that the adrenocortical zonational structure was substantially disorganized in COVID-19 patients, which long ICU-stay patients manifested a higher prevalence of severe disorganization (67%) than short ICU-stay patients (11%; P = 0.0058). The adrenal cortex of COVID-19 patients exhibited a 40% decrease in the zona glomerulosa (ZG) area and a 74% increase in the zona fasciculata (ZF) area (both P < 0.0001) relative to controls. Furthermore, among long ICU-stay COVID-19 patients, the ZG area diminished by 31% (P = 0.0004), and the ZF area expanded by 27% (P = 0.0004) in comparison to short ICU-stay patients. The zona reticularis (ZR) area remained unaltered. Nuclear translocation of corticosteroid receptors in the liver and kidney of long ICU-stay COVID-19 patients was at least 43% lower than in short ICU-stay patients (both P < 0.05). These findings underscore the necessity for clinicians to monitor adrenal function in long-stay COVID-19 patients.
Collapse
Affiliation(s)
- Tian-Zi Wen
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Wen-Juan Fu
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Shi-Qi Xiao
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Shuai Wang
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Tian-Ran Li
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xin-Yu Chen
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - He-Yuan Chen
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Jie Luo
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China.
| | - Xiao-Hong Yao
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China.
| |
Collapse
|
8
|
Cozma D, Siatra P, Bornstein SR, Steenblock C. Sensitivity of the Neuroendocrine Stress Axis in Metabolic Diseases. Horm Metab Res 2024; 56:65-77. [PMID: 38171373 DOI: 10.1055/a-2201-6641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Metabolic diseases are prevalent in modern society and have reached pandemic proportions. Metabolic diseases have systemic effects on the body and can lead to changes in the neuroendocrine stress axis, the critical regulator of the body's stress response. These changes may be attributed to rising insulin levels and the release of adipokines and inflammatory cytokines by adipose tissue, which affect hormone production by the neuroendocrine stress axis. Chronic stress due to inflammation may exacerbate these effects. The increased sensitivity of the neuroendocrine stress axis may be responsible for the development of metabolic syndrome, providing a possible explanation for the high prevalence of severe comorbidities such as heart disease and stroke associated with metabolic disease. In this review, we address current knowledge of the neuroendocrine stress axis in response to metabolic disease and discuss its role in developing metabolic syndrome.
Collapse
Affiliation(s)
- Diana Cozma
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Panagiota Siatra
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland
| | - Charlotte Steenblock
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
9
|
Shawky HA, Abdel Hafez SMN, Hasan NAK, Elbassuoni E, Abdelbaky FAF, AboBakr AHS. Changes in Rat Adrenal Cortex and Pineal Gland in Inverted Light-Dark Cycle: A Biochemical, Histological, and Immunohistochemical Study. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:2037-2052. [PMID: 37738357 DOI: 10.1093/micmic/ozad101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 08/13/2023] [Accepted: 08/25/2023] [Indexed: 09/24/2023]
Abstract
Poor sleep standards are common in everyday life; it is frequently linked to a rise in stress levels. The adrenal gland interacts physiologically with the pineal gland in the stress response. Pineal gland is a small endocrine organ that modulates sleep patterns. This work aimed to evaluate the inverted light-dark cycle rhythm on the histological changes within the adrenal cortex and pineal gland in adult male albino rats. Twenty adult male albino rats were equally divided into two groups: For the first control group, animals were kept on daylight-darkness for 12-12 h. The second group was kept under an inverted 12- to 12-h light-darkness cycle for 4 weeks. Adrenal sections were subjected to biochemical, histological, and immunohistochemical study. Inverted light-dark cycle group recorded a significant elevation of plasma corticosterone, tissue malondialdehyde, tumor necrosis factor-α, and interleukin-1β (IL-1β) associated with a significant reduction of catalase and superoxide dismutase. Adrenal cortex showed biochemical and histological changes. Pineal glands also showed loss of lobular architecture. A significant upregulation in activated inducible nitric oxide synthase (iNOS) and B-cell lymphoma-associated X (Bax) immunohistochemical expression was recorded in adrenal cortex associating with downregulation in B-cell lymphoma 2 (Bcl-2). It could be concluded that subchronic inverted light-dark cycle exerted direct effects on adrenal cortex and the pineal glands.
Collapse
Affiliation(s)
- Heba A Shawky
- Anatomy and Embryology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| | - Sara Mohamed Naguib Abdel Hafez
- Histology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| | - Nabil Abdel Kader Hasan
- Anatomy and Embryology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| | - Eman Elbassuoni
- Physiology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| | - Fatma Alzhraa Fouad Abdelbaky
- Anatomy and Embryology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| | - Abdel Hamid Sayed AboBakr
- Anatomy and Embryology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| |
Collapse
|
10
|
Della Porta M, Maier JA, Cazzola R. Effects of Withania somnifera on Cortisol Levels in Stressed Human Subjects: A Systematic Review. Nutrients 2023; 15:5015. [PMID: 38140274 PMCID: PMC10745833 DOI: 10.3390/nu15245015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Withania somnifera (WS), a popular medicinal plant of the Solanaceae family, contains active ingredients with antioxidant, anti-inflammatory, immunomodulatory, and anti-stress activities. However, its precise mechanisms of action and optimal use as a supplement are not yet fully understood. The objective of this systematic review is to assess the impact of WS supplementation on cortisol levels in stressed humans by analyzing clinical trials conducted prior to May 2023. METHODS The assessment was carried out following the guidelines of Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) by exploring the databases of EMBASE, PubMed, Google Scholar, CENTRAL, and Scopus. RESULTS Of the 4788 articles identified, only 9 studies met the selection criteria. The selected studies varied in terms of design, results, formulations, dosages, and treatment duration (30-112 days), and involved subjects with varying degrees of stress. WS supplementation decreases cortisol secretion with no significant adverse effects. Nonetheless, none of the studies evaluated the potential impact of cortisol reduction on adrenal function and long-term effects. CONCLUSIONS Brief-term supplementation with WS appears to have a stress-reducing effect in stressed individuals. However, since the long-term effects of WS supplementation are not yet fully understood, WS supplements should be used under medical supervision.
Collapse
Affiliation(s)
- Matteo Della Porta
- Department of Biomedical and Clinical Sciences, University of Milano, 20157 Milan, Italy; (J.A.M.); (R.C.)
| | | | | |
Collapse
|
11
|
Wang X, Luo T, Yang Y, Zhou Y, Hou J, Wang P. Unilateral chemical ablation of the adrenal gland lowers blood pressure and alleviates target organ damage in spontaneously hypertensive rats. Hypertens Res 2023; 46:2693-2704. [PMID: 37789113 DOI: 10.1038/s41440-023-01444-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/11/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
Adrenal gland hormones play a critical role in the development and maintenance of hypertension. Adrenal ablation has been used to treat primary aldosteronism but not essential hypertension. The present study aimed to investigate the effectiveness and safety of unilateral adrenal gland ablation in treating spontaneously hypertensive rats (SHR). SHR and Wistar-Kyoto rats (WKY) were subjected to unilateral adrenal ablation with injection of anhydrous ethanol or a sham procedure. Blood pressure was monitored by both tail-cuff plethysmography and telemetry until 6 months after the procedure. Adrenal ablation significantly lowered systolic and diastolic blood pressure of the SHR (P < 0.05 or P < 0.01) but not WKY from 4 to 24 weeks after the procedure. Adrenal ablation substantially damaged adrenal cortex and medulla with fibrosis in SHR and WKY rats. The ablation procedure remarkably reduced the levels of renin, angiotensin II, aldosterone, cortisol, noradrenaline, and epinephrine in SHR (all P < 0.05) but not in WKY. Hypokalemia in SHR was significantly improved by adrenal ablation (P < 0.05), while the serum sodium levels were not affected by adrenal ablation in either SHR or WKY rats. Additionally, adrenal ablation improved cardiac, renal, and vascular remodeling and function measured 3 months after the procedure in SHR. In conclusion, the present study shows that ethanol ablation of adrenal gland can effectively lower blood pressure and prevent target organ damage in SHR. These findings suggest that unilateral debulking of the adrenal gland using ethanol ablation is a promising therapeutic strategy for treating hypertension. METHODS Spontaneously hypertensive rats (SHR) and Wistar-Kyoto rats (WKY) were subjected to unilateral adrenal ablation with injection of ethanol or a sham procedure. Blood pressure was monitored for 24 weeks. RESULTS Adrenal ablation significantly lowered systolic and diastolic blood pressure of SHR but not WKY from 4 to 24 weeks after the procedure. CONCLUSION Unilateral debulking of the adrenal gland using ethanol ablation is a promising therapeutic strategy for treating hypertension.
Collapse
Affiliation(s)
- Xinquan Wang
- Department of Cardiology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
- Key Laboratory of Aging and Vascular Homeostasis of Sichuan Higher Education Institutes, Chengdu, Sichuan, 610500, China
| | - Tao Luo
- Department of Cardiology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
- Key Laboratory of Aging and Vascular Homeostasis of Sichuan Higher Education Institutes, Chengdu, Sichuan, 610500, China
| | - Yi Yang
- Department of Cardiology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
- Key Laboratory of Aging and Vascular Homeostasis of Sichuan Higher Education Institutes, Chengdu, Sichuan, 610500, China
| | - Yaqiong Zhou
- Department of Cardiology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
- Key Laboratory of Aging and Vascular Homeostasis of Sichuan Higher Education Institutes, Chengdu, Sichuan, 610500, China
| | - Jixin Hou
- Department of Cardiology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China.
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China.
- Key Laboratory of Aging and Vascular Homeostasis of Sichuan Higher Education Institutes, Chengdu, Sichuan, 610500, China.
| | - Peijian Wang
- Department of Cardiology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China.
- Sichuan Clinical Research Center for Geriatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China.
- Key Laboratory of Aging and Vascular Homeostasis of Sichuan Higher Education Institutes, Chengdu, Sichuan, 610500, China.
| |
Collapse
|
12
|
Okeowo OM, Oke OO, David GO, Ijomone OM. Caffeine Administration Mitigates Chronic Stress-Induced Behavioral Deficits, Neurochemical Alterations, and Glial Disruptions in Rats. Brain Sci 2023; 13:1663. [PMID: 38137111 PMCID: PMC10741929 DOI: 10.3390/brainsci13121663] [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: 10/17/2023] [Revised: 11/11/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Prolonged exposure to stress has detrimental effects on health, and the consumption of caffeine, mostly contained in energy drinks, has become a widely adopted stress coping strategy. Currently, there is limited information regarding the effects of caffeine intake on chronic stress exposure. Thus, this study investigated the effects of caffeine administration on chronic stress-induced behavioral deficits, neurochemical alterations, and glial disruptions in experimental rats. Thirty male Wistar rats were randomly assigned to five groups (n = 6): non-stress control, stress control, and caffeine groups of doses 12.5, 25, and 50 mg/kg. The stress control and caffeine groups were subjected to an unpredictable chronic mild stress (UCMS) protocol daily for 14 days. The rats were evaluated for phenotypic and neurobehavioral assessments. Thereafter, the rat brains were processed for biochemical and immunohistochemical assays. Caffeine administration was found to ameliorate behavioral dysfunctions in rats exposed to UCMS. The UCMS-induced changes in brain levels of monoamines, cholinesterases, and some oxidative stress biomarkers were reversed by caffeine. Caffeine administration also produced mild protective effects against UCMS-induced changes in GFAP and Iba-1 expression in stress-specific brain regions. These results showed that low and moderate doses of caffeine reversed most of the stress-induced changes, suggesting its ameliorative potential against chronic stress-induced alterations.
Collapse
Affiliation(s)
- Oritoke M. Okeowo
- Department of Physiology, School of Basic Medical Sciences, Federal University of Technology, Akure 340252, Ondo State, Nigeria; (O.M.O.); (O.O.O.)
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo 351101, Ondo State, Nigeria
| | - Olanrewaju O. Oke
- Department of Physiology, School of Basic Medical Sciences, Federal University of Technology, Akure 340252, Ondo State, Nigeria; (O.M.O.); (O.O.O.)
| | - Gloria O. David
- Department of Anatomy, School of Basic Medical Sciences, Federal University of Technology, Akure 340252, Ondo State, Nigeria;
| | - Omamuyovwi M. Ijomone
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo 351101, Ondo State, Nigeria
- Department of Anatomy, School of Basic Medical Sciences, Federal University of Technology, Akure 340252, Ondo State, Nigeria;
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo 351101, Ondo State, Nigeria
| |
Collapse
|
13
|
Zhang M, Yin X, Li W, Zha Y, Zeng X, Zhang X, Cui J, Xue Z, Wang R, Liu C. A radiomics based approach using adrenal gland and periadrenal fat CT images to allocate COVID-19 health care resources fairly. BMC Med Imaging 2023; 23:181. [PMID: 37950171 PMCID: PMC10636917 DOI: 10.1186/s12880-023-01145-9] [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: 01/27/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND The value of radiomics features from the adrenal gland and periadrenal fat CT images for predicting disease progression in patients with COVID-19 has not been studied extensively. We assess the value of radiomics features from the adrenal gland and periadrenal fat CT images in predicting COVID-19 disease exacerbation. METHODS A total of 1,245 patients (685 moderate and 560 severe patients) were enrolled in a retrospective study. We proposed a 3D V-net to segment adrenal glands in onset CT images automatically, and periadrenal fat was obtained using inflation operation around the adrenal gland. Next, we built a clinical model (CM), three radiomics models (adrenal gland model [AM], periadrenal fat model [PM], and fusion of adrenal gland and periadrenal fat model [FM]), and radiomics nomogram (RN) after radiomics features extracted. RESULTS The auto-segmentation framework yielded a dice value 0.79 in the training set. CM, AM, PM, FM, and RN obtained AUCs of 0.717, 0.716, 0.736, 0.760, and 0.833 in the validation set. FM and RN had better predictive efficacy than CM (P < 0.0001) in the training set. RN showed that there was no significant difference in the validation set (mean absolute error [MAE] = 0.04) and test set (MAE = 0.075) between predictive and actual results. Decision curve analysis showed that if the threshold probability was between 0.4 and 0.8 in the validation set or between 0.3 and 0.7 in the test set, it could gain more net benefits using RN than FM and CM. CONCLUSIONS Radiomics features extracted from the adrenal gland and periadrenal fat CT images are related to disease exacerbation in patients with COVID-19.
Collapse
Affiliation(s)
- Mudan Zhang
- Department of Medical Imaging, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, No. 83 Zhongshan East Road, Nan Ming District, 550002, Guiyang, Guiyang, Guizhou Province, China
- School Of Medicine, Guizhou University, 550000, Guiyang, Guizhou province, China
| | - Xuntao Yin
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wuchao Li
- Department of Medical Imaging, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, No. 83 Zhongshan East Road, Nan Ming District, 550002, Guiyang, Guiyang, Guizhou Province, China
- School Of Medicine, Guizhou University, 550000, Guiyang, Guizhou province, China
| | - Yan Zha
- Department of Nephrology, Guizhou Provincial People's Hospital, 550002, Guiyang, Guizhou province, China
| | - Xianchun Zeng
- Department of Medical Imaging, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, No. 83 Zhongshan East Road, Nan Ming District, 550002, Guiyang, Guiyang, Guizhou Province, China
| | - Xiaoyong Zhang
- Department of Medical Imaging, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, No. 83 Zhongshan East Road, Nan Ming District, 550002, Guiyang, Guiyang, Guizhou Province, China
| | - Jingjing Cui
- Shanghai United Imaging Intelligence, Co., Ltd, 201807, Shanghai, China
| | - Zhong Xue
- Shanghai United Imaging Intelligence, Co., Ltd, 201807, Shanghai, China
| | - Rongpin Wang
- Department of Medical Imaging, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, No. 83 Zhongshan East Road, Nan Ming District, 550002, Guiyang, Guiyang, Guizhou Province, China.
- School Of Medicine, Guizhou University, 550000, Guiyang, Guizhou province, China.
| | - Chen Liu
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| |
Collapse
|
14
|
Ayalew M, Deribe B, Hussen S, Defar S, Tesfaye E, Gedefaw A. Insomnia and common mental disorder among patients with pre-existing chronic non-communicable diseases in southern Ethiopia: a survey during COVID-19 pandemic. Front Psychiatry 2023; 14:1142926. [PMID: 37779630 PMCID: PMC10540445 DOI: 10.3389/fpsyt.2023.1142926] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/29/2023] [Indexed: 10/03/2023] Open
Abstract
Background COVID-19 has been causing significant mental health problems and other health-related issues. Despite the fact that COVID-19 has a significant impact on chronic disease patients, there is scant research on insomnia, common mental health disorders (CMD), and their associated factors among chronic disease patients. Objective The purpose of this study was to assess the prevalence of insomnia and common mental disorders (CMD) and their associated factors among patients with pre-existing chronic NCDs in Sidama, southern Ethiopia. Methods A multicenter cross-sectional study was undertaken between June 1 and September 1, 2021. The study included 633 participants. CMD and insomnia were assessed using a 20-item Self-Reported Questionnaire (SRQ-20) and a 7-item Insomnia Severity Index (ISI) scale, respectively. To describe the various variables, descriptive statistics were used. We performed multivariable logistic regression analysis to identify independent factors associated with CMD and insomnia. A value of p < 0.05 was considered statistically significant at a 95% confidence interval. Results The prevalence of insomnia and CMD was found to be 39.3% and 46.8%, respectively. Being merchant (AOR = 0.33; 95% CI = 0.13, 0.82), having a diagnosis of diabetes mellitus (AOR = 1.89; 95% CI = 1.04, 3.46), comorbid diagnosis (AOR = 3.96; 95% CI = 2.27, 6.89), low social support (poor (AOR = 3.37; 95% CI = 1.51, 7.57) and moderate (AOR = 3.13; 95% CI = 1.46, 6.69)), symptoms of insomnia (AOR = 12.08; 95% CI = 7.41, 19.72) and poor quality of life (QOL) (AOR = 1.67; 95% CI = 1.04, 2.72) were independent predictors of CMD. We also found out that, having cardiovascular disorders (CVDs) (AOR = 2.48; 95% CI = 1.18, 5.19), CMD (AOR = 12.09; 95% CI = 7.46, 19.61), and poor QOL (AOR = 2.04; 95% CI = 1.27, 3.26) were significantly associated with insomnia symptoms. Conclusion Our study suggests that substantially high prevalence of CMD and insomnia. Significant association between CMD and occupation, diagnosis, comorbidity, social support, insomnia, and QOL were found. We also revealed that having CVDs, CMD, and poor QOL were significantly associated with insomnia symptoms. Therefore, dealing with the mental health problems of patients with chronic NCDs is an essential component of public health intervention during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Mohammed Ayalew
- School of Nursing, College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
| | - Bedilu Deribe
- School of Nursing, College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
| | - Siraj Hussen
- School of Medical Laboratory, College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
| | - Semira Defar
- Department of Midwifery, College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
| | - Emnet Tesfaye
- Department of Emergency and Critical Care Medicine, School of Medicine, College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
| | - Abel Gedefaw
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine and Health Sciences, Hawassa University, Hawassa, Ethiopia
| |
Collapse
|
15
|
Shahyad S, Sahraei H, Mousallo K, Pirzad Jahromi G, Mohammadi MT. Effect of Papaver rhoeas hydroalcoholic extract on blood corticosterone and psychosocial behaviors in the mice model of predator exposure-induced post-traumatic stress disorder. Heliyon 2023; 9:e18084. [PMID: 37483762 PMCID: PMC10362233 DOI: 10.1016/j.heliyon.2023.e18084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/16/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
The function of hypothalamic-pituitary-adrenal (HPA) axis and psychosocial behaviors are affected in post-traumatic stress disorder (PTSD). Based on presence of several beneficial alkaloids in Papaver rhoeas (PR) plant, we assessed the effects of PR hydroalcoholic extract on blood corticosterone and psychosocial behaviors in the mice model of predator exposure-induced PTSD. Male NMARI mice were assigned into two main groups (control or PTSD) according to stress exposure (presence or absent of the predator). Each main group was divided into four subgroups according to treatment with the different doses of PR extract. Mice were treated intraperitoneally by PR extract at three different doses (1,5&10 mg/kg) 30 min before the beginning of test on days 1, 2&3. Corticosterone concentration determined in the blood samples on days 1, 3&21, and mice examined for the psychosocial behaviors on the third day. PTSD induction in mice by exposing to hungry predator increased blood corticosterone and changed the psychosocial and physiological behaviors. PR extract decreased blood corticosterone in PTSD mice on the third day as well as 21st day. Also, PR extract improved the psychosocial and physiological behaviors in PTSD mice. Moreover, PR extract increased blood corticosterone in control mice at a dose-response manner. PR extract is able to decrease blood corticosterone in PTSD condition and probably prevent the HPA hyperactivity in PTSD mice when exposed to the stress stimuli. Accordingly, decreased blood corticosterone by PR extract might be involved in improvement of the physiological and psychosocial behaviors in PTSD mice.
Collapse
Affiliation(s)
- Shima Shahyad
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hedayat Sahraei
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Kamal Mousallo
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Gila Pirzad Jahromi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Mohammadi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
- Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Czycholl I, Büttner K, Becker D, Schwennen C, Baumgärtner W, Otten W, Wendt M, Puff C, Krieter J. Are biters sick? Health status of tail biters in comparison to control pigs. Porcine Health Manag 2023; 9:19. [PMID: 37161469 PMCID: PMC10170755 DOI: 10.1186/s40813-023-00314-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/27/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Tail biting is a multifactorial problem. As the health status is one of the factors commonly linked to tail biting, this study focuses on the health of identified biters. 30 (obsessive) biters are compared to 30 control animals by clinical and pathological examination as well as blood and cerebrospinal fluid samples. In that way, altogether 174 variables are compared between the groups. Moreover, connections between the variables are analysed. RESULTS In the clinical examination, 6 biters, but only 2 controls (P = 0.019) were noticeably agitated in the evaluation of general behaviour, while 8 controls were noticeably calmer (2 biters, P = 0.02). Biters had a lower body weight (P = 0.0007) and 13 biters had overlong bristles (4 controls, P = 0.008). In the pathological examination, 5 biters, but none of the controls had a hyperceratosis or inflammation of the pars proventricularis of the stomach (P = 0.018). However, 7 controls and only 3 biters were affected by gut inflammation (P = 0.03). In the blood sample, protein and albumin levels were below normal range for biters (protein: 51.6 g/l, albumin: 25.4 g/l), but not for controls (protein: 53.7 g/l, albumin: 27.4 g/l), (protein: P = 0.05, albumin: P = 0.02). Moreover, 14 biters, but only 8 controls had poikilocytosis (P = 0.05). Although not statistically different between groups, many animals (36/60) were affected by hypoproteinemia and hyponatremia as well as by hypokalemia (53/60) and almost all animals (58/60) had hypomagnesemia. For hypomagnesemia, significant connections with variables linked to tail damage and ear necrosis were detected (rs/V/ρ ≥ 0.4, P ≤ 0.05). CONCLUSION The results suggest that behavioural tests might be helpful in identifying biters. Moreover, cornification and inflammation of the pars proventricularis is linked to becoming a biter. Furthermore, the results highlight the need for appropriate and adjusted nutrient and mineral supply, especially with regard to magnesium.
Collapse
Affiliation(s)
- I Czycholl
- Institute of Animal Breeding and Husbandry, Kiel University, 24098, Kiel, Germany.
- Pig Improvement Company (PIC), Hendersonville, TN, 37075, USA.
- Department for Animal Welfare and Disease Control, University of Copenhagen, 1870, Frederiksberg, Denmark.
| | - K Büttner
- Unit for Biomathematics and Data Processing, Faculty of Veterinary Medicine, Justus Liebig University, 35392, Giessen, Germany
| | - D Becker
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - C Schwennen
- Clinic for Swine, Small Ruminants and Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hanover, Foundation, 30173, Hanover, Germany
| | - W Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hanover, Foundation, 30559, Hanover, Germany
| | - W Otten
- Institute of Behavioural Physiology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - M Wendt
- Clinic for Swine, Small Ruminants and Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hanover, Foundation, 30173, Hanover, Germany
| | - C Puff
- Department of Pathology, University of Veterinary Medicine Hanover, Foundation, 30559, Hanover, Germany
| | - J Krieter
- Institute of Animal Breeding and Husbandry, Kiel University, 24098, Kiel, Germany
| |
Collapse
|
17
|
Harfmann D, Florea A. Experimental envenomation with honeybee venom melittin and phospholipase A2 induced multiple ultrastructural changes in adrenocortical mitochondria. Toxicon 2023; 229:107136. [PMID: 37116588 DOI: 10.1016/j.toxicon.2023.107136] [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: 01/27/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
Bee stings represent a public health subject, but the mechanisms involved in bee venom toxicity are not yet fully understood. To evaluate the reactions of adrenocortical cells, through which organisms respond to stress, two honeybee venom components: melittin (Mlt) and phospholipase A2 (PLA2) were tested as potential chemical stressors. Modifications were investigated with transmission electron microscopy and microanalysis. A single dose of Mlt (31 mg/kg) or PLA2 (9.3 mg/kg) was injected in rats of groups ML and PL; daily doses of Mlt (350 μg/kg) or PLA2 (105 μg/kg) were injected 30 days in rats of groups M30 and P30. Adrenocortical cells in ML group showed ultrastructural degenerative alterations of nuclei, endoplasmic reticulum, and mitochondria that exhibited lipid inclusions and mitochondrial cristae (MC) re-organized into mono- or multimembrane large vesicles, and whorls of membranes. Many MC were degenerated. In the M30 group, similar ultrastructural changes, but of lower amplitude were noted; lipid cytosolic droplets were heterogenous. MC diameters in Mlt groups (melittin treated groups) were significantly higher than in control (C) group. In PL group, mitochondria contained large lipid inclusions, vesicular MC of different sizes and multiple membranes, and debris, or whorl structures. In P30 group MC were tubular with increased diameters. In both PLA2 groups (PLA2 treated groups) MC were significantly larger than in C group. We concluded that Mlt and PLA2 were powerful stressors, toxic at the tested doses, cellular reactions concerning in all groups mainly mitochondria, but also other cellular compartments. Apart from degenerative regression of MC, the rearrangement of tubular MC occurred into one or multiple large multimembrane vesicular MC. Reactions to the high doses were more pronounced, with the highest amplitude in ML group, and the lowest in P30 group.
Collapse
Affiliation(s)
- Diana Harfmann
- Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Adrian Florea
- Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| |
Collapse
|
18
|
Wang X, Eguchi A, Fujita Y, Wan X, Chang L, Yang Y, Shan J, Qu Y, Ma L, Shirayama Y, Mori C, Yang J, Hashimoto K. Abnormal compositions of gut microbiota and metabolites are associated with susceptibility versus resilience in rats to inescapable electric stress. J Affect Disord 2023; 331:369-379. [PMID: 36972851 DOI: 10.1016/j.jad.2023.03.073] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/30/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Increasing evidence suggests the role of gut microbiota in resilience versus vulnerability after stress. However, the role of gut microbiota and microbiome-derived metabolites in resilience versus susceptibility in rodents exposed to stress remains unclear. METHODS Adult male rats were exposed to inescapable electric stress under the learned helplessness (LH) paradigm. The composition of gut microbiota and metabolites in the brain and blood from control (no stress) rats, LH resilient rats, and LH susceptible rats were examined. RESULTS At the genus level, the relative abundances of Asaccharobacter, Eisenbergiella, and Klebsiella in LH susceptible rats were significantly higher than that of LH resilient rats. At the species level, the relative abundances of several microbiome were significantly altered between LH susceptible rats and LH resilient rats. Furthermore, there were several metabolites in the brain and blood altered between LH susceptible rats and LH resilient rats. A network analysis showed correlations between the abundance of several microbiome and metabolites in the brain (or blood). LIMITATIONS Detailed roles of microbiome and metabolites are unclear. CONCLUSIONS These findings suggest that abnormal compositions of the gut microbiota and metabolites might contribute to susceptibility versus resilience in rats subjected to inescapable electric foot shock.
Collapse
Affiliation(s)
- Xingming Wang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Akifumi Eguchi
- Department of Sustainable Health Science, Chiba University Center for Preventive Medical Sciences, Chiba 263-8522, Japan
| | - Yuko Fujita
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Xiayun Wan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Lijia Chang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Yong Yang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Jiajing Shan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Youge Qu
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Li Ma
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Yukihiko Shirayama
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Psychiatry, Teikyo University Chiba Medical Center, Ichihara 299-0111, Japan
| | - Chisato Mori
- Department of Sustainable Health Science, Chiba University Center for Preventive Medical Sciences, Chiba 263-8522, Japan; Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| |
Collapse
|
19
|
Frankowska K, Zarobkiewicz M, Sławiński MA, Wawryk-Gawda E, Abramiuk M, Jodłowska-Jędrych B. Changes in the Histological Structure of Adrenal Glands and Corticosterone Level after Whey Protein or Bee Pollen Supplementation in Running and Non-Running Rats. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4105. [PMID: 36901116 PMCID: PMC10002451 DOI: 10.3390/ijerph20054105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 06/18/2023]
Abstract
Due to the many health-promoting properties of bee pollen and whey protein, both products are widely used as dietary supplements. According to these reports on their health-promoting properties, the aim of our study is to assess whether these products can influence the structure and function of the adrenal glands in rats. Thirty male Wistar rats were divided into six equal groups. Among them, there were three groups which included non-running rats and three groups which included running rats. Both of these running (n = 3) and non-running (n = 3) groups included non-supplemented (control groups), bee-pollen-supplemented groups, and whey-protein-supplemented groups. After 8 weeks, the rats were decapitated, their adrenal glands were collected, and paraffin slides were prepared. Then, staining according to the standard H&E and Masson's trichrome protocols was performed. Fecal and urine samples were collected prior to the end of the study to measure corticosterone levels. In the group of non-running rats, the consumption of bee pollen was noted to be significantly higher when compared to the group of running rats (p < 0.05). The thickness of the particular adrenal cortex layers was similar among all of the groups (p > 0.05). The statistically significant changes in the microscopic structure of the adrenal glands, especially regarding cell nuclei diameter and structure, as well as the architecture of sinusoids, were observed between the groups. Moreover, urine corticosterone concentrations were found to vary between all of the analyzed groups (p < 0.05). These results indicate that both bee pollen and whey protein have limited stress-reducing potential.
Collapse
Affiliation(s)
- Karolina Frankowska
- Chair and Department of Histology, Embryology and Cytophysiology, Student Scientific Association, Medical University of Lublin, 20-080 Lublin, Poland
- First Chair and Department of Oncological Gynaecology and Gynaecology, Student Scientific Association, Medical University of Lublin, 20-081 Lublin, Poland
| | - Michał Zarobkiewicz
- Chair and Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Mirosław A. Sławiński
- Chair and Department of Histology, Embryology and Cytophysiology, Medical University of Lublin, 20-080 Lublin, Poland
| | - Ewelina Wawryk-Gawda
- Department of Paediatric Pulmonology and Rheumatology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Monika Abramiuk
- First Chair and Department of Oncological Gynecology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland
| | - Barbara Jodłowska-Jędrych
- Chair and Department of Histology, Embryology and Cytophysiology, Medical University of Lublin, 20-080 Lublin, Poland
| |
Collapse
|
20
|
Relav L, Doghman-Bouguerra M, Ruggiero C, Muzzi JCD, Figueiredo BC, Lalli E. Steroidogenic Factor 1, a Goldilocks Transcription Factor from Adrenocortical Organogenesis to Malignancy. Int J Mol Sci 2023; 24:3585. [PMID: 36835002 PMCID: PMC9959402 DOI: 10.3390/ijms24043585] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Steroidogenic factor-1 (SF-1, also termed Ad4BP; NR5A1 in the official nomenclature) is a nuclear receptor transcription factor that plays a crucial role in the regulation of adrenal and gonadal development, function and maintenance. In addition to its classical role in regulating the expression of P450 steroid hydroxylases and other steroidogenic genes, involvement in other key processes such as cell survival/proliferation and cytoskeleton dynamics have also been highlighted for SF-1. SF-1 has a restricted pattern of expression, being expressed along the hypothalamic-pituitary axis and in steroidogenic organs since the time of their establishment. Reduced SF-1 expression affects proper gonadal and adrenal organogenesis and function. On the other hand, SF-1 overexpression is found in adrenocortical carcinoma and represents a prognostic marker for patients' survival. This review is focused on the current knowledge about SF-1 and the crucial importance of its dosage for adrenal gland development and function, from its involvement in adrenal cortex formation to tumorigenesis. Overall, data converge towards SF-1 being a key player in the complex network of transcriptional regulation within the adrenal gland in a dosage-dependent manner.
Collapse
Affiliation(s)
- Lauriane Relav
- Institut de Pharmacologie Moleculaire et Cellulaire CNRS UMR 7275, 06560 Valbonne, France
- Universite Cote d’Azur, 06560 Valbonne, France
| | - Mabrouka Doghman-Bouguerra
- Institut de Pharmacologie Moleculaire et Cellulaire CNRS UMR 7275, 06560 Valbonne, France
- Universite Cote d’Azur, 06560 Valbonne, France
| | - Carmen Ruggiero
- Institut de Pharmacologie Moleculaire et Cellulaire CNRS UMR 7275, 06560 Valbonne, France
- Universite Cote d’Azur, 06560 Valbonne, France
| | - João C. D. Muzzi
- Laboratório de Imunoquímica (LIMQ), Pós-Graduação em Microbiologia, Parasitologia e Patologia, Departamento de Patologia Básica, Universidade Federal do Paraná (UFPR), Curitiba 81530-990, PR, Brazil
- Laboratório de Bioinformática e Biologia de Sistemas, Pós-Graduação em Bioinformática, Universidade Federal do Paraná (UFPR), Curitiba 81520-260, PR, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Oncology Division, Curitiba 80250-060, PR, Brazil
| | - Bonald C. Figueiredo
- Instituto de Pesquisa Pelé Pequeno Príncipe, Oncology Division, Curitiba 80250-060, PR, Brazil
- Centro de Genética Molecular e Pesquisa do Câncer em Crianças (CEGEMPAC), Molecular Oncology Laboratory, Curitiba 80030-110, PR, Brazil
| | - Enzo Lalli
- Institut de Pharmacologie Moleculaire et Cellulaire CNRS UMR 7275, 06560 Valbonne, France
- Universite Cote d’Azur, 06560 Valbonne, France
- Inserm, 06560 Valbonne, France
| |
Collapse
|
21
|
Feng Y, Lv Y, Feng Q, Song X, Li X, Wang Y. Improvement of stress adaptation and insulin resistance in women with GDM by WeChat group management during novel coronavirus pneumonia. Front Nutr 2023; 9:1017472. [PMID: 36698457 PMCID: PMC9868617 DOI: 10.3389/fnut.2022.1017472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/12/2022] [Indexed: 01/11/2023] Open
Abstract
Aim To evaluate the improvement of glycemic control and stress adaptation in patients with GDM by mobile phone WeChat management during novel coronavirus pneumonia. Methods In this study, 75 women with GDM were included, of whom 35 were included in mobile WeChat group management as the GDM-M group and 40 as the GDM group. Results After mobile WeChat group management for 4 weeks, E and NE were lower. MDA was lower, and SOD was higher. HOMA-IR was lower. E, NE, and cortisol were related to HOMA-IR positively, MDA was positively related to HOMA-IR, and SOD was negatively related to HOMA-IR. E and cortisol were positively related to MDA but negatively related to SOD. Conclusion The stress adaptation disorder and insulin resistance in patients with GDM who have completed mobile WeChat group management can be improved during novel coronavirus pneumonia. Mobile WeChat management played a positive role in improving the insulin resistance of women with GDM under special circumstances, which may reduce the risk of maternal and fetal complications.
Collapse
Affiliation(s)
- Yan Feng
- Department of Clinical Nutrition, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China,*Correspondence: Yan Feng ✉
| | - Yuping Lv
- Department of Medical Oncology, Yan Tai Zhifu Hospital, Yantai, China
| | - Qi Feng
- Department of General Surgery, Xi'an, China
| | - Xinna Song
- Department of Clinical Nutrition, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
| | - Xiaoyan Li
- Department of Obstetrics, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
| | - Yongjun Wang
- Department of Clinical Nutrition, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong, China,Yongjun Wang ✉
| |
Collapse
|
22
|
Bellastella G, Cirillo P, Carbone C, Scappaticcio L, Maio A, Botta G, Tomasuolo M, Longo M, Pontillo A, Bellastella A, Esposito K, De Bellis A. Neuroimmunoendocrinology of SARS-CoV 2 Infection. Biomedicines 2022; 10:2855. [PMID: 36359373 PMCID: PMC9687863 DOI: 10.3390/biomedicines10112855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2023] Open
Abstract
This review is aimed at illustrating and discussing the neuroimmune endocrinological aspects of the SARS-CoV-2 infection in light of the studies on this topic that have so far appeared in the literature. The most characteristic findings and pending controversies were derived by PubMed and Scopus databases. We included original and observational studies, reviews, meta-analysis, and case reports. The entry of the coronavirus into susceptible cells is allowed by the interaction with an ecto-enzyme located on human cells, the angiotensin-converting enzyme 2 (ACE2). SARS-CoV-2 also targets the central nervous system (CNS), including hypothalamic-pituitary structures, as their tissues express ACE2, and ACE2 mRNA expression in hypothalamus and pituitary gland cells has been confirmed in an autoptic study on patients who died of COVID 19. SARS-CoV-2 infection may cause central endocrine disorders in acute phase and in post-COVID period, particularly due to the effects of this virus at CNS level involving the hypothalamic-pituitary axis. The aggression to the hypothalamus-pituitary region may also elicit an autoimmune process involving this axis, responsible consequently for functional disorders of the satellite glands. Adrenal, thyroid and gonadal dysfunctions, as well as pituitary alterations involving GH and prolactin secretions, have so far been reported. However, the extent to which COVID-19 contributes to short- and long-term effects of infection to the endocrine system is currently being discussed and deserves further detailed research.
Collapse
Affiliation(s)
- Giuseppe Bellastella
- Unit of Endocrinology and Metabolic Diseases, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Paolo Cirillo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Carla Carbone
- Unit of Endocrinology and Metabolic Diseases, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Lorenzo Scappaticcio
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Antonietta Maio
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Graziella Botta
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Maria Tomasuolo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Miriam Longo
- Unit of Endocrinology and Metabolic Diseases, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Alessandro Pontillo
- Unit of Endocrinology and Metabolic Diseases, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | | | - Katherine Esposito
- Unit of Endocrinology and Metabolic Diseases, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Annamaria De Bellis
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| |
Collapse
|
23
|
Dasmahapatra AK, Tchounwou PB. Histopathological evaluation of the interrenal gland (adrenal homolog) of Japanese medaka (Oryzias latipes) exposed to graphene oxide. ENVIRONMENTAL TOXICOLOGY 2022; 37:2460-2482. [PMID: 35809259 PMCID: PMC9463118 DOI: 10.1002/tox.23610] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 05/23/2023]
Abstract
Due to unique physicochemical properties and wide industrial and biomedical applications, graphene oxide (GO) is ubiquitous in the aquatic ecosystem. Using Japanese medaka (Oryzias latipes) fish as a model, we previously demonstrated minimal endocrine disrupting (ED) effects of GO on reproductive organs, and thyroids. Current study investigated the ED-effects of GO on the interrenal gland (IRG) of medaka. Breeding pairs of adult male and female fish were exposed to 0 mg/L (control) or 20 mg/L GO by continuous immersion for 96 h, or to 0 or 100 μg/g GO by intraperitoneal administration. Also, 1 day post-hatch (dph) larvae were exposed to different concentrations of GO (2.5-20 mg/L) for 96 h. IRG was evaluated by immunohistochemical techniques after 21 days depuration in adults and 6 weeks in larvae. IRG cells were counted and the nuclear area was measured in hematoxylin-eosin stained sections using ImageJ software. We found that IRG is distributed adjacent to the posterior cardinal vein and its branches within the head kidney. Columnar/oval shaped periodic acid-Schiff negative, tyrosine hydroxylase positive cells are arranged either in a single, or in groups, sometimes encircling a sinusoid, or in a straight chord, laying adjacent to the endothelium of the cardinal vein, and having eosinophilic cytoplasm with round/oval basophilic nuclei. GO effect on nuclei and cell population in IRG was inconsistent; depending on exposure route, sex, and/or age of the fish. Also, because of its high adsorptive property and sharp edges, GO probably agglomerated on IRG, and induced physical injury, and ED effects.
Collapse
Affiliation(s)
- Asok K. Dasmahapatra
- RCMI Center for Environmental HealthJackson State UniversityJacksonMississippiUSA
- Department of Biomolecular Sciences, Environmental Toxicology DivisionUniversity of MississippiOxfordMississippiUSA
| | - Paul B. Tchounwou
- RCMI Center for Environmental HealthJackson State UniversityJacksonMississippiUSA
| |
Collapse
|
24
|
Lim HS, Lee SH, Seo H, Lee HH, Yoon K, Kim YU, Park MK, Chung JH, Lee YS, Lee DH, Park G. Early stage ultraviolet irradiation damage to skin collagen can be suppressed by HPA axis control via controlled CYP11B. Biomed Pharmacother 2022; 155:113716. [PMID: 36162374 DOI: 10.1016/j.biopha.2022.113716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/07/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022] Open
Abstract
UV rays constitute an extremely important environmental factor known to operate adaptative mechanisms that maintain biological homeostasis in the skin, adrenal glands, and the brain. The skin is extremely vulnerable to UV rays. UV rays deform collagen, the main component of elastic fibers, decreasing its normal function, and ultimately reducing skin's elasticity. We confirmed that psychological stress occurring during the early stages of UVB-irradiation degraded collagen function by inhibiting production rather than the decomposition of collagen, thereby promoting skin aging. UV irradiation for 0-2 weeks increased the level of a stress factor, corticosterone (CORT). High-performance liquid chromatography and western blot analysis confirmed that the increase was caused by enhanced CYP11B1/2 levels during steroid synthesis in the adrenal gland. Precursor levels decreased significantly during the two weeks of UV irradiation. Skin collagen and collagen fibers reduced drastically during this time. Furthermore, the administration of osilodrostat, a USFDA-approved drug that selectively inhibits CYP11B1/2, preserved skin collagen. The mechanism underlying the reduction of CORT by osilodrostat confirmed that the amount of skin collagen could be preserved with treatment. In addition, upon suppression of the CORT receptor, the amount of collagen was controlled, and skin aging was suppressed by the hypothalamic-pituitary-adrenal axis. Therefore, this study confirmed an inverse relationship between adrenal CYP11B1/2 levels and collagen during the initial stages of UV irradiation of the skin. The findings of this study may be useful for developing new detection mechanisms for aging, following their further verification.
Collapse
Affiliation(s)
- Hye-Sun Lim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, 111 Geonjae-ro, Naju-si, Jeollanam-do 58245, the Republic of Korea
| | - Seung Hoon Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University School of Medicine, 282 Munhwa-ro, Jung-gu, Daejeon 35015, the Republic of Korea
| | - Huiyun Seo
- Center for Genome Engineering, Institute for Basic Science, 55 Expo-ro, Yuseong-gu, Daejeon 34126, the Republic of Korea
| | - Hwi-Ho Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, the Republic of Korea
| | - Kyeongno Yoon
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, the Republic of Korea
| | - Yong-Ung Kim
- Department of Pharmaceutical Engineering, College of Biomedical Science, Daegu Haany University, 290 Yugok-dong, Gyeongsan-si, Gyeongsangbuk-do 38610, the Republic of Korea
| | - Moon-Ki Park
- Department of Pharmaceutical Engineering, College of Biomedical Science, Daegu Haany University, 290 Yugok-dong, Gyeongsan-si, Gyeongsangbuk-do 38610, the Republic of Korea
| | - Jin Ho Chung
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, the Republic of Korea; Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, the Republic of Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, the Republic of Korea; Institute on Aging, Seoul National University, Seoul 03080, the Republic of Korea
| | - Yong-Seok Lee
- Department of Physiology, Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, the Republic of Korea
| | - Dong Hun Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, the Republic of Korea; Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, the Republic of Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, the Republic of Korea; Institute on Aging, Seoul National University, Seoul 03080, the Republic of Korea
| | - Gunhyuk Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, 111 Geonjae-ro, Naju-si, Jeollanam-do 58245, the Republic of Korea.
| |
Collapse
|
25
|
Anxiolytic Effect of Carvedilol in Chronic Unpredictable Stress Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6906722. [PMID: 36035219 PMCID: PMC9417788 DOI: 10.1155/2022/6906722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 07/13/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022]
Abstract
Anxiety disorders are the most prevalent psychiatric disorders being also a comorbid state of other diseases. We aimed to evaluate the anxiolytic-like effects of carvedilol (CVD), a drug used to treat high blood pressure and heart failure with potent antioxidant effects, in animals exposed to chronic unpredictable stress (CUS). To do this, female Swiss mice were exposed to different stressors for 21 days. Between days 15 and 21, the animals received oral CVD (5 or 10 mg/kg) or the antidepressant desvenlafaxine (DVS 10 mg/kg). On the 22nd day, behavioral tests were conducted to evaluate locomotor activity (open field) and anxiety-like alterations (elevated plus-maze—EPM and hole board—HB tests). After behavioral determinations, the animals were euthanized, and the adrenal gland, blood and brain areas, prefrontal cortex (PFC), and hippocampus were removed for biochemical analysis. CUS reduced the crossings while increased rearing and grooming, an effect reversed by both doses of CVD and DVS. CUS decreased the number of entries and permanence time in the open arms of the EPM, while all treatments reversed this effect. CUS reduced the number of head dips in the HB, an effect reversed by CVD. The CUS reduced weight gain, while only CVD5 reversed this effect. A reduction in the cortical layer size of the adrenal gland was observed in stressed animals, which CVD reversed. Increased myeloperoxidase activity (MPO) and interferon-γ (IFN-γ), as well as reduction of interleukin-4 (IL-4) induced by CUS, were reversed by CVD. DVS and CVD increased IL-6 in both brain areas. In the hippocampus, DVS caused an increase in IFN-γ. Our data show that CVD presents an anxiolytic effect partially associated with immune-inflammatory mechanism regulation.
Collapse
|
26
|
Rodgers KA, Kigerl KA, Schwab JM, Popovich PG. Immune dysfunction after spinal cord injury - A review of autonomic and neuroendocrine mechanisms. Curr Opin Pharmacol 2022; 64:102230. [PMID: 35489214 PMCID: PMC9372819 DOI: 10.1016/j.coph.2022.102230] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 02/05/2023]
Abstract
Infections impair neurological outcome and increase mortality after spinal cord injury (SCI). Emerging data show that pathogens more easily infect individuals with SCI because SCI disrupts neural and humoral control of immune cells, culminating with the development of "SCI-induced immune deficiency syndrome" (SCI-IDS). Here, we review data that implicate autonomic dysfunction and impaired neuroendocrine signaling as key determinants of SCI-IDS. Although it is widely appreciated that mature leukocyte dysfunction is a canonical feature of SCI-IDS, new data indicate that SCI impairs the development and mobilization of immune cell precursors in bone marrow. Thus, this review will also explore how the post-injury acquisition of a "bone marrow failure syndrome" may be the earliest manifestation of SCI-IDS.
Collapse
Affiliation(s)
- Kyleigh A Rodgers
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA
| | - Kristina A Kigerl
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA; The Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA
| | - Jan M Schwab
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA; The Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA; Departments of Neurology and Physical Medicine and Rehabilitation, The Ohio State University, Columbus, OH 43210, USA
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA; The Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
27
|
Khalid W, Maqbool Z, Arshad MS, Kousar S, Akram R, Siddeeg A, Ali A, Qin H, Aziz A, Saeed A, Rahim MA, Zubair Khalid M, Ali H. Plant-derived functional components: prevent from various disorders by regulating the endocrine glands. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2022. [DOI: 10.1080/10942912.2022.2070643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Waseem Khalid
- Department of Food Science, Government College University, Faisalabad, Pakistan
| | - Zahra Maqbool
- Department of Food Science, Government College University, Faisalabad, Pakistan
| | | | - Safura Kousar
- Department of Food Science, Government College University, Faisalabad, Pakistan
| | - Ramish Akram
- Department of Rehabilitation Sciences, The University of Faisalabad, Pakistan
| | - Azhari Siddeeg
- Department of Food Engineering and Technology, Faculty of Engineering and Technology, University of Gezira, Wad Medani, Sudan
| | - Anwar Ali
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, China
| | - Hong Qin
- Department of Nutrition Science and Food Hygiene, Xiangya School of Public Health, Central South University, China
| | - Afifa Aziz
- Department of Food Science, Government College University, Faisalabad, Pakistan
| | - Ayesha Saeed
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | | | | | - Hina Ali
- Department of Botany, Government College University, Faisalabad, Pakistan
| |
Collapse
|
28
|
Neuroinflammation in Post-Traumatic Stress Disorder. Biomedicines 2022; 10:biomedicines10050953. [PMID: 35625690 PMCID: PMC9138406 DOI: 10.3390/biomedicines10050953] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 12/07/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a well-known mental illness, which is caused by various stressors, including memories of past physical assaults and psychological pressure. It is diagnosed as a mental and behavioral disorder, but increasing evidence is linking it to the immune system and inflammatory response. Studies on the relationship between inflammation and PTSD revealed that patients with PTSD had increased levels of inflammatory cytokine biomarkers, such as interleukin-1, interleukin-6, tumor necrosis factor-α, nuclear factor-κB, and C-reactive protein, compared with healthy controls. In addition, animal model experiments imitating PTSD patients suggested the role of inflammation in the pathogenesis and pathophysiology of PTSD. In this review, we summarize the definition of PTSD and its association with increased inflammation, its mechanisms, and future predictable diseases and treatment possibilities. We also discuss anti-inflammatory treatments to address inflammation in PTSD.
Collapse
|
29
|
Vogel F, Reincke M. Endocrine risk factors for COVID-19: Endogenous and exogenous glucocorticoid excess. Rev Endocr Metab Disord 2022; 23:233-250. [PMID: 34241765 PMCID: PMC8267234 DOI: 10.1007/s11154-021-09670-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2021] [Indexed: 01/08/2023]
Abstract
Patients with endogenous or exogenous glucocorticoid (GC) excess exhibit a range of side effects, including an increased risk of infections. Via both mechanism, immune impairments and cardiometabolic concomitant diseases, patients with GC excess could be at increased risk for COVID-19. The impact on incidence and outcome of a SARS-CoV-2 infection in this population are not yet completely clear. This review aims to compile the data available to date and to discuss the existing literature on this topic. Further we highlight potential effects of SARS-CoV-2 on the hypothalamic-pituitary-adrenal axis as well as the influence of endogenous or exogenous GC excess on SARS-CoV-2 mRNA vaccination. There is growing evidence suggesting an increased risk of infection and severe outcome in patients with high-dose GC therapy after contracting SARS-CoV-2. The few data and case reports on patients with endogenous GC excess and SARS-CoV-2 infection point in a similar direction: chronic GC excess seems to be associated with an unfavorable course of COVID-19. Whether this is mainly a primary immune-mediated effect, or also triggered by the many GC-associated comorbidities in this population, is not yet fully understood. Patients with endogenous or exogenous GC excess should be considered as a vulnerable group during the SARS-CoV-2 pandemic. Regardless of the cause, vaccination and consistent surveillance and control of associated comorbidities are recommended.
Collapse
Affiliation(s)
- Frederick Vogel
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
30
|
Corticosterone induces discrete epigenetic signatures in the dorsal and ventral hippocampus that depend upon sex and genotype: focus on methylated Nr3c1 gene. Transl Psychiatry 2022; 12:109. [PMID: 35296634 PMCID: PMC8927334 DOI: 10.1038/s41398-022-01864-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023] Open
Abstract
The genomic effects of circulating glucocorticoids are particularly relevant in cortico-limbic structures, which express a high concentration of steroid hormone receptors. To date, no studies have investigated genomic differences in hippocampal subregions, namely the dorsal (dHPC) and ventral (vHPC) hippocampus, in preclinical models treated with exogenous glucocorticoids. Chronic oral corticosterone (CORT) in mouse is a pharmacological approach that disrupts the activity of the hypothalamic-pituitary-adrenal axis, increases affective behavior, and induces genomic changes after stress in the HPC of wildtype (WT) mice and mice heterozygous for the gene coding for brain-derived neurotrophic factor Val66Met (hMet), a variant associated with genetic susceptibility to stress. Using RNA-sequencing, we investigated the genomic signatures of oral CORT in the dHPC and vHPC of WT and hMet male and female mice, and examined sex and genotype differences in response to oral CORT. Males under CORT showed lower glycemia and increased anxiety- and depression-like behavior compared to females that showed instead opposite affective behavior in response to CORT. Rank-rank-hypergeometric overlap (RRHO) was used to identify genes from a continuous gradient of significancy that were concordant across groups. RRHO showed that CORT-induced differentially expressed genes (DEGs) in WT mice and hMet mice converged in the dHPC of males and females, while in the vHPC, DEGs converged in males and diverged in females. The vHPC showed a higher number of DEGs compared to the dHPC and exhibited sex differences related to glucocorticoid receptor (GR)-binding genes and epigenetic modifiers. Methyl-DNA-immunoprecipitation in the vHPC revealed differential methylation of the exons 1C and 1F of the GR gene (Nr3c1) in hMet females. Together, we report behavioral and endocrinological sex differences in response to CORT, as well as epigenetic signatures that i) differ in the dHPC and vHPC,ii) are distinct in males and females, and iii) implicate differential methylation of Nr3c1 selectively in hMet females.
Collapse
|
31
|
Steenblock C, Schwarz PEH, Perakakis N, Brajshori N, Beqiri P, Bornstein SR. The interface of COVID-19, diabetes, and depression. DISCOVER MENTAL HEALTH 2022; 2:5. [PMID: 35253006 PMCID: PMC8886346 DOI: 10.1007/s44192-022-00007-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/27/2022] [Indexed: 02/07/2023]
Abstract
Comorbid diabetes with depression is a challenging and often under-recognized clinical problem. During the current COVID-19 pandemic, a communicable disease is thriving on the increasing incidences of these non-communicable diseases. These three different health problems are bidirectionally connected forming a vicious cycle. Firstly, depressed individuals show a higher risk of developing diabetes and patients with diabetes have a higher risk of developing symptoms of depression. Secondly, patients with diabetes have a higher risk of developing severe COVID-19 as well as of experiencing breakthrough infections. Thirdly, in both patients with type 2 diabetes and in COVID-19 survivors the prevalence of depression seems to be increased. Fourthly, lockdown and quarantine measurements during the COVID-19 pandemic has led to an increase in depression. Therefore, it is of importance to increase the awareness of this interface between depression, diabetes and COVID-19. Finally, as symptoms of post-COVID, diabetes and depression may be overlapping, there is a need for educating skilled personnel in the management of these comorbidities.
Collapse
Affiliation(s)
- Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Peter E. H. Schwarz
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD E.V.), Neuherberg, Germany
| | - Nikolaos Perakakis
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD E.V.), Neuherberg, Germany
| | - Naime Brajshori
- Research Unit, Heimerer College, Prishtina, Republic of Kosovo
| | - Petrit Beqiri
- Research Unit, Heimerer College, Prishtina, Republic of Kosovo
| | - Stefan R. Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD E.V.), Neuherberg, Germany
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland
- Department of Diabetes, School of Life Course Science and Medicine, Kings College London, London, UK
| |
Collapse
|
32
|
Stress vulnerability shapes disruption of motor cortical neuroplasticity. Transl Psychiatry 2022; 12:91. [PMID: 35246507 PMCID: PMC8897461 DOI: 10.1038/s41398-022-01855-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023] Open
Abstract
Chronic stress is a major cause of neuropsychiatric conditions such as depression. Stress vulnerability varies individually in mice and humans, measured by behavioral changes. In contrast to affective symptoms, motor retardation as a consequence of stress is not well understood. We repeatedly imaged dendritic spines of the motor cortex in Thy1-GFP M mice before and after chronic social defeat stress. Susceptible and resilient phenotypes were discriminated by symptom load and their motor learning abilities were assessed by a gross and fine motor task. Stress phenotypes presented individual short- and long-term changes in the hypothalamic-pituitary-adrenal axis as well as distinct patterns of altered motor learning. Importantly, stress was generally accompanied by a marked reduction of spine density in the motor cortex and spine dynamics depended on the stress phenotype. We found astrogliosis and altered microglia morphology along with increased microglia-neuron interaction in the motor cortex of susceptible mice. In cerebrospinal fluid, proteomic fingerprints link the behavioral changes and structural alterations in the brain to neurodegenerative disorders and dysregulated synaptic homeostasis. Our work emphasizes the importance of synaptic integrity and the risk of neurodegeneration within depression as a threat to brain health.
Collapse
|
33
|
Lu Y, Shi C, Jin X, He J, Yin Z. Domestication of farmed fish via the attenuation of stress responses mediated by the hypothalamus-pituitary-inter-renal endocrine axis. Front Endocrinol (Lausanne) 2022; 13:923475. [PMID: 35937837 PMCID: PMC9353172 DOI: 10.3389/fendo.2022.923475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/29/2022] [Indexed: 12/13/2022] Open
Abstract
Human-directed domestication of terrestrial animals traditionally requires thousands of years for breeding. The most prominent behavioral features of domesticated animals include reduced aggression and enhanced tameness relative to their wild forebears, and such behaviors improve the social tolerance of domestic animals toward both humans and crowds of their own species. These behavioral responses are primarily mediated by the hypothalamic-pituitary-adrenal (inter-renal in fish) (HPA/I) endocrine axis, which is involved in the rapid conversion of neuronal-derived perceptual information into hormonal signals. Over recent decades, growing evidence implicating the attenuation of the HPA/I axis during the domestication of animals have been identified through comprehensive genomic analyses of the paleogenomic datasets of wild progenitors and their domestic congeners. Compared with that of terrestrial animals, domestication of most farmed fish species remains at early stages. The present review focuses on the application of HPI signaling attenuation to accelerate the domestication and genetic breeding of farmed fish. We anticipate that deeper understanding of HPI signaling and its implementation in the domestication of farmed fish will benefit genetic breeding to meet the global demands of the aquaculture industry.
Collapse
Affiliation(s)
- Yao Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Chuang Shi
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Xia Jin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jiangyan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
- Hubei Hongshan Laboratory, Wuhan, China
- Hainan Yazhou Bay Seed Laboratory, Sanya, China
- *Correspondence: Zhan Yin,
| |
Collapse
|
34
|
He X, Ewing AG. Concentration of stimulant regulates initial exocytotic molecular plasticity at single cells. Chem Sci 2022; 13:1815-1822. [PMID: 35282618 PMCID: PMC8826951 DOI: 10.1039/d1sc05278k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/20/2022] [Indexed: 11/21/2022] Open
Abstract
Activity-induced synaptic plasticity has been intensively studied, but is not yet well understood. We examined the temporal and concentration effects of exocytotic molecular plasticity during and immediately after chemical stimulation (30 s K+ stimulation) via single cell amperometry. Here the first and the second 15 s event periods from individual event traces were compared. Remarkably, we found that the amount of catecholamine release and release dynamics depend on the stimulant concentration. No changes were observed at 10 mM K+ stimulation, but changes observed at 30 and 50 mM (i.e., potentiation, increased number of molecules) were opposite to those at 100 mM (i.e., depression, decreased number of events), revealing changes in exocytotic plasticity based on the concentration of the stimulant solution. These results show that molecular changes initiating exocytotic plasticity can be regulated by the concentration strength of the stimulant solution. These different effects on early plasticity offer a possible link between stimulation intensity and synaptic (or adrenal) plasticity. Amperometric measurement of exocytosis (SCA) and vesicle content (IVIEC) over 15 s intervals reveals plasticity (none, potentiation, or depression), that is regulated by the concentration of stimulant solution (e.g., 30 s 10, 30, 50, and 100 mM K+).![]()
Collapse
Affiliation(s)
- Xiulan He
- Department of Chemistry and Molecular Biology, University of Gothenburg 412 96 Gothenburg Sweden
| | - Andrew G Ewing
- Department of Chemistry and Molecular Biology, University of Gothenburg 412 96 Gothenburg Sweden
| |
Collapse
|
35
|
Tseilikman V, Komelkova M, Kondashevskaya MV, Manukhina E, Downey HF, Chereshnev V, Chereshneva M, Platkovskii P, Goryacheva A, Pashkov A, Fedotova J, Tseilikman O, Maltseva N, Cherkasova O, Steenblock C, Bornstein SR, Ettrich B, Chrousos GP, Ullmann E. A Rat Model of Post-Traumatic Stress Syndrome Causes Phenotype-Associated Morphological Changes and Hypofunction of the Adrenal Gland. Int J Mol Sci 2021; 22:ijms222413235. [PMID: 34948031 PMCID: PMC8705403 DOI: 10.3390/ijms222413235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 01/12/2023] Open
Abstract
Background: Rats exposed to chronic predator scent stress mimic the phenotype of complex post-traumatic stress disorder (PTSD) in humans, including altered adrenal morphology and function. High- and low-anxiety phenotypes have been described in rats exposed to predator scent stress (PSS). This study aimed to determine whether these high- and low-anxiety phenotypes correlate with changes in adrenal histomorphology and corticosteroid production. Methods: Rats were exposed to PSS for ten days. Thirty days later, the rats’ anxiety index (AI) was assessed with an elevated plus-maze test. Based on differences in AI, the rats were segregated into low- (AI ≤ 0.8, n = 9) and high- (AI > 0.8, n = 10) anxiety phenotypes. Plasma corticosterone (CORT) concentrations were measured by ELISA. Adrenal CORT, desoxyCORT, and 11-dehydroCORT were measured by high-performance liquid chromatography. After staining with hematoxylin and eosin, adrenal histomorphometric changes were evaluated by measuring the thickness of the functional zones of the adrenal cortex. Results: Decreased plasma CORT concentrations, as well as decreased adrenal CORT, desoxyCORT and 11-dehydroCORT concentrations, were observed in high- but not in low-anxiety phenotypes. These decreases were associated with increases in AI. PSS led to a significant decrease in the thickness of the zona fasciculata and an increase in the thickness of the zona intermedia. The increase in the thickness of the zona intermedia was more pronounced in low-anxiety than in high-anxiety rats. A decrease in the adrenal capsule thickness was observed only in low-anxiety rats. The nucleus diameter of cells in the zona fasciculata of high-anxiety rats was significantly smaller than that of control or low-anxiety rats. Conclusion: Phenotype-associated changes in adrenal function and histomorphology were observed in a rat model of complex post-traumatic stress disorder.
Collapse
Affiliation(s)
- Vadim Tseilikman
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (V.T.); (M.K.); (E.M.); (H.F.D.); (P.P.); (A.P.); (O.T.); (N.M.); (G.P.C.)
| | - Maria Komelkova
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (V.T.); (M.K.); (E.M.); (H.F.D.); (P.P.); (A.P.); (O.T.); (N.M.); (G.P.C.)
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia; (V.C.); (M.C.)
- Faculty of Medicine, Chelyabinsk State University, 454001 Chelyabinsk, Russia
| | - Marina V. Kondashevskaya
- Laboratory for Immunomorphology of Inflammation, Research Institute of Human Morphology, 117418 Moscow, Russia;
| | - Eugenia Manukhina
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (V.T.); (M.K.); (E.M.); (H.F.D.); (P.P.); (A.P.); (O.T.); (N.M.); (G.P.C.)
- Laboratory for Regulatory Mechanisms of Stress and Adaptation, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - H. Fred Downey
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (V.T.); (M.K.); (E.M.); (H.F.D.); (P.P.); (A.P.); (O.T.); (N.M.); (G.P.C.)
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Valerii Chereshnev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia; (V.C.); (M.C.)
| | - Margarita Chereshneva
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia; (V.C.); (M.C.)
| | - Pavel Platkovskii
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (V.T.); (M.K.); (E.M.); (H.F.D.); (P.P.); (A.P.); (O.T.); (N.M.); (G.P.C.)
| | - Anna Goryacheva
- Laboratory for Regulatory Mechanisms of Stress and Adaptation, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
| | - Anton Pashkov
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (V.T.); (M.K.); (E.M.); (H.F.D.); (P.P.); (A.P.); (O.T.); (N.M.); (G.P.C.)
| | - Julia Fedotova
- Laboratory of Neuroendocrinology, I.P. Pavlov Institute of Physiology RAS, 6 Emb. Makarova, 199034 Saint Petersburg, Russia;
- International Research Centre “Biotechnologies of the Third Millennium”, ITMO University, 191002 Saint Petersburg, Russia
| | - Olga Tseilikman
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (V.T.); (M.K.); (E.M.); (H.F.D.); (P.P.); (A.P.); (O.T.); (N.M.); (G.P.C.)
| | - Natalya Maltseva
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (V.T.); (M.K.); (E.M.); (H.F.D.); (P.P.); (A.P.); (O.T.); (N.M.); (G.P.C.)
| | - Olga Cherkasova
- Biophysics Laboratory, Institute of Laser Physics, Siberian Branch of the Russian Academy of Science, 630090 Novosibirsk, Russia;
| | - Charlotte Steenblock
- Department of Medicine, Technical University of Dresden, 01309 Dresden, Germany; (C.S.); (S.R.B.)
| | - Stefan R. Bornstein
- Department of Medicine, Technical University of Dresden, 01309 Dresden, Germany; (C.S.); (S.R.B.)
- Rayne Institute, Division of Diabetes & Nutritional Sciences, Endocrinology and Diabetes, Faculty of Life Sciences & Medicine, Kings College London, London SE5 9PJ, UK
| | - Barbara Ettrich
- Department of Psychiatry and Psychotherapy, University of Leipzig, 04107 Leipzig, Germany;
| | - George P. Chrousos
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (V.T.); (M.K.); (E.M.); (H.F.D.); (P.P.); (A.P.); (O.T.); (N.M.); (G.P.C.)
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Enrico Ullmann
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia; (V.T.); (M.K.); (E.M.); (H.F.D.); (P.P.); (A.P.); (O.T.); (N.M.); (G.P.C.)
- Department of Medicine, Technical University of Dresden, 01309 Dresden, Germany; (C.S.); (S.R.B.)
- Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University of Leipzig, 04107 Leipzig, Germany
- Correspondence:
| |
Collapse
|
36
|
Dado-Senn B, Field SL, Davidson BD, Casarotto LT, Marrero MG, Ouellet V, Cunha F, Sacher MA, Rice CL, Maunsell FP, Dahl GE, Laporta J. Late-Gestation in utero Heat Stress Limits Dairy Heifer Early-Life Growth and Organ Development. FRONTIERS IN ANIMAL SCIENCE 2021. [DOI: 10.3389/fanim.2021.750390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Dairy calves exposed to late-gestation heat stress weigh less, have impaired immunity, produce less milk across multiple lactations, and have reduced productive life. However, less is known about the relationship between in utero heat stress and organ morphology and development. Herein, we characterized the consequences of late-gestation in utero heat stress on body and organ growth trajectories during early-life development. Holstein heifers were either in utero heat-stressed (IU-HT, n = 36, dams exposed to THI > 68) or cooled (IU-CL, n = 37, dams exposed to THI > 68 with access to active cooling) during late gestation (54 ± 5 d prepartum). All heifers were reared identically from birth to weaning. Upon birth, calves were weighed and fed 3.78 L of colostrum followed by 0.87 kg DM/d milk replacer (MR) over two feedings and ad libitum starter concentrate daily. Weaning began at 49 d and ended at 56 d of age. Feed intake was recorded daily, and body weight (BW) and other growth measures were recorded at 0, 28, 56, and 63 d. Blood was collected at d 1 then weekly. Subsets of heifers were selected for euthanasia at birth and 7 d after complete weaning (n = 8 per group each) to harvest and weigh major organs. Reduced BW and stature measures persisted in IU-HT heifers from 0 to 63 d of age with a 7% lower average daily gain and reduced starter consumption relative to IU-CL heifers. IU-HT heifers had lower hematocrit percentages and reduced apparent efficiency of absorption of IgG relative to IU-CL heifers. Additionally, IU-HT heifers had reduced gross thymus, spleen, thyroid gland, and heart weight at birth and larger adrenal glands and kidneys but smaller ovaries relative to BW at 63 d. The mammary gland of IU-HT heifers was smaller relative to IU-CL heifers at birth and 63 d adjusted for BW, suggesting mechanisms leading to impaired milk yield in mature IU-HT cows are initiated early in development. In summary, in utero heat stress reduces whole-body size and limits development of key organs with potential repercussions on dairy calf metabolic adaptation, immune function, and future productivity.
Collapse
|
37
|
Steenblock C, Schwarz PEH, Ludwig B, Linkermann A, Zimmet P, Kulebyakin K, Tkachuk VA, Markov AG, Lehnert H, de Angelis MH, Rietzsch H, Rodionov RN, Khunti K, Hopkins D, Birkenfeld AL, Boehm B, Holt RIG, Skyler JS, DeVries JH, Renard E, Eckel RH, Alberti KGMM, Geloneze B, Chan JC, Mbanya JC, Onyegbutulem HC, Ramachandran A, Basit A, Hassanein M, Bewick G, Spinas GA, Beuschlein F, Landgraf R, Rubino F, Mingrone G, Bornstein SR. COVID-19 and metabolic disease: mechanisms and clinical management. Lancet Diabetes Endocrinol 2021; 9:786-798. [PMID: 34619105 PMCID: PMC8489878 DOI: 10.1016/s2213-8587(21)00244-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/02/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023]
Abstract
Up to 50% of the people who have died from COVID-19 had metabolic and vascular disorders. Notably, there are many direct links between COVID-19 and the metabolic and endocrine systems. Thus, not only are patients with metabolic dysfunction (eg, obesity, hypertension, non-alcoholic fatty liver disease, and diabetes) at an increased risk of developing severe COVID-19 but also infection with SARS-CoV-2 might lead to new-onset diabetes or aggravation of pre-existing metabolic disorders. In this Review, we provide an update on the mechanisms of how metabolic and endocrine disorders might predispose patients to develop severe COVID-19. Additionally, we update the practical recommendations and management of patients with COVID-19 and post-pandemic. Furthermore, we summarise new treatment options for patients with both COVID-19 and diabetes, and highlight current challenges in clinical management.
Collapse
Affiliation(s)
- Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Peter E H Schwarz
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Paul Langerhans Institute Dresden, Helmholtz Center Munich, University Hospital Carl Gustav Carus, Dresden, Germany; German Center for Diabetes Research, Neuherberg, Germany
| | - Barbara Ludwig
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; DFG-Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; Paul Langerhans Institute Dresden, Helmholtz Center Munich, University Hospital Carl Gustav Carus, Dresden, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland
| | - Andreas Linkermann
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Paul Zimmet
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Konstantin Kulebyakin
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia; Institute for Regenerative Medicine, Medical Research and Education Centre, Lomonosov Moscow State University, Moscow, Russia
| | - Vsevolod A Tkachuk
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia; Institute for Regenerative Medicine, Medical Research and Education Centre, Lomonosov Moscow State University, Moscow, Russia
| | - Alexander G Markov
- Department of General Physiology, St Petersburg State University, St Petersburg, Russia
| | | | - Martin Hrabě de Angelis
- German Center for Diabetes Research, Neuherberg, Germany; Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany; School of Life Sciences, Technische Universität München, Freising, Germany
| | - Hannes Rietzsch
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Roman N Rodionov
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kamlesh Khunti
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - David Hopkins
- Department of Diabetes, School of Life Course Science and Medicine, Kings College London, London, UK
| | - Andreas L Birkenfeld
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Department of Diabetes, School of Life Course Science and Medicine, Kings College London, London, UK; Department of Diabetology, Endocrinology and Nephrology, University Hospital Tübingen, Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich, University of Tübingen, Tübingen, Germany; Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | - Bernhard Boehm
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Richard I G Holt
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jay S Skyler
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - J Hans DeVries
- Amsterdam UMC, Internal Medicine, University of Amsterdam, Amsterdam, Netherlands; Profil Institute for Metabolic Research, Neuss, Germany
| | - Eric Renard
- Department of Endocrinology, Diabetes, Nutrition, Montpellier University Hospital, Montpellier, France; Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Robert H Eckel
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Bruno Geloneze
- Obesity and Comorbidities Research Center, Universidade de Campinas, Campinas, Brazil
| | - Juliana C Chan
- Department of Medicine and Therapeutics, Hong Kong Institute of Diabetes and Obesity, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Science, Chinese University of Hong Kong and Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Jean Claude Mbanya
- Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé, Yaounde, Cameroon
| | - Henry C Onyegbutulem
- Endocrine, Diabetes and Metabolic Unit, Department of Internal Medicine, Nile University of Nigeria-Asokoro Hospital, Abuja, Nigeria
| | - Ambady Ramachandran
- India Diabetes Research Foundation, Dr A Ramachandran's Diabetes Hospitals, Chennai, India
| | - Abdul Basit
- Baqai Institute of Diabetology and Endocrinology, Baqai Medical University, Karachi, Pakistan
| | - Mohamed Hassanein
- Dubai Hospital, Dubai Health Authority and Gulf Medical University, Dubai, United Arab Emirates
| | - Gavin Bewick
- Department of Diabetes, School of Life Course Science and Medicine, Kings College London, London, UK
| | - Giatgen A Spinas
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland
| | | | - Francesco Rubino
- Department of Diabetes, School of Life Course Science and Medicine, Kings College London, London, UK; Bariatric and Metabolic Surgery, King's College Hospital, London, UK
| | - Geltrude Mingrone
- Department of Diabetes, School of Life Course Science and Medicine, Kings College London, London, UK; Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Paul Langerhans Institute Dresden, Helmholtz Center Munich, University Hospital Carl Gustav Carus, Dresden, Germany; German Center for Diabetes Research, Neuherberg, Germany; Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland; Department of Diabetes, School of Life Course Science and Medicine, Kings College London, London, UK.
| |
Collapse
|
38
|
Impact of Glucocorticoid on a Cellular Model of Parkinson's Disease: Oxidative Stress and Mitochondrial Function. Brain Sci 2021; 11:brainsci11081106. [PMID: 34439724 PMCID: PMC8391158 DOI: 10.3390/brainsci11081106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 01/21/2023] Open
Abstract
Stress seems to contribute to the neuropathology of Parkinson's disease (PD), possibly by dysregulation of the hypothalamic-pituitary-adrenal axis. Oxidative distress and mitochondrial dysfunction are key factors involved in the pathophysiology of PD and neuronal glucocorticoid-induced toxicity. Animal PD models have been generated to study the effects of hormonal stress, but no in vitro model has yet been developed. Our aim was to examine the impact of corticosterone (CORT) administration on a dopaminergic neuronal cell model of PD induced by the neurotoxin MPP+, as a new combined PD model based on the marker of endocrine response to stress, CORT, and oxidative-mitochondrial damage. We determined the impact of CORT, MPP+ and their co-incubation on reactive oxygen species production (O2-•), oxidative stress cellular markers (advanced-oxidation protein products and total antioxidant status), mitochondrial function (mitochondrial membrane potential and mitochondrial oxygen consumption rate) and neurodegeneration (Fluoro-Jade staining). Accordingly, the administration of MPP+ or CORT individually led to cell damage compared to controls (p < 0.05), as determined by several methods, whereas their co-incubation produced strong cell damage (p < 0.05). The combined model described here could be appropriate for investigating neuropathological hallmarks and for evaluating potential new therapeutic tools for PD patients suffering mild to moderate emotional stress.
Collapse
|
39
|
Lim HS, Yoon KN, Chung JH, Lee YS, Lee DH, Park G. Chronic Ultraviolet Irradiation to the Skin Dysregulates Adrenal Medulla and Dopamine Metabolism In Vivo. Antioxidants (Basel) 2021; 10:antiox10060920. [PMID: 34200115 PMCID: PMC8228565 DOI: 10.3390/antiox10060920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/02/2021] [Accepted: 06/02/2021] [Indexed: 11/16/2022] Open
Abstract
Ultraviolet (UV) radiation has a strong biological effect on skin biology, and it switches on adaptive mechanisms to maintain homeostasis in organs such as the skin, adrenal glands, and brain. In this study, we examined the adaptation of the body to repeated bouts of UVB radiation, especially with respect to the catecholamine synthesis pathway of the adrenal glands. The effects of UVB on catecholamine-related enzymes were determined by neurochemical and histological analyses. To evaluate catecholamine changes after chronic excessive UVB irradiation of mouse skin, we examined dopamine and norepinephrine levels in the adrenal glands and blood from UV-irradiated and sham-irradiated mice. We found that chronic excessive UVB exposure significantly reduced dopamine levels in both tissues but did not affect norepinephrine levels. In addition, UVB irradiation significantly increased the levels of related enzymes tyrosine hydroxylase and dopamine-β-hydroxylase. Furthermore, we also found that apoptosis-associated markers were increased and that oxidative defense proteins were decreased, which might have contributed to the marked structural abnormalities in the adrenal medullas of the chronically UVB-irradiated mice. This is the first evidence of the damage to the adrenal gland and subsequent dysregulation of catecholamine metabolism induced by chronic exposure to UVB.
Collapse
Affiliation(s)
- Hye-Sun Lim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, 111 Geonjae-ro, Naju 58245, Korea;
| | - Kyeong-No Yoon
- Department of Biomedical Sciences, Graduate School, Seoul National University, Seoul 03080, Korea; (K.-N.Y.); (J.H.C.)
| | - Jin Ho Chung
- Department of Biomedical Sciences, Graduate School, Seoul National University, Seoul 03080, Korea; (K.-N.Y.); (J.H.C.)
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea
- Medical Research Center, Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Korea
- Institute on Aging, Seoul National University, Seoul 03080, Korea
| | - Yong-Seok Lee
- Department of Physiology, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea;
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Dong Hun Lee
- Department of Biomedical Sciences, Graduate School, Seoul National University, Seoul 03080, Korea; (K.-N.Y.); (J.H.C.)
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea
- Medical Research Center, Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Korea
- Institute on Aging, Seoul National University, Seoul 03080, Korea
- Correspondence: (D.H.L.); (G.P.); Tel.: +82-2-2072-2415 (D.H.L.); +82-61-338-7112 (G.P.)
| | - Gunhyuk Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, 111 Geonjae-ro, Naju 58245, Korea;
- Correspondence: (D.H.L.); (G.P.); Tel.: +82-2-2072-2415 (D.H.L.); +82-61-338-7112 (G.P.)
| |
Collapse
|
40
|
Dumontet T, Martinez A. Adrenal androgens, adrenarche, and zona reticularis: A human affair? Mol Cell Endocrinol 2021; 528:111239. [PMID: 33676986 DOI: 10.1016/j.mce.2021.111239] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/11/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022]
Abstract
In humans, reticularis cells of the adrenal cortex fuel the production of androgen steroids, constituting the driver of numerous morphological changes during childhood. These steps are considered a precocious stage of sexual maturation and are grouped under the term "adrenarche". This review describes the molecular and enzymatic characteristics of the zona reticularis, along with the possible signals and mechanisms that control its emergence and the associated clinical features. We investigate the differences between species and discuss new studies such as genetic lineage tracing and transcriptomic analysis, highlighting the rodent inner cortex's cellular and molecular heterogeneity. The recent development and characterization of mouse models deficient for Prkar1a presenting with adrenocortical reticularis-like features prompt us to review our vision of the mouse adrenal gland maturation. We expect these new insights will help increase our understanding of the adrenarche process and the pathologies associated with its deregulation.
Collapse
Affiliation(s)
- Typhanie Dumontet
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA; Training Program in Organogenesis, Center for Cell Plasticity and Organ Design, University of Michigan, Ann Arbor, MI, USA.
| | - Antoine Martinez
- Génétique, Reproduction et Développement (GReD), Centre National de La Recherche Scientifique CNRS, Institut National de La Santé & de La Recherche Médicale (INSERM), Université Clermont-Auvergne (UCA), France.
| |
Collapse
|
41
|
Bechmann N, Berger I, Bornstein SR, Steenblock C. Adrenal medulla development and medullary-cortical interactions. Mol Cell Endocrinol 2021; 528:111258. [PMID: 33798635 DOI: 10.1016/j.mce.2021.111258] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/12/2021] [Accepted: 03/22/2021] [Indexed: 01/10/2023]
Abstract
The mammalian adrenal gland is composed of two distinct tissue types in a bidirectional connection, the catecholamine-producing medulla derived from the neural crest and the mesoderm-derived cortex producing steroids. The medulla mainly consists of chromaffin cells derived from multipotent nerve-associated descendants of Schwann cell precursors. Already during adrenal organogenesis, close interactions between cortex and medulla are necessary for proper differentiation and morphogenesis of the gland. Moreover, communication between the cortex and the medulla ensures a regular function of the adult adrenal. In tumor development, interfaces between the two parts are also common. Here, we summarize the development of the mammalian adrenal medulla and the current understanding of the cortical-medullary interactions under development and in health and disease.
Collapse
Affiliation(s)
- Nicole Bechmann
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Ilona Berger
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Diabetes and Nutritional Sciences Division, King's College London, London, UK
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
42
|
Bechmann N, Watts D, Steenblock C, Wallace PW, Schürmann A, Bornstein SR, Wielockx B, Eisenhofer G, Peitzsch M. Adrenal Hormone Interactions and Metabolism: A Single Sample Multi-Omics Approach. Horm Metab Res 2021; 53:326-334. [PMID: 33902135 PMCID: PMC8105089 DOI: 10.1055/a-1440-0278] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The adrenal gland is important for many physiological and pathophysiological processes, but studies are often restricted by limited availability of sample material. Improved methods for sample preparation are needed to facilitate analyses of multiple classes of adrenal metabolites and macromolecules in a single sample. A procedure was developed for preparation of chromaffin cells, mouse adrenals, and human chromaffin tumors that allows for multi-omics analyses of different metabolites and preservation of native proteins. To evaluate the new procedure, aliquots of samples were also prepared using conventional procedures. Metabolites were analyzed by liquid-chromatography with mass spectrometry or electrochemical detection. Metabolite contents of chromaffin cells and tissues analyzed with the new procedure were similar or even higher than with conventional methods. Catecholamine contents were comparable between both procedures. The TCA cycle metabolites, cis-aconitate, isocitate, and α-ketoglutarate were detected at higher concentrations in cells, while in tumor tissue only isocitrate and potentially fumarate were measured at higher contents. In contrast, in a broad untargeted metabolomics approach, a methanol-based preparation procedure of adrenals led to a 1.3-fold higher number of detected metabolites. The established procedure also allows for simultaneous investigation of adrenal hormones and related enzyme activities as well as proteins within a single sample. This novel multi-omics approach not only minimizes the amount of sample required and overcomes problems associated with tissue heterogeneity, but also provides a more complete picture of adrenal function and intra-adrenal interactions than previously possible.
Collapse
Affiliation(s)
- Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, Technische
Universität Dresden, Dresden, Germany
- Department of Medicine III, Technische Universität Dresden,
Dresden, Germany
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of
Experimental Diabetology, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg,
Germany
- Correspondence Dr. Nicole Bechmann Institute of Clinical Chemistry and Laboratory Medicine,University Hospital Carl Gustav Carus Dresden, TechnischeUniversität DresdenFetscherstrasse 7401307 DresdenGermany+ 49 351 45819687+ 49 351
4587346
| | - Deepika Watts
- Institute of Clinical Chemistry and Laboratory Medicine, Technische
Universität Dresden, Dresden, Germany
| | | | - Paal William Wallace
- Institute of Clinical Chemistry and Laboratory Medicine, Technische
Universität Dresden, Dresden, Germany
| | - Annette Schürmann
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of
Experimental Diabetology, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg,
Germany
| | - Stefan R. Bornstein
- Department of Medicine III, Technische Universität Dresden,
Dresden, Germany
| | - Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, Technische
Universität Dresden, Dresden, Germany
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine, Technische
Universität Dresden, Dresden, Germany
- Department of Medicine III, Technische Universität Dresden,
Dresden, Germany
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, Technische
Universität Dresden, Dresden, Germany
| |
Collapse
|
43
|
Lopez JP, Brivio E, Santambrogio A, De Donno C, Kos A, Peters M, Rost N, Czamara D, Brückl TM, Roeh S, Pöhlmann ML, Engelhardt C, Ressle A, Stoffel R, Tontsch A, Villamizar JM, Reincke M, Riester A, Sbiera S, Fassnacht M, Mayberg HS, Craighead WE, Dunlop BW, Nemeroff CB, Schmidt MV, Binder EB, Theis FJ, Beuschlein F, Andoniadou CL, Chen A. Single-cell molecular profiling of all three components of the HPA axis reveals adrenal ABCB1 as a regulator of stress adaptation. SCIENCE ADVANCES 2021; 7:eabe4497. [PMID: 33571131 PMCID: PMC7840126 DOI: 10.1126/sciadv.abe4497] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/09/2020] [Indexed: 05/03/2023]
Abstract
Chronic activation and dysregulation of the neuroendocrine stress response have severe physiological and psychological consequences, including the development of metabolic and stress-related psychiatric disorders. We provide the first unbiased, cell type-specific, molecular characterization of all three components of the hypothalamic-pituitary-adrenal axis, under baseline and chronic stress conditions. Among others, we identified a previously unreported subpopulation of Abcb1b+ cells involved in stress adaptation in the adrenal gland. We validated our findings in a mouse stress model, adrenal tissues from patients with Cushing's syndrome, adrenocortical cell lines, and peripheral cortisol and genotyping data from depressed patients. This extensive dataset provides a valuable resource for researchers and clinicians interested in the organism's nervous and endocrine responses to stress and the interplay between these tissues. Our findings raise the possibility that modulating ABCB1 function may be important in the development of treatment strategies for patients suffering from metabolic and stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Juan Pablo Lopez
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
- The Max Planck Society-Weizmann Institute of Science Laboratory for Experimental Neuropsychiatry and Behavioral Neurogenetics, Rehovot 76100, Israel and Munich, Bavaria 80804, Germany
| | - Elena Brivio
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
- The Max Planck Society-Weizmann Institute of Science Laboratory for Experimental Neuropsychiatry and Behavioral Neurogenetics, Rehovot 76100, Israel and Munich, Bavaria 80804, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Bavaria 80804, Germany
| | - Alice Santambrogio
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE11UL, UK
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Saxony 01307, Germany
| | - Carlo De Donno
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
- The Max Planck Society-Weizmann Institute of Science Laboratory for Experimental Neuropsychiatry and Behavioral Neurogenetics, Rehovot 76100, Israel and Munich, Bavaria 80804, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria 85764, Germany
| | - Aron Kos
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
- The Max Planck Society-Weizmann Institute of Science Laboratory for Experimental Neuropsychiatry and Behavioral Neurogenetics, Rehovot 76100, Israel and Munich, Bavaria 80804, Germany
| | - Miriam Peters
- Department for Endocrinology, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, Munich, Bavaria 80336, Germany
| | - Nicolas Rost
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Bavaria 80804, Germany
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Tanja M Brückl
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Simone Roeh
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Max L Pöhlmann
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Clara Engelhardt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Andrea Ressle
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Rainer Stoffel
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Alina Tontsch
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Javier M Villamizar
- Department for Endocrinology, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, Munich, Bavaria 80336, Germany
| | - Martin Reincke
- Department for Endocrinology, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, Munich, Bavaria 80336, Germany
| | - Anna Riester
- Department for Endocrinology, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, Munich, Bavaria 80336, Germany
| | - Silviu Sbiera
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Bavaria 97080, Germany
| | - Martin Fassnacht
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Bavaria 97080, Germany
| | - Helen S Mayberg
- Departments of Neurology and Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - W Edward Craighead
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Boadie W Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Charles B Nemeroff
- Department of Psychiatry, University of Texas at Austin Dell Medical School, Austin, TX 78738, USA
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Elisabeth B Binder
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Bavaria 85764, Germany
- Department of Mathematics, Technische Universität München, Munich, Bavaria 85748, Germany
| | - Felix Beuschlein
- Department for Endocrinology, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-University, Munich, Bavaria 80336, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zurich 8091, Switzerland
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London SE11UL, UK
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Saxony 01307, Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Bavaria 80804, Germany.
- The Max Planck Society-Weizmann Institute of Science Laboratory for Experimental Neuropsychiatry and Behavioral Neurogenetics, Rehovot 76100, Israel and Munich, Bavaria 80804, Germany
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
44
|
SUBMICROSCOPIC CHANGES OF THE HEMOCAPILLARIES IN THE ADRENAL GLAND CORTEX IN DYNAMICS AFTER EXPERIMENTAL THERMAL INJURY. WORLD OF MEDICINE AND BIOLOGY 2021. [DOI: 10.26724/2079-8334-2021-4-78-224-228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
45
|
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological agent of the coronavirus 2019 (COVID-19) disease, which has caused a pandemic in 2020. This healthcare crisis has had a significant repercussion on the mental health of people who have had the disease itself, those who have undergone restrictive lockdowns, and healthcare professionals who have been working directly or indirectly in patient care. Although the pathological mechanisms related to neuropsychiatric manifestations in patients with COVID-19 are currently unknown, different methods of action through which the central nervous system could be affected have been proposed, including direct or indirect methods. Among them, inflammatory activation through what is known as a "cytokine storm," which is present in both COVID-19 and some mental disorders, seems to play a fundamental role. We also analyze the effects the pandemic has had on the general population, which has had to be remain in lockdown, as well as on healthcare professionals who have been working.
Collapse
Affiliation(s)
- A Rodríguez-Quiroga
- Servicio de Psiquiatría y Salud Mental, Hospital Universitario Infanta Leonor, Madrid, España
- Departamento de Medicina Legal, Psiquiatría y Patología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España
| | - C Buiza
- Servicio de Psiquiatría y Salud Mental, Hospital Universitario Infanta Leonor, Madrid, España
- Departamento de Medicina Legal, Psiquiatría y Patología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España
| | - M A Álvarez de Mon
- Servicio de Psiquiatría y Salud Mental, Hospital Universitario Infanta Leonor, Madrid, España
- Departamento de Medicina Legal, Psiquiatría y Patología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España
| | - J Quintero
- Servicio de Psiquiatría y Salud Mental, Hospital Universitario Infanta Leonor, Madrid, España
- Departamento de Medicina Legal, Psiquiatría y Patología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España
| |
Collapse
|
46
|
Werdermann M, Berger I, Scriba LD, Santambrogio A, Schlinkert P, Brendel H, Morawietz H, Schedl A, Peitzsch M, King AJF, Andoniadou CL, Bornstein SR, Steenblock C. Insulin and obesity transform hypothalamic-pituitary-adrenal axis stemness and function in a hyperactive state. Mol Metab 2020; 43:101112. [PMID: 33157254 PMCID: PMC7691554 DOI: 10.1016/j.molmet.2020.101112] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/22/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022] Open
Abstract
Objective Metabolic diseases are an increasing problem in society with the brain-metabolic axis as a master regulator of the human body for sustaining homeostasis under metabolic stress. However, metabolic inflammation and disease will trigger sustained activation of the hypothalamic-pituitary-adrenal axis. In this study, we investigated the role of metabolic stress on progenitor cells in the hypothalamic-pituitary-adrenal axis. Methods In vitro, we applied insulin and leptin to murine progenitor cells isolated from the pituitary and adrenal cortex and examined the role of these hormones on proliferation and differentiation. In vivo, we investigated two different mouse models of metabolic disease, obesity in leptin-deficient ob/ob mice and obesity achieved via feeding with a high-fat diet. Results Insulin was shown to lead to enhanced proliferation and differentiation of both pituitary and adrenocortical progenitors. No alterations in the progenitors were noted in our chronic metabolic stress models. However, hyperactivation of the hypothalamic-pituitary-adrenal axis was observed and the expression of the appetite-regulating genes Npy and Agrp changed in both the hypothalamus and adrenal. Conclusions It is well-known that chronic stress and stress hormones such as glucocorticoids can induce metabolic changes including obesity and diabetes. In this article, we show for the first time that this might be based on an early sensitization of stem cells of the hypothalamic-pituitary-adrenal axis. Thus, pituitary and adrenal progenitor cells exposed to high levels of insulin are metabolically primed to a hyper-functional state leading to enhanced hormone production. Likewise, obese animals exhibit a hyperactive hypothalamic-pituitary-adrenal axis leading to adrenal hyperplasia. This might explain how stress in early life can increase the risk for developing metabolic syndrome in adulthood. Insulin enhances proliferation and differentiation of adrenocortical and pituitary progenitors. Obesity leads to hyperactivation and priming of the HPA axis. Obesity leads to overexpression of appetite-regulating genes in the hypothalamus. Obesity leads to a decrease in the expression of appetite-regulating genes in the adrenal gland.
Collapse
Affiliation(s)
- Martin Werdermann
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Ilona Berger
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Laura D Scriba
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Alice Santambrogio
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany; Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, SE1 9RT, UK.
| | - Pia Schlinkert
- Department of Pharmacology and Toxicology, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Heike Brendel
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus Dresden, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Andreas Schedl
- University of Côte d'Azur, INSERM, CNRS, iBV, Parc Valrose, Nice, 06108, France.
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| | - Aileen J F King
- Department of Diabetes, School of Life Course Sciences, King's College London, Great Maze Pond, London, SE1 9RT, UK.
| | - Cynthia L Andoniadou
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany; Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, SE1 9RT, UK.
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany; Diabetes and Nutritional Sciences Division, King's College London, Guy's Campus, London, SE1 1UL, UK.
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, Dresden, 01307, Germany.
| |
Collapse
|
47
|
McCormick CM, Smith K, Baumbach JL, de Lima APN, Shaver M, Hodges TE, Marcolin ML, Ismail N. Adolescent social instability stress leads to immediate and lasting sex-specific changes in the neuroendocrine-immune-gut axis in rats. Horm Behav 2020; 126:104845. [PMID: 32846188 DOI: 10.1016/j.yhbeh.2020.104845] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022]
Abstract
Social instability stress (SS; daily 1 h isolation and change of cage partner from postnatal day (P) 30-45) in adolescence produces elevations in corticosterone during the procedure in male and female rats, but no lasting changes in hypothalamic-pituitary-adrenal (HPA) responses to psychological stressors, although deficits in social and cognitive function are evident in adulthood. Here we investigated the effects of SS in corticosterone response to an immune challenge (lipopolysaccharide, LPS, 0.1 mg/kg), on gene expression in the hippocampus, and on gut microbiota, when tested soon- (P46) or long- (P70) after SS. The temporal pattern of corticosterone release after LPS differed between SS and control rats irrespective of the time since SS exposure in females, whereas in males, SS did not alter corticosterone release after LPS. Expression of genes in the hippocampus relevant to immune and HPA function differed between saline-treated SS and control rats depending on sex and time tested, but with lasting consequences of SS in both sexes. LPS-treatment altered hippocampal gene expression, with bigger effects of LPS evident in control than in SS female rats, and the opposite in male rats. Further, effects sometimes depended on the age at time of LPS treatment. SS and control rats differed in both fecal and colon microbiome composition in all but P46 males, and stress history, sex, and age influenced the effects of an immune challenge on the gut microbiome. In sum, adolescent stress history has consequences for immune function into adulthood that may involve effects on the gut microbiome.
Collapse
Affiliation(s)
- Cheryl M McCormick
- Department of Psychology, Brock University, St. Catharines, ON L2S 3A1, Canada; Centre for Neuroscience, Brock University, St. Catharines, ON L2S 3A1, Canada.
| | - Kevin Smith
- Department of Psychology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Jennet L Baumbach
- Department of Psychology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | | | - Madeleine Shaver
- Centre for Neuroscience, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Travis E Hodges
- Department of Psychology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Marina L Marcolin
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Nafissa Ismail
- Department of Psychology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
48
|
Sarapultsev A, Sarapultsev P, Dremencov E, Komelkova M, Tseilikman O, Tseilikman V. Low glucocorticoids in stress-related disorders: the role of inflammation. Stress 2020; 23:651-661. [PMID: 32401103 DOI: 10.1080/10253890.2020.1766020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
There is evidence that plasma cortisol concentration can be either increased or decreased in patients with depression and related anxiety and stress-related disorders; the exact pathophysiological mechanisms of this state are not almost clear. Several distinct theories were proposed and mechanisms, which could lead to decreased glucocorticoid signaling and/or levels, were described. However, there is a possible drawback in almost all the theories proposed: insufficient attention to the inflammatory process, which is undoubtedly present in several stress-related disorders, including post-traumatic stress disorder (PTSD). Previous studies only briefly mentioned the presence of an inflammatory reaction's signs in PTSD, without giving it due importance, although recognizing that it can affect the course of the disease. With that, the state of biochemical changes, characterized by the low glucocorticoids, glucocorticoid receptor's resistance and the signs of the persistent inflammation (with the high levels of circulating cytokines) might be observed not only in PTSD but in coronary heart diseases and systemic chronic inflammatory diseases (rheumatoid arthritis) as well. That is why the present review aims to depict the pathophysiological mechanisms, which lead to a decrease in glucocorticoids in PTSD due to the action of inflammatory stimuli. We described changes in the glucocorticoid system and inflammatory reaction as parts of an integral system, where glucocorticoids and the glucocorticoid receptor reside at the apex of a regulatory network that blocks several inflammatory pathways, while decreased glucocorticoid signaling and/or level leads to unchecked inflammatory reactions to promote pathologies such as PTSD. LAY SUMMARY This review emphasizes the importance of inflammatory reaction in the development of puzzling conditions sometimes observed in severe diseases including post-traumatic stress disorder - the decreased levels of glucocorticoids in the blood. Following the classical concepts, one would expect an increase in glucocorticoid hormones, since they are part of the feedback mechanism in the immune system, which reduces stress and inflammation. However, low levels of glucocorticoid hormones are also observed. Thus, this review describes potential mechanisms, which can lead to the development of such a state.
Collapse
Affiliation(s)
- Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Division of the Russian Academy of Sciences, Ekaterinburg, Russia
| | - Petr Sarapultsev
- Institute of Immunology and Physiology, Ural Division of the Russian Academy of Sciences, Ekaterinburg, Russia
| | - Eliyahu Dremencov
- Institute of Molecular Physiology and Genetics, Centre for Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Maria Komelkova
- Institute of Immunology and Physiology, Ural Division of the Russian Academy of Sciences, Ekaterinburg, Russia
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Olga Tseilikman
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Vadim Tseilikman
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| |
Collapse
|
49
|
Abstract
In a number of adult tissues, Nestin-positive stem cells/progenitors have been identified and shown to be involved in maintenance and remodeling. Various studies have shown that under stressful conditions, quiescent Nestin-positive progenitor cells are activated. Thereby, they migrate to their target location and differentiate into mature cells. In the current paper, we discuss if Nestin-positive progenitors in the hippocampus and adrenal gland belong to unique cell populations that are responsive to stress. Furthermore, we speculate about the mechanism behind their activation and the clinical importance of this stress-response.
Collapse
Affiliation(s)
- Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Diabetes and Nutritional Sciences Division, King's College London, London, UK
| | - Ilona Berger
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
50
|
Di Lorenzo M, Barra T, Rosati L, Valiante S, Capaldo A, De Falco M, Laforgia V. Adrenal gland response to endocrine disrupting chemicals in fishes, amphibians and reptiles: A comparative overview. Gen Comp Endocrinol 2020; 297:113550. [PMID: 32679158 DOI: 10.1016/j.ygcen.2020.113550] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/09/2020] [Indexed: 12/18/2022]
Abstract
The adrenal gland is an essential component of the body stress response; it is formed by two portions: a steroidogenic and a chromaffin tissue. Despite the anatomy of adrenal gland is different among classes of vertebrates, the hormones produced are almost the same. During stress, these hormones contribute to body homeostasis and maintenance of ion balance. The adrenal gland is very sensitive to toxic compounds, many of which behave like endocrine-disruptor chemicals (EDCs). They contribute to alter the endocrine system in wildlife and humans and are considered as possible responsible of the decline of several vertebrate ectotherms. Considering that EDCs regularly can be found in all environmental matrices, the aim of this review is to collect information about the impact of these chemical compounds on the adrenal gland of fishes, amphibians and reptiles. In particular, this review shows the different behavior of these "sentinel species" when they are exposed to stress condition. The data supplied in this review can help to further elucidate the role of EDCs and their harmful impact on the survival of these vertebrates.
Collapse
Affiliation(s)
- Mariana Di Lorenzo
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy.
| | - Teresa Barra
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy
| | - Luigi Rosati
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy
| | - Salvatore Valiante
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy
| | - Anna Capaldo
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy
| | - Maria De Falco
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy; National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| | - Vincenza Laforgia
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy; National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| |
Collapse
|