1
|
Shi XX, Tian XF, He B, Liu SN, Shi CG, Shi Y, Yang YS. Lycium barbarum polysaccharide ameliorates corticosterone-induced cognition decline with modulation of CRHR1. Brain Res Bull 2025; 225:111346. [PMID: 40220965 DOI: 10.1016/j.brainresbull.2025.111346] [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: 01/28/2025] [Revised: 03/18/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Lycium barbarum polysaccharide (LBP) has anti-inflammatory, anti-oxidation and anti-aging properties, but the mechanism of LBP on stress-induced cognitive dysfunction caused by elevated GC level is still unclear. OBJECTIVES Therefore, the present study aimed to investigate the mechanism of LBP on corticosterone-injected(CORT-injected) cognitive impairment. METHODS The rat model was induced by corticosterone in vivo. Water maze test and HE staining were used to observe the effect of LBP on cognitive function and brain morphology in CORT-injected rats. RT-qPCR, Western blot, and immunofluorescence were used to detect the expression of proteins. SH-SY5Y cells were treated with CORT and D-gal in vitro, respectively. The effect of LBP on cell proliferation was observed, and western blotting was detected in the protein expressions. RESULTS In this study, LBP treatment ameliorated CORT-induced learning and cognitive function and protected hypothalamic and hippocampal neurons from injury in vivo. In addition, LBP reduced plasma corticosterone concentrations in CORT-injected rats. The results also indicated that LBP enhanced the expression of synapsis-related proteins PSD95 and SYN by up-regulating the expression of CRHR1 and RGS2 in the hippocampus and hypothalamus of the model group. Meanwhile, we confirmed that LBP enhanced CORT - and D-Gal-induced proliferation of SH-SY5Y cells in vitro, and further verified the expression changes of CRHR1, RGS2, and synapse-related proteins. CONCLUSIONS This study demonstrates that LBP ameliorated CORT-induced cognition decline by regulating CRHR1. Therefore, LBP may represent a potential drug for the prevention of cognition dysfunction in patients caused by increased GCs.
Collapse
Affiliation(s)
- Xiao-Xiao Shi
- Department of Clinical Laboratory, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Feng Tian
- Department of Clinical Laboratory, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bin He
- NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning, Beijing, China
| | - Su-Na Liu
- Department of Neonatal Screening, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cui-Ge Shi
- NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning, Beijing, China.
| | - Ying Shi
- Department of Clinical Laboratory, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Department of Neonatal Screening, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yi-Shu Yang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Cavalhas-Almeida C, Cristo MI, Cavadas C, Ramalho-Santos J, Álvaro AR, Amaral S. Sleep and male (In)Fertility: A comprehensive overview. Sleep Med Rev 2025; 81:102080. [PMID: 40153995 DOI: 10.1016/j.smrv.2025.102080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 01/29/2025] [Accepted: 03/14/2025] [Indexed: 04/01/2025]
Abstract
Infertility affects millions of couples globally, with male factors contributing up to 50 % of cases. Despite the existing Assisted Reproduction Technologies (ART), decreased sperm quality and male infertility persist. Evidence suggests that sleep is crucial for normal reproductive function, and the noticeable increase in infertility rates aligns with the growing prevalence of sleep deprivation, suggesting a connection between both conditions. This literature review explores the possible links between sleep disturbances, with a special emphasis on insomnia, the most prevalent sleep disorder, and male reproductive health. It delves into the latest findings regarding factors such as sperm quality, hormone levels, and overall reproductive function from studies in mammals, both rodents and humans. Through a concise synthesis of these findings, we unveil potential mechanisms and provide crucial insights for clinicians and researchers in the field of reproductive health.
Collapse
Affiliation(s)
- Catarina Cavalhas-Almeida
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Maria Inês Cristo
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; University of Coimbra, Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Portugal
| | - Cláudia Cavadas
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - João Ramalho-Santos
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Ana Rita Álvaro
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - Sandra Amaral
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
3
|
Guo L, Lee HK, Oh S, Koirala GR, Kim TI. Smart Bioelectronics for Real-Time Diagnosis and Therapy of Body Organ Functions. ACS Sens 2025; 10:3239-3273. [PMID: 40310273 DOI: 10.1021/acssensors.5c00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Noncommunicable diseases (NCDs) associated with cardiovascular, neurological, and gastrointestinal disorders remain a leading cause of global mortality, sounding the alarm for the urgent need for better diagnostic and therapeutic solutions. Wearable and implantable biointegrated electronics offer a groundbreaking solution, combining real-time, high-resolution monitoring with innovative treatment capabilities tailored to specific organ functions. In this comprehensive review, we focus on the diseases affecting the brain, heart, gastrointestinal organs, bladder, and adrenal gland, along with their associated physiological parameters. Additionally, we provide an overview of the characteristics of these parameters and explore the potential of bioelectronic devices for in situ sensing and therapeutic applications and highlight the recent advancements in their deployment across specific organs. Finally, we analyze the current challenges and prospects of implementing closed-loop feedback control systems in integrated sensor-therapy applications. By emphasizing organ-specific applications and advocating for closed-loop systems, this review highlights the potential of future bioelectronics to address physiological needs and serves as a guide for researchers navigating the interdisciplinary fields of diagnostics, therapeutics, and personalized medicine.
Collapse
Affiliation(s)
- Lili Guo
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Hin Kiu Lee
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Suyoun Oh
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Gyan Raj Koirala
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Tae-Il Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| |
Collapse
|
4
|
Stergianos S, Spelman T, Eriksson D, Öster S, Björnsdottir S, Kämpe O, Skov J, Bensing S. Increased risk of osteoporotic fractures and osteoporosis in patients with Addison's disease in Sweden: A nationwide population-based cohort study. J Intern Med 2025; 297:518-531. [PMID: 40190018 PMCID: PMC12033000 DOI: 10.1111/joim.20085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
BACKGROUND The risk of major osteoporotic fractures (MOFs) and osteoporosis in patients with autoimmune Addison's disease (AAD) is unclear. OBJECTIVE To investigate the risk of MOF in patients with AAD and the possible correlation with adrenal hormone replacement doses. METHODS Swedish national health registers were used to identify 1869 subjects with AAD and 16,844 matched controls. The primary outcome was MOF, and the secondary outcome was treatment with osteoporosis medications. Marginal Cox models were used to compare time-to-event outcomes. The study period spanned from 1 July 2005 until 31 December 2020. Individuals at risk were followed from inclusion until censored or the end of the study period. RESULTS A total of 77 patients with AAD (7.1/1000 person-years [PY]), and 387 matched controls (3.9/1000 PY) were diagnosed with MOF. The risk of MOF was higher in patients with AAD compared to matched controls, with an adjusted hazard ratio (aHR) of 1.82 (95% confidence interval [CI], 1.41-2.35) and increased in both male and female patients, with aHR of 2.51 (95% CI, 1.56-4.02) and 1.65 (95% CI, 1.22-2.24), respectively. Patients with AAD had an increased risk of treatment with osteoporosis medications: aHR 3.25 (95% CI, 2.71-3.99), compared to controls. No significant differences in MOF rates were observed between patients treated with intermediate or high doses of glucocorticoids compared to low doses (p = 0.967 and p = 0.580, respectively). Similarly, stratification by mineralocorticoid dose (<0.10 vs. ≥0.10 mg/day) showed no significant association regarding MOF (p = 0.915). CONCLUSIONS The risk of MOF is increased in patients with AAD without any apparent correlation to adrenal hormone replacement doses.
Collapse
Affiliation(s)
- Stavros Stergianos
- Department of EndocrinologyKarolinska University HospitalStockholmSweden
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
| | - Tim Spelman
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Daniel Eriksson
- Department of Medicine (Solna)Center for Molecular MedicineKarolinska InstitutetSweden
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| | - Sara Öster
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
| | | | - Olle Kämpe
- Department of EndocrinologyKarolinska University HospitalStockholmSweden
- Department of Medicine (Solna)Center for Molecular MedicineKarolinska InstitutetSweden
| | - Jakob Skov
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
- Department of MedicineKarlstad Central HospitalKarlstadSweden
| | - Sophie Bensing
- Department of EndocrinologyKarolinska University HospitalStockholmSweden
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
| |
Collapse
|
5
|
Huang Q, Hu B, Zhang P, Yuan Y, Yue S, Chen X, Liang J, Tang Z, Zhang B. Neuroscience of cancer: unraveling the complex interplay between the nervous system, the tumor and the tumor immune microenvironment. Mol Cancer 2025; 24:24. [PMID: 39825376 PMCID: PMC11740516 DOI: 10.1186/s12943-024-02219-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025] Open
Abstract
The study of the multifaceted interactions between neuroscience and cancer is an emerging field with significant implications for understanding tumor biology and the innovation in therapeutic approaches. Increasing evidence suggests that neurological functions are connected with tumorigenesis. In particular, the peripheral and central nervous systems, synapse, neurotransmitters, and neurotrophins affect tumor progression and metastasis through various regulatory approaches and the tumor immune microenvironment. In this review, we summarized the neurological functions that affect tumorigenesis and metastasis, which are controlled by the central and peripheral nervous systems. We also explored the roles of neurotransmitters and neurotrophins in cancer progression. Moreover, we examined the interplay between the nervous system and the tumor immune microenvironment. We have also identified drugs that target the nervous system for cancer treatment. In this review we present the work supporting that therapeutic agent targeting the nervous system could have significant potential to improve cancer therapy.
Collapse
Affiliation(s)
- Qibo Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China
| | - Bai Hu
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ye Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Shiwei Yue
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China.
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China.
| |
Collapse
|
6
|
Stalder T, Oster H, Abelson JL, Huthsteiner K, Klucken T, Clow A. The Cortisol Awakening Response: Regulation and Functional Significance. Endocr Rev 2025; 46:43-59. [PMID: 39177247 DOI: 10.1210/endrev/bnae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/21/2024] [Accepted: 08/21/2024] [Indexed: 08/24/2024]
Abstract
In healthy individuals, the majority of cortisol secretion occurs within several hours surrounding morning awakening. A highly studied component of this secretory period is the cortisol awakening response (CAR), the rapid increase in cortisol levels across the first 30 to 45 minutes after morning awakening. This strong cortisol burst at the start of the active phase has been proposed to be functional in preparing the organism for the challenges of the upcoming day. Here, we review evidence on key regulatory and functional processes of the CAR and develop an integrative model of its functional role. Specifically, we propose that, in healthy individuals, the CAR is closely regulated by an intricate dual-control system, which draws upon key circadian, environmental, and neurocognitive processes to best predict the daily need for cortisol-related action. Fine-tuned CAR expression, in turn, is then assumed to induce potent glucocorticoid action via rapid nongenomic and slower genomic pathways (eg, affecting circadian clock gene expression) to support and modulate daily activity through relevant metabolic, immunological, and neurocognitive systems. We propose that this concerted action is adaptive in mediating two main functions: a primary process to mobilize resources to meet activity-related demands and a secondary process to help the organism counterregulate adverse prior-day emotional experiences.
Collapse
Affiliation(s)
- Tobias Stalder
- Department of Psychology, University of Siegen, 57076 Siegen, Germany
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, 23562 Lübeck, Germany
| | - James L Abelson
- Department of Psychiatry, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Tim Klucken
- Department of Psychology, University of Siegen, 57076 Siegen, Germany
| | - Angela Clow
- Department of Psychology, University of Westminster, London W1W 6UW, UK
| |
Collapse
|
7
|
Li C, Lu T, Pan C, Hu C. Glucocorticoids Selectively Inhibit Hippocampal CA1 Pyramidal Neurons Activity Through HCN Channels. Int J Mol Sci 2024; 25:11971. [PMID: 39596039 PMCID: PMC11593447 DOI: 10.3390/ijms252211971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Glucocorticoids are known to influence hippocampal function, but their rapid non-genomic effects on specific neurons in the hippocampal trisynaptic circuit remain underexplored. This study investigated the immediate effects of glucocorticoids on CA1 and CA3 pyramidal neurons, and dentate gyrus (DG) granule neurons in rats using the patch-clamp technique. We found that a 5 min extracellular application of corticosterone significantly reduced action potential firing frequency in CA1 pyramidal neurons, while no effects were observed in CA3 or DG neurons. The corticosterone-induced inhibition in CA1 was blocked by the glucocorticoid receptor antagonist CORT125281, but remained unaffected by the mineralocorticoid receptor antagonist spironolactone. Notably, membrane-impermeable bovine serum albumin-conjugated dexamethasone mimicked corticosterone's effects on CA1 neurons, which exhibited prominent hyperpolarization-activated cyclic nucleotide-gated (HCN) channel currents. Pyramidal neurons in CA3 and granular neurons in the DG showed little HCN channel currents. Corticosterone enhanced HCN channel activity in CA1 neurons via glucocorticoid receptors, and the HCN channel inhibitor ZD7288 abolished corticosterone's suppressive effects on action potentials. These findings suggest that glucocorticoids selectively inhibit CA1 pyramidal neuron activity through HCN channels, providing new insight into the mechanisms of glucocorticoid action in hippocampal circuits.
Collapse
Affiliation(s)
- Chenyang Li
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
- International Human Phenome Institute (Shanghai), Shanghai 200433, China
| | - Tongchuang Lu
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
- International Human Phenome Institute (Shanghai), Shanghai 200433, China
| | - Chengfang Pan
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
- International Human Phenome Institute (Shanghai), Shanghai 200433, China
| | - Changlong Hu
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, China
- International Human Phenome Institute (Shanghai), Shanghai 200433, China
| |
Collapse
|
8
|
Wan D, Lu T, Li C, Hu C. Glucocorticoids Rapidly Modulate Ca V1.2-Mediated Calcium Signals through Kv2.1 Channel Clusters in Hippocampal Neurons. J Neurosci 2024; 44:e0179242024. [PMID: 39299804 PMCID: PMC11551909 DOI: 10.1523/jneurosci.0179-24.2024] [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: 01/25/2024] [Revised: 08/15/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
The precise regulation of Ca2+ signals plays a crucial role in the physiological functions of neurons. Here, we investigated the rapid effect of glucocorticoids on Ca2+ signals in cultured hippocampal neurons from both female and male rats. In cultured hippocampal neurons, glucocorticoids inhibited the spontaneous somatic Ca2+ spikes generated by Kv2.1-organized Ca2+ microdomains. Furthermore, glucocorticoids rapidly reduced the cell surface expressions of Kv2.1 and CaV1.2 channels in hippocampal neurons. In HEK293 cells transfected with Kv2.1 alone, glucocorticoids significantly reduced the surface expression of Kv2.1 with little effect on K+ currents. In HEK293 cells transfected with CaV1.2 alone, glucocorticoids inhibited CaV1.2 currents but had no effect on the cell surface expression of CaV1.2. Notably, in the presence of wild-type Kv2.1, glucocorticoids caused a decrease in the surface expression of CaV1.2 channels in HEK293 cells. However, this effect was not observed in the presence of nonclustering Kv2.1S586A mutant channels. Live-cell imaging showed that glucocorticoids rapidly decreased Kv2.1 clusters on the plasma membrane. Correspondingly, Western blot results indicated a significant increase in the cytoplasmic level of Kv2.1, suggesting the endocytosis of Kv2.1 clusters. Glucocorticoids rapidly decreased the intracellular cAMP concentration and the phosphorylation level of PKA in hippocampal neurons. The PKA inhibitor H89 mimicked the effect of glucocorticoids on Kv2.1, while the PKA agonist forskolin abrogated the effect. In conclusion, glucocorticoids rapidly suppress CaV1.2-mediated Ca2+ signals in hippocampal neurons by promoting the endocytosis of Kv2.1 channel clusters through reducing PKA activity.
Collapse
Affiliation(s)
- Di Wan
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| | - Tongchuang Lu
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| | - Chenyang Li
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| | - Changlong Hu
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China,
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| |
Collapse
|
9
|
Lee JH, Meyer EJ, Nenke MA, Lightman SL, Torpy DJ. Cortisol, Stress, and Disease-Bidirectional Associations; Role for Corticosteroid-Binding Globulin? J Clin Endocrinol Metab 2024; 109:2161-2172. [PMID: 38941154 DOI: 10.1210/clinem/dgae412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Indexed: 06/30/2024]
Abstract
Selye described stress as a unified neurohormonal mechanism maintaining homeostasis. Acute stress system activation is adaptive through neurocognitive, catecholaminergic, and immunomodulation mechanisms, followed by a reset via cortisol. Stress system components, the sympathoadrenomedullary system, hypothalamic-pituitary-adrenal axis, and limbic structures are implicated in many chronic diseases by establishing an altered homeostatic state, allostasis. Consequent "primary stress system disorders" were popularly accepted, with phenotypes based on conditions such as Cushing syndrome, pheochromocytoma, and adrenal insufficiency. Cardiometabolic and major depressive disorders are candidates for hypercortisolemic etiology, contrasting the "hypocortisolemic symptom triad" of stress sensitivity, chronic fatigue, and pain. However, acceptance of chronic stress etiology requires cause-and-effect associations, and practical utility such as therapeutics altering stress system function. Inherent predispositions to stress system perturbations may be relevant. Glucocorticoid receptor (GR) variants have been associated with metabolic/neuropsychological states. The SERPINA6 gene encoding corticosteroid-binding globulin (CBG), was the sole genetic factor in a single-nucleotide variation-genome-wide association study linkage study of morning plasma cortisol, a risk factor for cardiovascular disease, with alterations in tissue-specific GR-related gene expression. Studies showed genetically predicted high cortisol concentrations are associated with hypertension and anxiety, and low CBG concentrations/binding affinity, with the hypocortisolemic triad. Acquired CBG deficiency in septic shock results in 3-fold higher mortality when hydrocortisone administration produces equivocal results, consistent with CBG's role in spatiotemporal cortisol delivery. We propose some stress system disorders result from constitutional stress system variants rather than stressors themselves. Altered CBG:cortisol buffering may influence interstitial cortisol ultradian surges leading to pathological tissue effects, an example of stress system variants contributing to stress-related disorders.
Collapse
Affiliation(s)
- Jessica H Lee
- Department of Medicine, Adelaide University, Adelaide, SA 5000, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Emily Jane Meyer
- Department of Medicine, Adelaide University, Adelaide, SA 5000, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Endocrine and Diabetes Services, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Marni Anne Nenke
- Department of Medicine, Adelaide University, Adelaide, SA 5000, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Endocrine and Diabetes Services, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Stafford L Lightman
- Systems Neuroendocrinology Research Group, University of Bristol, Bristol, BS1 3NY, UK
| | - David J Torpy
- Department of Medicine, Adelaide University, Adelaide, SA 5000, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| |
Collapse
|
10
|
Lightman SL, Conway-Campbell BL. Circadian and ultradian rhythms: Clinical implications. J Intern Med 2024; 296:121-138. [PMID: 38825772 DOI: 10.1111/joim.13795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The hypothalamic-pituitary-adrenal axis is an extremely dynamic system with a combination of both circadian and ultradian oscillations. This state of 'continuous dynamic equilibration' provides a platform that is able to anticipate events, is sensitive in its response to stressors, remains robust during perturbations of both the internal and external environments and shows plasticity to adapt to a changed environment. In this review, we describe these oscillations of glucocorticoid (GC) hormones and why they are so important for GC-dependent gene activation in the brain and liver, and their consequent effects on the regulation of synaptic and memory function as well as appetite control and metabolic regulation. Abnormalities of mood, appetite and metabolic regulation are well-known consequences of GC therapy, and we suggest that the pattern of GC treatment and hormone replacement should be a much higher priority for endocrinologists and the pharmaceutical industry. One of the major impediments to our research on the importance of these cortisol rhythms in our patients has been our inability to measure repeated levels of hormones across the day in patients in their home or work surroundings. We describe how new wearable methodologies now allow the measurement of 24-h cortisol profiles - including during sleep - and will enable us to define physiological normality and allow us both to develop better diagnostic tests and inform, at an individual patient level, how to improve replacement therapy.
Collapse
Affiliation(s)
- Stafford L Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Becky L Conway-Campbell
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
11
|
Ilkevič E, Hausmann M, Grikšienė R. Emotion recognition and regulation in males: Role of sex and stress steroids. Front Neuroendocrinol 2024; 74:101145. [PMID: 38862092 DOI: 10.1016/j.yfrne.2024.101145] [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: 11/23/2023] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/13/2024]
Abstract
Understanding emotions in males is crucial given their higher susceptibility to substance use, interpersonal violence, and suicide compared to females. Steroid hormones are assumed to be critical biological factors that affect and modulate emotion-related behaviors, together with psychological and social factors. This review explores whether males' abilities to recognize emotions of others and regulate their own emotions are associated with testosterone, cortisol, and their interaction. Higher levels of testosterone were associated with improved recognition and heightened sensitivity to threatening faces. In contrast, higher cortisol levels positively impacted emotion regulation ability. Indirect evidence from neuroimaging research suggested a link between higher testosterone levels and difficulties in cognitive emotion regulation. However, this notion must be investigated in future studies using different emotion regulation strategies and considering social status. The present review contributes to the understanding of how testosterone and cortisol affect psychological well-being and emotional behavior in males.
Collapse
Affiliation(s)
- Erik Ilkevič
- Department of Neurobiology and Biophysics, Life Science Center, Vilnius University, Lithuania
| | | | - Ramunė Grikšienė
- Department of Neurobiology and Biophysics, Life Science Center, Vilnius University, Lithuania.
| |
Collapse
|
12
|
Jørgensen NT, Boesen VB, Borresen SW, Christoffersen T, Jørgensen NR, Plomgaard P, Christoffersen C, Watt T, Feldt-Rasmussen U, Klose M. Dual-release hydrocortisone improves body composition and the glucometabolic profile in patients with secondary adrenal insufficiency. Endocrine 2024; 84:1182-1192. [PMID: 38345683 PMCID: PMC11208214 DOI: 10.1007/s12020-024-03711-9] [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: 12/15/2023] [Accepted: 01/21/2024] [Indexed: 06/27/2024]
Abstract
PURPOSE Studies have suggested improved metabolic profiles in patients with adrenal insufficiency treated with dual-release hydrocortisone (DR-HC) compared with conventional hydrocortisone (C-HC). This study investigates the effect of DR-HC compared with C-HC treatment on five health variables: diurnal salivary cortisol/cortisone, body composition, bone health, glucose metabolism, lipids, and blood pressure. METHODS Prospective study of 27 participants (24 men) with secondary adrenal insufficiency with measurements during stable C-HC and 16 weeks after treatment switch to DR-HC. OUTCOMES Diurnal salivary-cortisol/cortisone, body composition assessed by Dual-Energy X-ray absorptiometry scan, bone status indices (serum type I N-terminal procollagen [PINP], collagen type I cross-linked C-telopeptide [CTX], osteocalcin, receptor activator kappa-B [RANK] ligand, osteoprotegerin, and sclerostin), lipids, haemoglobin A1c (HbA1c), and 24-hour blood pressure. RESULTS After the switch to DR-HC, the diurnal salivary-cortisol area under the curve (AUC) decreased non-significantly (mean difference: -55.9 nmol/L/day, P = 0.06). The salivary-cortisone-AUC was unchanged. Late-evening salivary-cortisol and cortisone were lower (-1.6 and -1.7 nmol/L, P = 0.002 and 0.004). Total and abdominal fat mass (-1.5 and -0.5 kg, P = 0.003 and 0.02), HbA1c (-1.2 mmol/mol, P = 0.02), and osteocalcin decreased (-7.0 µg/L, P = 0.03) whereas sclerostin increased (+41.1 pg/mL, P = 0.0001). The remaining bone status indices, lipids, and blood pressure were unchanged. CONCLUSION This study suggests that switching to DR-HC leads to lower late-evening cortisol/cortisone exposure and a more favourable metabolic profile and body composition. In contrast, decreased osteocalcin with increasing sclerostin might indicate a negative impact on bones. CLINICAL TRIAL REGISTRATION EudraCT201400203932.
Collapse
Affiliation(s)
- Nanna Thurmann Jørgensen
- Department of Endocrinology and Metabolism, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Victor Brun Boesen
- Department of Endocrinology and Metabolism, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Stina Willemoes Borresen
- Department of Endocrinology and Metabolism, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Thea Christoffersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
- Department of Clinical Pharmacology, Copenhagen University Hospital, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Niklas Rye Jørgensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Peter Plomgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Torquil Watt
- Department of Endocrinology and Metabolism, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Ulla Feldt-Rasmussen
- Department of Endocrinology and Metabolism, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Marianne Klose
- Department of Endocrinology and Metabolism, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
13
|
Tsimpolis A, Kalafatakis K, Charalampopoulos I. Recent advances in the crosstalk between the brain-derived neurotrophic factor and glucocorticoids. Front Endocrinol (Lausanne) 2024; 15:1362573. [PMID: 38645426 PMCID: PMC11027069 DOI: 10.3389/fendo.2024.1362573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a key neurotrophin within the brain, by selectively activating the TrkB receptor, exerts multimodal effects on neurodevelopment, synaptic plasticity, cellular integrity and neural network dynamics. In parallel, glucocorticoids (GCs), vital steroid hormones, which are secreted by adrenal glands and rapidly diffused across the mammalian body (including the brain), activate two different groups of intracellular receptors, the mineralocorticoid and the glucocorticoid receptors, modulating a wide range of genomic, epigenomic and postgenomic events, also expressed in the neural tissue and implicated in neurodevelopment, synaptic plasticity, cellular homeostasis, cognitive and emotional processing. Recent research evidences indicate that these two major regulatory systems interact at various levels: they share common intracellular downstream pathways, GCs differentially regulate BDNF expression, under certain conditions BDNF antagonises the GC-induced effects on long-term potentiation, neuritic outgrowth and cellular death, while GCs regulate the intraneuronal transportation and the lysosomal degradation of BDNF. Currently, the BDNF-GC crosstalk features have been mainly studied in neurons, although initial findings show that this crosstalk could be equally important for other brain cell types, such as astrocytes. Elucidating the precise neurobiological significance of BDNF-GC interactions in a tempospatial manner, is crucial for understanding the subtleties of brain function and dysfunction, with implications for neurodegenerative and neuroinflammatory diseases, mood disorders and cognitive enhancement strategies.
Collapse
Affiliation(s)
- Alexandros Tsimpolis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas (IMBB-FORTH), Heraklion, Greece
| | - Konstantinos Kalafatakis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Faculty of Medicine and Dentistry (Malta Campus), Queen Mary University of London, Victoria, Malta
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas (IMBB-FORTH), Heraklion, Greece
| |
Collapse
|
14
|
Gubin D. Chronotherapeutic Approaches. CHRONOBIOLOGY AND CHRONOMEDICINE 2024:536-577. [DOI: 10.1039/bk9781839167553-00536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
The chapter provides a comprehensive review of current approaches to personalized chronodiagnosis and chronotherapy. We discuss circadian clock drug targets that aim to affect cellular clock machinery, circadian mechanisms of pharmacokinetics/pharmacodynamics, and chronotherapeutic approaches aimed at increasing treatment efficacy and minimizing its side effects. We explore how chronotherapy can combat acquired and compensatory drug resistance. Non-pharmacological interventions for clock preservation and enhancement are also overviewed, including light treatment, melatonin, sleep scheduling, time-restricted feeding, physical activity, and exercise.
Collapse
Affiliation(s)
- Denis Gubin
- aTyumen State Medical University, Tyumen, Russia
- bTyumen Cardiology Research Center, Tomsk National Research Medical Center, Russian Academy of Science, Tomsk, Russia
| |
Collapse
|
15
|
Lightman SL. Clinical Endocrinology-Time for a Reset? J Endocr Soc 2024; 8:bvae024. [PMID: 38440109 PMCID: PMC10910589 DOI: 10.1210/jendso/bvae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Indexed: 03/06/2024] Open
Abstract
Measurement of blood levels of circulating hormones has always been the cornerstone of the biochemical diagnosis of endocrine diseases, with the objective of detecting hormone excess or insufficiency. Unfortunately, the dynamic nature of hormone secretion means single-point measurements of many hormones often lack diagnostic validity. Endocrinologists have devised complex dynamic tests as indirect assessments of the functioning of the hormone system under investigation. Recent advances in the measurement of dynamic hormone changes across the day now offer an opportunity to reconsider whether there might be better ways both to diagnose and to monitor the therapy of endocrine conditions.
Collapse
Affiliation(s)
- Stafford L Lightman
- Translational Health Sciences, The Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| |
Collapse
|
16
|
Vieluf S, Cantley S, Krishnan V, Loddenkemper T. Ultradian rhythms in accelerometric and autonomic data vary based on seizure occurrence in paediatric epilepsy patients. Brain Commun 2024; 6:fcae034. [PMID: 38454964 PMCID: PMC10919479 DOI: 10.1093/braincomms/fcae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/18/2023] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
Ultradian rhythms are physiological oscillations that resonate with period lengths shorter than 24 hours. This study examined the expression of ultradian rhythms in patients with epilepsy, a disease defined by an enduring seizure risk that may vary cyclically. Using a wearable device, we recorded heart rate, body temperature, electrodermal activity and limb accelerometry in patients admitted to the paediatric epilepsy monitoring unit. In our case-control design, we included recordings from 29 patients with tonic-clonic seizures and 29 non-seizing controls. We spectrally decomposed each signal to identify cycle lengths of interest and compared average spectral power- and period-related markers between groups. Additionally, we related seizure occurrence to the phase of ultradian rhythm in patients with recorded seizures. We observed prominent 2- and 4-hour-long ultradian rhythms of accelerometry, as well as 4-hour-long oscillations in heart rate. Patients with seizures displayed a higher peak power in the 2-hour accelerometry rhythm (U = 287, P = 0.038) and a period-lengthened 4-hour heart rate rhythm (U = 291.5, P = 0.037). Those that seized also displayed greater mean rhythmic electrodermal activity (U = 261; P = 0.013). Most seizures occurred during the falling-to-trough quarter phase of accelerometric rhythms (13 out of 27, χ2 = 8.41, P = 0.038). Fluctuations in seizure risk or the occurrence of seizures may interrelate with ultradian rhythms of movement and autonomic function. Longitudinal assessments of ultradian patterns in larger patient samples may enable us to understand how such rhythms may improve the temporal precision of seizure forecasting models.
Collapse
Affiliation(s)
- Solveig Vieluf
- Division of Epilepsy and Clinical Neurophysiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine I, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Sarah Cantley
- Division of Epilepsy and Clinical Neurophysiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vaishnav Krishnan
- Departments of Neurology, Neuroscience and Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tobias Loddenkemper
- Division of Epilepsy and Clinical Neurophysiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Russell G, Kalafatakis K, Durant C, Marchant N, Thakrar J, Thirard R, King J, Bowles J, Upton T, Thai NJ, Brooks JCW, Wilson A, Phillips K, Ferguson S, Grabski M, Rogers CA, Lampros T, Wilson S, Harmer C, Munafo M, Lightman SL. Ultradian hydrocortisone replacement alters neuronal processing, emotional ambiguity, affect and fatigue in adrenal insufficiency: The PULSES trial. J Intern Med 2024; 295:51-67. [PMID: 37857352 PMCID: PMC10952319 DOI: 10.1111/joim.13721] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
BACKGROUND Primary adrenal insufficiency (PAI) mortality and morbidity remain unacceptably high, possibly arising as glucocorticoid replacement does not replicate natural physiology. A pulsatile subcutaneous pump can closely replicate cortisol's circadian and ultradian rhythm. OBJECTIVES To assess the effect of pump therapy on quality of life, mood, functional neuroimaging, behavioural/cognitive responses, sleep and metabolism. METHODS A 6-week randomised, crossover, double-blinded and placebo-controlled feasibility study of usual dose hydrocortisone in PAI administered as either pulsed subcutaneous or standard care in Bristol, United Kingdom (ISRCTN67193733). Participants were stratified by adrenal insufficiency type. All participants who received study drugs are included in the analysis. The primary outcome, the facial expression recognition task (FERT), occurred at week 6. RESULTS Between December 2014 and 2017, 22 participants were recruited - 20 completed both arms, and 21 were analysed. The pump was well-tolerated. No change was seen in the FERT primary outcome; however, there were subjective improvements in fatigue and mood. Additionally, functional magnetic resonance imaging revealed differential neural processing to emotional cues and visual stimulation. Region of interest analysis identified the left amygdala and insula, key glucocorticoid-sensitive regions involved in emotional ambiguity. FERT post hoc analysis confirmed this response. There were four serious adverse events (AE): three intercurrent illnesses requiring hospitalisation (1/3, 33.3% pump) and a planned procedure (1/1, 100% pump). There was a small number of expected AEs: infusion site bruising/itching (3/5, 60% pump), intercurrent illness requiring extra (3/7, 42% pump) and no extra (4/6, 66% pump) steroid. CONCLUSIONS These findings support the administration of hormone therapy that mimics physiology.
Collapse
Affiliation(s)
- Georgina Russell
- Laboratories of Integrative Neuroscience and EndocrinologyBristol Medical SchoolUniversity of BristolBristolUK
- University Hospital Bristol and Weston NHS Foundation TrustBristolUK
- North Bristol NHS TrustBristolUK
| | - Konstantinos Kalafatakis
- Laboratories of Integrative Neuroscience and EndocrinologyBristol Medical SchoolUniversity of BristolBristolUK
- Department of Informatics and Telecommunications, Human‐Computer Interaction LaboratoryUniversity of IoanninaArtaGreece
- Clinical Research and Imaging CentreUniversity of BristolBristolUK
- Faculty of Medicine and Dentistry (Malta Campus)Queen Mary University of LondonVictoriaMalta
| | - Claire Durant
- Department of Brain SciencesFaculty of MedicineImperial College LondonLondonUK
| | - Nicola Marchant
- Laboratories of Integrative Neuroscience and EndocrinologyBristol Medical SchoolUniversity of BristolBristolUK
- University Hospital Bristol and Weston NHS Foundation TrustBristolUK
| | - Jamini Thakrar
- Laboratories of Integrative Neuroscience and EndocrinologyBristol Medical SchoolUniversity of BristolBristolUK
- Clinical Research and Imaging CentreUniversity of BristolBristolUK
| | - Russell Thirard
- Bristol Trials CentreBristol Medical SchoolUniversity of BristolBristolUK
| | - Jade King
- University Hospital Bristol and Weston NHS Foundation TrustBristolUK
- North Bristol NHS TrustBristolUK
| | - Jane Bowles
- Laboratories of Integrative Neuroscience and EndocrinologyBristol Medical SchoolUniversity of BristolBristolUK
- University Hospital Bristol and Weston NHS Foundation TrustBristolUK
| | - Thomas Upton
- Laboratories of Integrative Neuroscience and EndocrinologyBristol Medical SchoolUniversity of BristolBristolUK
| | - Ngoc Jade Thai
- Clinical Research and Imaging CentreUniversity of BristolBristolUK
- Neurosciences and Mental HealthLiverpool Health PartnersLiverpoolUK
| | | | - Aileen Wilson
- Clinical Research and Imaging CentreUniversity of BristolBristolUK
| | - Kirsty Phillips
- University Hospital Bristol and Weston NHS Foundation TrustBristolUK
| | - Stuart Ferguson
- School of MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | | | - Chris A. Rogers
- Bristol Trials CentreBristol Medical SchoolUniversity of BristolBristolUK
| | - Theodoros Lampros
- Department of Informatics and Telecommunications, Human‐Computer Interaction LaboratoryUniversity of IoanninaArtaGreece
| | - Sue Wilson
- Department of Brain SciencesFaculty of MedicineImperial College LondonLondonUK
| | - Catherine Harmer
- Department of PsychiatryOxford University and Oxford Health NHS Foundation TrustOxfordUK
| | - Marcus Munafo
- MRC Integrative Epidemiology UnitSchool of Psychological ScienceUniversity of BristolBristolUK
| | - Stafford L. Lightman
- Laboratories of Integrative Neuroscience and EndocrinologyBristol Medical SchoolUniversity of BristolBristolUK
- University Hospital Bristol and Weston NHS Foundation TrustBristolUK
| |
Collapse
|
18
|
Lightman S, Upton T. How an appreciation of dynamics has altered our understanding of the HPA axis. Stress 2024; 27:2297371. [PMID: 38258508 PMCID: PMC7617274 DOI: 10.1080/10253890.2023.2297371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/07/2023] [Indexed: 01/24/2024] Open
Abstract
Rhythmicity is a intrinsic feature of biological systems, including the hypothalamic-pituitary-adrenal axis, a mammalian neurohormonal system crucial both in daily life and as a network that responds to stressful stimuli. Circadian and ultradian rhythmicity underlie HPA activity in rodents and in humans, regulating gene expression, metabolism and behavior, and adverse consequences occur when rhythms are disturbed. In the assessment of human disease, the complexity of HPA rhythmicity is rarely acknowledged or understood, and is currently a limitation to better diagnosis and treatment. However, the recent emergence of ambulatory, high frequency and blood-free hormone sampling techniques has the promise to substantially change our understanding of the function of HPA axis in healthy normal life, and provide new opportunities for the diagnosis and treatment of disease.
Collapse
Affiliation(s)
- Stafford Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Bristol, UK
| | - Thomas Upton
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
19
|
Pokharel SS, Brown JL. Physiological plasticity in elephants: highly dynamic glucocorticoids in African and Asian elephants. CONSERVATION PHYSIOLOGY 2023; 11:coad088. [PMID: 39583302 PMCID: PMC10673820 DOI: 10.1093/conphys/coad088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 10/04/2023] [Accepted: 11/01/2023] [Indexed: 09/27/2024]
Abstract
Slowly reproducing and long-lived terrestrial mammals are often more at risk from challenges that influence fitness and survival. It is, therefore, important to understand how animals cope with such challenges and how coping mechanisms translate over generations and affect phenotypic plasticity. Rapidly escalating anthropogenic challenges may further diminish an animal's ability to reinstate homeostasis. Research to advance insights on elephant stress physiology has predominantly focused on relative or comparative analyses of a major stress response marker, glucocorticoids (GCs), across different ecological, anthropogenic, and reproductive contexts. This paper presents an extensive review of published findings on Asian and African elephants from 1980 to 2023 (May) and reveals that stress responses, as measured by alterations in GCs in different sample matrices, often are highly dynamic and vary within and across individuals exposed to similar stimuli, and not always in a predictable fashion. Such dynamicity in physiological reactivity may be mediated by individual differences in personality traits or coping styles, ecological conditions, and technical factors that often are not considered in study designs. We describe probable causations under the 'Physiological Dynamicity Model', which considers context-experience-individuality effects. Highly variable adrenal responses may affect physiological plasticity with potential fitness and survival consequences. This review also addresses the significance of cautious interpretations of GCs data in the context of normal adaptive stress versus distress. We emphasize the need for long-term assessments of GCs that incorporate multiple markers of 'stress' and 'well-being' to decipher the probable fitness consequences of highly dynamic physiological adrenal responses in elephants. Ultimately, we propose that assessing GC responses to current and future challenges is one of the most valuable and informative conservation tools we have for guiding conservation strategies.
Collapse
Affiliation(s)
- Sanjeeta Sharma Pokharel
- Center for Species Survival, Smithsonian National Zoo Conservation Biology Institute, 1500 Remount Road, Front Royal, VA 22630, USA
| | - Janine L Brown
- Center for Species Survival, Smithsonian National Zoo Conservation Biology Institute, 1500 Remount Road, Front Royal, VA 22630, USA
| |
Collapse
|
20
|
de Kloet ER. Glucocorticoid feedback paradox: a homage to Mary Dallman. Stress 2023; 26:2247090. [PMID: 37589046 DOI: 10.1080/10253890.2023.2247090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/07/2023] [Indexed: 08/18/2023] Open
Abstract
As the end product of the hypothalamus-pituitary-adrenal (HPA) axis, the glucocorticoid hormones cortisol and corticosterone coordinate circadian activities, stress-coping, and adaptation to change. For this purpose, the hormone promotes energy metabolism and controls defense reactions in the body and brain. This life-sustaining action exerted by glucocorticoids occurs in concert with the autonomic nervous and immune systems, transmitters, growth factors/cytokines, and neuropeptides. The current contribution will focus on the glucocorticoid feedback paradox in the HPA-axis: the phenomenon that stress responsivity remains resilient if preceded by stress-induced secretion of glucocorticoid hormone, but not if this hormone is previously administered. Furthermore, in animal studies, the mixed progesterone/glucocorticoid antagonist RU486 or mifepristone switches to an apparent partial agonist upon repeated administration. To address these enigmas several interesting phenomena are highlighted. These include the conditional nature of the excitation/inhibition balance in feedback regulation, the role of glucose as a determinant of stress responsivity, and the potential of glucocorticoids in resetting the stress response system. The analysis of the feedback paradox provides also a golden opportunity to review the progress in understanding the role of glucocorticoid hormone in resilience and vulnerability during stress, the science that was burned deeply in Mary Dallman's emotions.
Collapse
Affiliation(s)
- Edo Ronald de Kloet
- Department of Clinical Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
21
|
Ting IJ, Psomas A, Skene DJ, Van der Veen DR. Reduced glucose concentration enhances ultradian rhythms in Pdcd5 promoter activity in vitro. Front Physiol 2023; 14:1244497. [PMID: 37904794 PMCID: PMC10613464 DOI: 10.3389/fphys.2023.1244497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/25/2023] [Indexed: 11/01/2023] Open
Abstract
Intrinsically driven ultradian rhythms in the hourly range are often co-expressed with circadian rhythms in various physiological processes including metabolic processes such as feeding behaviour, gene expression and cellular metabolism. Several behavioural observations show that reduced energy intake or increased energy expenditure leads to a re-balancing of ultradian and circadian timing, favouring ultradian feeding and activity patterns when energy availability is limited. This suggests a close link between ultradian rhythmicity and metabolic homeostasis, but we currently lack models to test this hypothesis at a cellular level. We therefore transduced 3T3-L1 pre-adipocyte cells with a reporter construct that drives a destabilised luciferase via the Pdcd5 promotor, a gene we previously showed to exhibit robust ultradian rhythms in vitro. Ultradian rhythmicity in Pdcd5 promotor driven bioluminescence was observed in >80% of all cultures that were synchronised with dexamethasone, whereas significantly lower numbers exhibited ultradian rhythmicity in non-synchronised cultures (∼11%). Cosine fits to ultradian bioluminescence rhythms in cells cultured and measured in low glucose concentrations (2 mM and 5 mM), exhibited significantly higher amplitudes than all other cultures, and a shorter period (6.9 h vs. 8.2 h, N = 12). Our findings show substantial ultradian rhythmicity in Pdcd5 promotor activity in cells in which the circadian clocks have been synchronised in vitro, which is in line with observations of circadian synchronisation of behavioural ultradian rhythms. Critically, we show that the amplitude of ultradian rhythms is enhanced in low glucose conditions, suggesting that low energy availability enhances ultradian rhythmicity at the cellular level in vitro.
Collapse
|
22
|
Yoshimura M, Flynn BP, Kershaw YM, Zhao Z, Ueta Y, Lightman SL, Conway-Campbell BL. Phase-shifting the circadian glucocorticoid profile induces disordered feeding behaviour by dysregulating hypothalamic neuropeptide gene expression. Commun Biol 2023; 6:998. [PMID: 37775688 PMCID: PMC10541449 DOI: 10.1038/s42003-023-05347-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 09/12/2023] [Indexed: 10/01/2023] Open
Abstract
Here we demonstrate, in rodents, how the timing of feeding behaviour becomes disordered when circulating glucocorticoid rhythms are dissociated from lighting cues; a phenomenon most commonly associated with shift-work and transmeridian travel 'jetlag'. Adrenalectomized rats are infused with physiological patterns of corticosterone modelled on the endogenous adrenal secretory profile, either in-phase or out-of-phase with lighting cues. For the in-phase group, food intake is significantly greater during the rats' active period compared to their inactive period; a feeding pattern similar to adrenal-intact control rats. In contrast, the feeding pattern of the out-of-phase group is significantly dysregulated. Consistent with a direct hypothalamic modulation of feeding behaviour, this altered timing is accompanied by dysregulated timing of anorexigenic and orexigenic neuropeptide gene expression. For Neuropeptide Y (Npy), we report a glucocorticoid-dependent direct transcriptional regulation mechanism mediated by the glucocorticoid receptor (GR). Taken together, our data highlight the adverse behavioural outcomes that can arise when two circadian systems have anti-phasic cues, in this case impacting on the glucocorticoid-regulation of a process as fundamental to health as feeding behaviour. Our findings further highlight the need for development of rational approaches in the prevention of metabolic dysfunction in circadian-disrupting activities such as transmeridian travel and shift-work.
Collapse
Affiliation(s)
- Mitsuhiro Yoshimura
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
- Department of Physiology, University of Occupational and Environmental Health, Japan 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Benjamin P Flynn
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Yvonne M Kershaw
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Zidong Zhao
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Yoichi Ueta
- Department of Physiology, University of Occupational and Environmental Health, Japan 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Stafford L Lightman
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK
| | - Becky L Conway-Campbell
- Translational Health Sciences, Bristol Medical School, University of Bristol Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| |
Collapse
|
23
|
Jeanneteau F. Stress and the risk of Alzheimer dementia: Can deconstructed engrams be rebuilt? J Neuroendocrinol 2023; 35:e13235. [PMID: 36775895 DOI: 10.1111/jne.13235] [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: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
The exact neuropathological mechanism by which the dementia process unfolds is under intense scrutiny. The disease affects about 38 million people worldwide, 70% of which are clinically diagnosed with Alzheimer's disease (AD). If the destruction of synapses essential for learning, planning and decision-making is part of the problem, must the restoration of previously lost synapses be part of the solution? It is plausible that neuronal capacity to restitute information corresponds with the adaptive capacity of its connectivity reserve. A challenge will be to promote the functional connectivity that can compensate for the lost one. This will require better clarification of the remodeling of functional connectivity during the progression of AD dementia and its reversal upon experimental treatment. A major difficulty is to promote the neural pathways that are atrophied in AD dementia while suppressing others that are bolstered. Therapeutic strategies should aim at scaling functional connectivity to a just balance between the atrophic and hypertrophic systems. However, the exact factors that can help reach this objective are still unclear. Similarities between the effects of chronic stress and some neuropathological mechanisms underlying AD dementia support the idea that common components deserve prime attention as therapeutic targets.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
24
|
De la Fuente-Moreno E, Paredes-Ramos P, Carrasco-García A, Hernandez-Cruz B, Alvarado M, Edwards C. Salivary Cortisol in Guide Dogs. Animals (Basel) 2023; 13:1981. [PMID: 37370493 DOI: 10.3390/ani13121981] [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: 04/19/2023] [Revised: 06/07/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023] Open
Abstract
Guide dogs work for extended periods and are exposed to multiple environmental stimuli that could lead to higher stress compared with companion dogs. Cortisol is the main hormone associated with stress in most mammals. This study included seven guide dogs and seven same-breed dogs that were trained as guide dogs but became companion dogs to compare their salivary cortisol levels before, during, and after a period of social isolation and exposure to a 110-decibel gunshot sound. Each dog was left alone in an empty room for 60 min. After 15 min, the dogs were exposed to the sound. We collected four saliva samples from each dog. The first one was taken 5 min before starting the social isolation period, and the following ones at 15, 30, and 45 min after the test started. A two-way ANOVA was used to compare the group effect and the time effect during isolation and noise exposure. The results showed higher levels of cortisol in the guide dogs compared with the companion dogs throughout the test. No differences were found in time or in the interaction between time and group. This suggests that being a guide dog increases levels of basal cortisol when compared with dogs that live as companion animals and family members.
Collapse
Affiliation(s)
| | - Pedro Paredes-Ramos
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Veracruzana, Veracruz 91190, Mexico
| | - Apolo Carrasco-García
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Veracruzana, Veracruz 91190, Mexico
| | - Bertha Hernandez-Cruz
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Veracruzana, Veracruz 91190, Mexico
| | - Mayvi Alvarado
- Instituto de Neuroetología, Xalapa Universidad Veracruzana, Xalapa 91190, Mexico
| | - Claudia Edwards
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
25
|
Birnie M, Claydon M, Troy O, Flynn B, Yoshimura M, Kershaw Y, Zhao Z, Demski-Allen R, Barker G, Warburton E, Bortolotto Z, Lightman S, Conway-Campbell B. Circadian regulation of hippocampal function is disrupted with corticosteroid treatment. Proc Natl Acad Sci U S A 2023; 120:e2211996120. [PMID: 37023133 PMCID: PMC10104554 DOI: 10.1073/pnas.2211996120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/24/2023] [Indexed: 04/07/2023] Open
Abstract
Disrupted circadian activity is associated with many neuropsychiatric disorders. A major coordinator of circadian biological systems is adrenal glucocorticoid secretion which exhibits a pronounced preawakening peak that regulates metabolic, immune, and cardiovascular processes, as well as mood and cognitive function. Loss of this circadian rhythm during corticosteroid therapy is often associated with memory impairment. Surprisingly, the mechanisms that underlie this deficit are not understood. In this study, in rats, we report that circadian regulation of the hippocampal transcriptome integrates crucial functional networks that link corticosteroid-inducible gene regulation to synaptic plasticity processes via an intrahippocampal circadian transcriptional clock. Further, these circadian hippocampal functions were significantly impacted by corticosteroid treatment delivered in a 5-d oral dosing treatment protocol. Rhythmic expression of the hippocampal transcriptome, as well as the circadian regulation of synaptic plasticity, was misaligned with the natural light/dark circadian-entraining cues, resulting in memory impairment in hippocampal-dependent behavior. These findings provide mechanistic insights into how the transcriptional clock machinery within the hippocampus is influenced by corticosteroid exposure, leading to adverse effects on critical hippocampal functions, as well as identifying a molecular basis for memory deficits in patients treated with long-acting synthetic corticosteroids.
Collapse
Affiliation(s)
- Matthew T. Birnie
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Matthew D. B. Claydon
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BristolBS8 1TD, United Kingdom
| | - Oliver Troy
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Benjamin P. Flynn
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Mitsuhiro Yoshimura
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Yvonne M. Kershaw
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Zidong Zhao
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Rebecca C. R. Demski-Allen
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BristolBS8 1TD, United Kingdom
| | - Gareth R. I. Barker
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BristolBS8 1TD, United Kingdom
| | - E. Clea Warburton
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BristolBS8 1TD, United Kingdom
| | - Zuner A. Bortolotto
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BristolBS8 1TD, United Kingdom
| | - Stafford L. Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| | - Becky L. Conway-Campbell
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Translational Health Sciences, Faculty of Health Sciences, School of Medicine, University of Bristol, BristolBS1 3NY, United Kingdom
| |
Collapse
|
26
|
Grant AD, Kriegsfeld LJ. Continuous body temperature as a window into adolescent development. Dev Cogn Neurosci 2023; 60:101221. [PMID: 36821877 PMCID: PMC9981811 DOI: 10.1016/j.dcn.2023.101221] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/06/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Continuous body temperature is a rich source of information on hormonal status, biological rhythms, and metabolism, all of which undergo stereotyped change across adolescence. Due to the direct actions of these dynamic systems on body temperature regulation, continuous temperature may be uniquely suited to monitoring adolescent development and the impacts of exogenous reproductive hormones or peptides (e.g., hormonal contraception, puberty blockers, gender affirming hormone treatment). This mini-review outlines how traditional methods for monitoring the timing and tempo of puberty may be augmented by markers derived from continuous body temperature. These features may provide greater temporal precision, scalability, and reduce reliance on self-report, particularly in females. Continuous body temperature data can now be gathered with ease across a variety of wearable form factors, providing the opportunity to develop tools that aid in individual, parental, clinical, and researcher awareness and education.
Collapse
Affiliation(s)
- Azure D Grant
- Levels Health, Inc., New York City, NY 10003, United States
| | - Lance J Kriegsfeld
- Department of Psychology, University of California, Berkeley, CA 94720, United States; Department of Integrative Biology, University of California, Berkeley, CA 94720, United States; Graduate Group in Endocrinology, University of California, Berkeley, CA 94720, United States; The Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, United States.
| |
Collapse
|
27
|
Meijer OC, Buurstede JC, Viho EMG, Amaya JM, Koning ASCAM, van der Meulen M, van Weert LTCM, Paul SN, Kroon J, Koorneef LL. Transcriptional glucocorticoid effects in the brain: Finding the relevant target genes. J Neuroendocrinol 2023; 35:e13213. [PMID: 36426812 DOI: 10.1111/jne.13213] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 10/25/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
Glucocorticoids are powerful modulators of brain function. They act via mineralocorticoid and glucocorticoid receptors (MR and GR). These are best understood as transcription factors. Although many glucocorticoid effects depend on the modulation of gene transcription, it is a major challenge to link gene expression to function given the large-scale, apparently pleiotropic genomic responses. The extensive sets of MR and GR target genes are highly specific per cell type, and the brain contains many different (neuronal and non-neuronal) cell types. Next to the set "trait" of cellular context, the "state" of other active signaling pathways will affect MR and GR transcriptional activity. Here, we discuss receptor specificity and contextual factors that determine the transcriptional outcome of MR/GR signaling, experimental possibilities offered by single-cell transcriptomics approaches, and reflect on how to make sense of lists of target genes in relation to understanding the functional effects of steroid receptor activation.
Collapse
Affiliation(s)
- Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacobus C Buurstede
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Eva M G Viho
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jorge Miguel Amaya
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne-Sophie C A M Koning
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Merel van der Meulen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa T C M van Weert
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Susana N Paul
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Kroon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa L Koorneef
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
28
|
Jeanneteau F, Meijer OC, Moisan MP. Structural basis of glucocorticoid receptor signaling bias. J Neuroendocrinol 2023; 35:e13203. [PMID: 36221223 DOI: 10.1111/jne.13203] [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: 05/21/2022] [Revised: 09/15/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022]
Abstract
Dissociation between the healthy and toxic effects of cortisol, a major stress-responding hormone has been a widely used strategy to develop anti-inflammatory glucocorticoids with fewer side effects. Such strategy falls short when treating brain disorders as timing and activity state within large-scale neuronal networks determine the physiological and behavioral specificity of cortisol response. Advances in structural molecular dynamics posit the bases for engineering glucocorticoids with precision bias for select downstream signaling pathways. Design of allosteric and/or cooperative control for the glucocorticoid receptor could help promote the beneficial and reduce the deleterious effects of cortisol on brain and behavior in disease conditions.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Onno C Meijer
- Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
29
|
Habets PC, Kalafatakis K, Dzyubachyk O, van der Werff SJ, Keo A, Thakrar J, Mahfouz A, Pereira AM, Russell GM, Lightman SL, Meijer OC. Transcriptional and cell type profiles of cortical brain regions showing ultradian cortisol rhythm dependent responses to emotional face stimulation. Neurobiol Stress 2023; 22:100514. [PMID: 36660181 PMCID: PMC9842700 DOI: 10.1016/j.ynstr.2023.100514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/02/2023] [Accepted: 01/02/2023] [Indexed: 01/05/2023] Open
Abstract
The characteristic endogenous circadian rhythm of plasma glucocorticoid concentrations is made up from an underlying ultradian pulsatile secretory pattern. Recent evidence has indicated that this ultradian cortisol pulsatility is crucial for normal emotional response in man. In this study, we investigate the anatomical transcriptional and cell type signature of brain regions sensitive to a loss of ultradian rhythmicity in the context of emotional processing. We combine human cell type and transcriptomic atlas data of high spatial resolution with functional magnetic resonance imaging (fMRI) data. We show that the loss of cortisol ultradian rhythm alters emotional processing response in cortical brain areas that are characterized by transcriptional and cellular profiles of GABAergic function. We find that two previously identified key components of rapid non-genomic GC signaling - the ANXA1 gene and retrograde endocannabinoid signaling - show most significant differential expression (q = 3.99e-10) and enrichment (fold enrichment = 5.56, q = 9.09e-4). Our results further indicate that specific cell types, including a specific NPY-expressing GABAergic neuronal cell type, and specific G protein signaling cascades underly the cerebral effects of a loss of ultradian cortisol rhythm. Our results provide a biological mechanistic underpinning of our fMRI findings, indicating specific cell types and cascades as a target for manipulation in future experimental studies.
Collapse
Affiliation(s)
- Philippe C. Habets
- Leiden University Medical Center, Department of Medicine, Division of Endocrinology, 2300 RC Leiden, the Netherlands
- Amsterdam University Medical Centre, Department of Psychiatry, Department of Anatomy and Neurosciences, 1081 HZ, Amsterdam, the Netherlands
| | - Konstantinos Kalafatakis
- Henry Wellcome Laboratories of Integrative Neuroscience and Endocrinology, Bristol Medical School, University of Bristol, BS1 3NY, Bristol, United Kingdom
- Institute of Health Science Education, Barts and the London School of Medicine & Dentistry, Queen Mary University of London Malta Campus, VCT 2520, Victoria Gozo, Malta
| | - Oleh Dzyubachyk
- Department of Radiology, Division of Medical Image Processing, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
- Leiden University Medical Center, Department of Cell and Chemical Biology, Section Electron Microscopy, 2300 RC, Leiden, the Netherlands
| | - Steven J.A. van der Werff
- Department of Psychiatry, Leiden University Medical Center LUMC, Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden, the Netherlands
| | - Arlin Keo
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, the Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
| | - Jamini Thakrar
- Henry Wellcome Laboratories of Integrative Neuroscience and Endocrinology, Bristol Medical School, University of Bristol, BS1 3NY, Bristol, United Kingdom
| | - Ahmed Mahfouz
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, the Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Alberto M. Pereira
- Leiden University Medical Center, Department of Medicine, Division of Endocrinology, 2300 RC Leiden, the Netherlands
- Department of Endocrinology & Metabolism, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Georgina M. Russell
- Henry Wellcome Laboratories of Integrative Neuroscience and Endocrinology, Bristol Medical School, University of Bristol, BS1 3NY, Bristol, United Kingdom
| | - Stafford L. Lightman
- Henry Wellcome Laboratories of Integrative Neuroscience and Endocrinology, Bristol Medical School, University of Bristol, BS1 3NY, Bristol, United Kingdom
| | - Onno C. Meijer
- Leiden University Medical Center, Department of Medicine, Division of Endocrinology, 2300 RC Leiden, the Netherlands
- Leiden Institute for Brain and Cognition, Leiden, the Netherlands
| |
Collapse
|
30
|
Kelly MR, Yuen F, Satterfield BC, Auchus RJ, Gaddameedhi S, Van Dongen HPA, Liu PY. Endogenous Diurnal Patterns of Adrenal and Gonadal Hormones During a 24-Hour Constant Routine After Simulated Shift Work. J Endocr Soc 2022; 6:bvac153. [PMID: 36330292 PMCID: PMC9620969 DOI: 10.1210/jendso/bvac153] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Indexed: 01/12/2023] Open
Abstract
Context Night-shift work causes circadian misalignment, predicts the development of metabolic diseases, and complicates the interpretation of hormone measurements. Objective To investigate endogenous circadian rhythms, dissociated from behavioral and environmental confounds, in adrenal and gonadal steroids after simulated shift work. Methods Fourteen healthy adults (ages 25.8 ± 3.2 years) were randomized to 3 days of night or day (control) shift work followed by a constant routine protocol designed to experimentally unveil rhythms driven endogenously by the central circadian pacemaker. Blood was sampled every 3 hours for 24 hours during the constant routine to concurrently obtain 16 Δ4 steroid profiles by mass spectrometry. Cosinor analyses of these profiles provided mesor (mean abundance), amplitude (oscillation magnitude), and acrophase (peak timing). Results Night-shift work marginally increased cortisol by 1 μg/dL (P = 0.039), and inactive/weak derivatives cortisone (P = 0.003) and 18-hydroxycortisol (P < 0.001), but did not alter the mesor of potent androgens testosterone and 11-ketotestosterone. Adrenal-derived steroids, including 11-ketotestosterone (P < 0.01), showed robust circadian rhythmicity after either day- or night-shift work. In contrast, testosterone and progesterone showed no circadian pattern after both shift work conditions. Night-shift work did not alter the amplitude or acrophase of any of the steroid profiles. Conclusion Experimental circadian misalignment had minimal effects on steroidogenesis. Adrenal steroids, but not gonadal hormones, showed endogenous circadian regulation robust to prior shift schedule. This dichotomy may predispose night-shift workers to metabolic ill health. Furthermore, adrenal steroids, including cortisol and the main adrenal androgen 11-ketostosterone, should always be evaluated during the biological morning whereas assessment of gonadal steroids, particularly testosterone, is dependent on the shift-work schedule.
Collapse
Affiliation(s)
- Monica R Kelly
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Geriatric Research, Education and Clinical Center, North Hills, CA, USA
| | - Fiona Yuen
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Division of Endocrinology, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Brieann C Satterfield
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Richard J Auchus
- Division of Metabolism, Diabetes, and Endocrinology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Shobhan Gaddameedhi
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Hans P A Van Dongen
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Peter Y Liu
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Division of Endocrinology, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| |
Collapse
|
31
|
Van't Westeinde A, Padilla N, Siqueiros Sanchez M, Fletcher-Sandersjöö S, Kämpe O, Bensing S, Lajic S. Brain structure in autoimmune Addison's disease. Cereb Cortex 2022; 33:4915-4926. [PMID: 36227196 PMCID: PMC10110435 DOI: 10.1093/cercor/bhac389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/12/2022] Open
Abstract
Long-term disturbances in cortisol levels might affect brain structure in individuals with autoimmune Addison's disease (AAD). This study investigated gray and white matter brain structure in a cohort of young adults with AAD. T1- and diffusion-weighted images were acquired for 52 individuals with AAD and 70 healthy controls, aged 19-43 years, using magnetic resonance imaging. Groups were compared on cortical thickness, surface area, cortical gray matter volume, subcortical volume (FreeSurfer), and white matter microstructure (FSL tract-based spatial statistics). Individuals with AAD had 4.3% smaller total brain volume. Correcting for head size, we did not find any regional structural differences, apart from reduced volume of the right superior parietal cortex in males with AAD. Within the patient group, a higher glucocorticoid (GC) replacement dose was associated with smaller total brain volume and smaller volume of the left lingual gyrus, left rostral anterior cingulate cortex, and right supramarginal gyrus. With the exception of smaller total brain volume and potential sensitivity of the parietal cortex to GC disturbances in men, brain structure seems relatively unaffected in young adults with AAD. However, the association between GC replacement dose and reduced brain volume may be reason for concern and requires follow-up study.
Collapse
Affiliation(s)
- Annelies Van't Westeinde
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Karolinskavagen 37A, SE-171 76 Stockholm, Sweden
| | - Nelly Padilla
- Unit for Neonatology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Karolinskavagen 37A, SE-171 76 Stockholm, Sweden
| | - Monica Siqueiros Sanchez
- Brain Imaging, Development and Genetics (BRIDGE) Lab, Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Stanford, CA 94305-5101, United States
| | - Sara Fletcher-Sandersjöö
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-171 76 Stockholm, Sweden.,Department of Endocrinology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Olle Kämpe
- Department of Endocrinology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden.,Department of Medicine (Solna), Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Sophie Bensing
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-171 76 Stockholm, Sweden.,Department of Endocrinology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Svetlana Lajic
- Pediatric Endocrinology Unit, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Karolinskavagen 37A, SE-171 76 Stockholm, Sweden
| |
Collapse
|
32
|
Jeanneteau F, Coutellier L. The glucocorticoid footprint on the memory engram. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 25:100378. [PMID: 38486965 PMCID: PMC10938917 DOI: 10.1016/j.coemr.2022.100378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
The complexity of the classical inverted U-shaped relationship between cortisol levels and responses transposable to stress reactivity has led to an incomplete understanding of the mechanisms enabling healthy and toxic effects of stress on brain and behavior. A clearer, more detailed, picture of those relationships can be obtained by integrating cortisol effects on large-scale brain networks, in particular, by focusing on neural network configurations from the perspective of inhibition and excitation. A unifying view of Semon and Hebb's theories of cellular memory links the biophysical and metabolic changes in neuronal ensembles to the strengthening of collective synapses. In that sense, the neuronal capacity to record, store, and retrieve information directly relates to the adaptive capacity of its connectivity and metabolic reserves. Here, we use task-activated cell ensembles or simply engram cells as an example to demonstrate that the adaptive behavioral responses to stress result from collective synapse strength within and across networks of interneurons and excitatory ones.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Laurence Coutellier
- Departments of Psychology and Neuroscience, Ohio State University, Columbus, USA
| |
Collapse
|
33
|
Dekkers AJ, Amaya JM, van der Meulen M, Biermasz NR, Meijer OC, Pereira AM. Long-term effects of glucocorticoid excess on the brain. J Neuroendocrinol 2022; 34:e13142. [PMID: 35980208 PMCID: PMC9541651 DOI: 10.1111/jne.13142] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 11/27/2022]
Abstract
The metabolic and cardiovascular clinical manifestations in patients with Cushing's syndrome (CS) are generally well known. However, recent studies have broadened the perspective of the effects of hypercortisolism, showing that both endogenous and exogenous glucocorticoid excess alter brain functioning on several time scales. Consequently, cognitive deficits and neuropsychological symptoms are highly prevalent during both active CS and CS in remission, as well as during glucocorticoid treatment. In this review, we discuss the effects of endogenous hypercortisolism and exogenously induced glucocorticoid excess on the brain, as well as the prevalence of cognitive and neuropsychological deficits and their course after biochemical remission. Furthermore, we propose possible mechanisms that may underly neuronal changes, based on experimental models and in vitro studies. Finally, we offer recommendations for future studies.
Collapse
Affiliation(s)
- Alies J. Dekkers
- Department of Medicine, Division of Endocrinology, Pituitary Center and Center for Endocrine TumorsLeiden University Medical CenterLeidenThe Netherlands
- Department of Medicine, Center for Endocrine Tumours LeidenLeiden University Medical CenterLeidenThe Netherlands
| | - Jorge Miguel Amaya
- Department of Medicine, Division of Endocrinology, Pituitary Center and Center for Endocrine TumorsLeiden University Medical CenterLeidenThe Netherlands
| | - Merel van der Meulen
- Department of Medicine, Division of Endocrinology, Pituitary Center and Center for Endocrine TumorsLeiden University Medical CenterLeidenThe Netherlands
- Department of Medicine, Center for Endocrine Tumours LeidenLeiden University Medical CenterLeidenThe Netherlands
| | - Nienke R. Biermasz
- Department of Medicine, Division of Endocrinology, Pituitary Center and Center for Endocrine TumorsLeiden University Medical CenterLeidenThe Netherlands
- Department of Medicine, Center for Endocrine Tumours LeidenLeiden University Medical CenterLeidenThe Netherlands
| | - Onno C. Meijer
- Department of Medicine, Division of Endocrinology, Pituitary Center and Center for Endocrine TumorsLeiden University Medical CenterLeidenThe Netherlands
| | - Alberto M. Pereira
- Department of Endocrinology & MetabolismAmsterdam UMC (AMC)AmsterdamThe Netherlands
| |
Collapse
|
34
|
Torres G, Mourad M, Leheste JR. Indoor Air Pollution and Decision-Making Behavior: An Interdisciplinary Review. Cureus 2022; 14:e26247. [PMID: 35911286 PMCID: PMC9313076 DOI: 10.7759/cureus.26247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 12/01/2022] Open
Abstract
The human brain is constantly exposed to air pollutants, some of which might be disruptive or even lethal to certain neurons implicated in abstract features of cognitive function. In this review, we present new evidence from behavioral and neural studies in humans, suggesting a link between indoor fine particulate matter and decision-making behavior. To illustrate this relationship, we use qualitative sources, such as historical documents of the Vietnam War to develop hypotheses of how aerial transmission of pollutants might obstruct alternative choices during the evaluation of policy decisions. We first describe the neural circuits driving decision-making processes by addressing how neurons and their cognate receptors directly evaluate and transduce physical phenomena into sensory perceptions that allow us to decide the best course of action among competing alternatives. We then raise the possibility that indoor air pollutants might also impact cell-signaling systems outside the brain parenchyma to further obstruct the computational analysis of the social environment. We also highlight how particulate matter might be pathologically integrated into the brain to override control of sensory decisions, and thereby perturb selection of choice. These lines of research aim to extend our understanding of how inhalation of airborne particulates and toxicants in smoke, for example, might contribute to cognitive impairment and negative health outcomes.
Collapse
|
35
|
Dromard Y, Arango-Lievano M, Borie A, Dedin M, Fontanaud P, Torrent J, Garabedian MJ, Ginsberg SD, Jeanneteau F. Loss of glucocorticoid receptor phosphorylation contributes to cognitive and neurocentric damages of the amyloid-β pathway. Acta Neuropathol Commun 2022; 10:91. [PMID: 35733193 PMCID: PMC9219215 DOI: 10.1186/s40478-022-01396-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 12/22/2022] Open
Abstract
Aberrant cortisol and activation of the glucocorticoid receptor (GR) play an essential role in age-related progression of Alzheimer's disease (AD). However, the GR pathways required for influencing the pathobiology of AD dementia remain unknown. To address this, we studied an early phase of AD-like progression in the well-established APP/PS1 mouse model combined with targeted mutations in the BDNF-dependent GR phosphorylation sites (serines 134/267) using molecular, behavioral and neuroimaging approaches. We found that disrupting GR phosphorylation (S134A/S267A) in mice exacerbated the deleterious effects of the APP/PS1 genotype on mortality, neuroplasticity and cognition, without affecting either amyloid-β deposition or vascular pathology. The dynamics, maturation and retention of task-induced new dendritic spines of cortical excitatory neurons required GR phosphorylation at the BDNF-dependent sites that amyloid-β compromised. Parallel studies in postmortem human prefrontal cortex revealed AD subjects had downregulated BDNF signaling and concomitant upregulated cortisol pathway activation, which correlated with cognitive decline. These results provide key evidence that the loss of neurotrophin-mediated GR phosphorylation pathway promotes the detrimental effects of the brain cortisol response that contributes to the onset and/or progression of AD dementia. These findings have important translational implications as they provide a novel approach to treating AD dementia by identifying drugs that increase GR phosphorylation selectively at the neurotrophic sites to improve memory and cognition.
Collapse
Affiliation(s)
- Yann Dromard
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France
| | - Margarita Arango-Lievano
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France
| | - Amelie Borie
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France
| | - Maheva Dedin
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France
| | - Pierre Fontanaud
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France
- Imagerie du Petit Animal de Montpellier, 34090, Montpellier, France
| | - Joan Torrent
- Institut de Neuroscience de Montpellier, INSERM, 34090, Montpellier, France
| | - Michael J Garabedian
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Stephen D Ginsberg
- Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, Neuroscience & Physiology, NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Freddy Jeanneteau
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France.
| |
Collapse
|
36
|
Van't Westeinde A, Ström S, Hirvikoski T, Dahlqvist P, Wahlberg J, Gezelius A, Kämpe O, Bensing S, Lajic S. Young adult Swedish patients with autoimmune Addison's disease report difficulties with executive functions in daily life despite overall good cognitive performance. Psychoneuroendocrinology 2022; 140:105714. [PMID: 35290880 DOI: 10.1016/j.psyneuen.2022.105714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Sub-optimal replacement of glucocorticoids (GC) in autoimmune Addison's disease (AAD) may affect cognitive functioning. The present study therefore sought to investigate cognitive performance and self-reported problems with executive functions in a cohort of young adult patients with AAD. DESIGN AND METHODS 67 patients with AAD (39 females), mean age 32 yrs. (range 19-41), and 80 control participants (43 females), mean age 29 yrs. (range 19-43), completed neuropsychological tests estimating verbal and non-verbal intellectual ability, learning, memory and executive functioning, in addition to self-report scales assessing problems with executive functions, fatigue and symptoms of anxiety and depression. RESULTS Patients performed within the average range on all cognitive tests compared to population norms. However, female AAD patients reported more problems than controls with both hot (emotion regulation) and cold (cognitive regulation) executive functions in daily life. Moreover, experienced problems with executive functions in both male and female patients were associated with increased mental fatigue and lower GC replacement doses. CONCLUSIONS Despite average performance in neuropsychological tests by both sexes, young adult female patients with AAD experience problems with executive functions in daily life. Coping with mental fatigue and optimization of pharmacotherapy may be important factors to be addressed in order to provide timely support for patients. Future research is needed to further determine other risk factors for experiencing executive function impairments in AAD.
Collapse
Affiliation(s)
- Annelies Van't Westeinde
- Department of Women's and Children's Health, Karolinska Institutet, Pediatric Endocrinology Unit, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Sara Ström
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Endocrinology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Tatja Hirvikoski
- Department of Women's and Children's Health, Pediatric Neuropsychiatry Unit, Center for Neurodevelopmental Disorders at Karolinska Institutet (KIND), Karolinska Institutet, SE-17177 Stockholm Sweden; Unit for Habilitation & Health, Stockholm County Council, Sweden
| | - Per Dahlqvist
- Department of Public Health and Clinical Medicine, Umeå University, SE-901 87 Umeå, Sweden
| | - Jeanette Wahlberg
- Department of Endocrinology and Department of Medical and Health Sciences, Linköpings University, SE-581 83 Linköping, Sweden; Department of Internal Medicine, School of Health and Medical Sciences, Örebro University, SE-702 81 Örebro, Sweden
| | - Anton Gezelius
- Department of Women's and Children's Health, Karolinska Institutet, Pediatric Endocrinology Unit, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Olle Kämpe
- Department of Medicine (Solna), Center for Molecular Medicine, Karolinska Institutet, Sweden
| | - Sophie Bensing
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Endocrinology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Svetlana Lajic
- Department of Women's and Children's Health, Karolinska Institutet, Pediatric Endocrinology Unit, Karolinska University Hospital, SE-171 76 Stockholm, Sweden.
| |
Collapse
|
37
|
Castle-Kirszbaum M, Shi MDY, Goldschlager T. Quality of Life in Craniopharyngioma: A Systematic Review. World Neurosurg 2022; 164:424-435.e2. [PMID: 35580780 DOI: 10.1016/j.wneu.2022.05.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Craniopharyngiomas are morbid tumors that significantly reduce patients' quality of life (QoL). The lifelong burden of endocrine, visual, hypothalamic, and limbic dysfunction can have disastrous consequences for the physical and psychosocial health of patients. Elucidating the factors that influence QoL could guide therapeutic interventions to improve patient well-being. METHODS A systematic review was performed in accordance with the PRISMA (preferred reporting items for systematic reviews and meta-analyses) statement using the PubMed and Medline databases. Studies that had reported patient QoL using validated metrics in both adult and pediatric populations were included. Bias and methodological rigor were assessed using the MINORS (methodological index for nonrandomized studies) criteria. RESULTS A total of 25 studies, including 2025 patients, were available for review. Most studies were small, retrospective, cohort studies with a high risk of bias. The QoL of the patients with craniopharyngioma was lower than that of the general population. Hypothalamic involvement was consistently the strongest predictor of QoL. Endocrinopathy contributed to morbidity but could be ameliorated by hormone replacement therapy. Social and emotional dysregulation and a poor memory are common complaints after surgery, and iatrogenic damage to the infundibulum, hypothalamus, limbic system, and frontal lobes might underlie these concerns. Sleep-wake cycle dysfunction and hypothalamic obesity are serious consequences of hypothalamic damage. CONCLUSIONS An experienced multidisciplinary team is necessary to optimally manage the complex cases of these patients. The poor QoL of patients with craniopharyngioma is multifactorial. However, the contribution of iatrogenesis is not insubstantial. Improved surgical techniques, focusing on hypothalamic preservation, and adjuvant treatment options are required to improve the well-being of these patients.
Collapse
Affiliation(s)
- Mendel Castle-Kirszbaum
- Department of Neurosurgery, Monash Health, Melbourne, Victoria, Australia; Department of Surgery, Monash University, Melbourne, Victoria, Australia.
| | - Margaret D Y Shi
- Department of Surgery, Northern Hospital, Melbourne, Victoria, Australia
| | - Tony Goldschlager
- Department of Neurosurgery, Monash Health, Melbourne, Victoria, Australia; Department of Surgery, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
38
|
Vaudin P, Augé C, Just N, Mhaouty-Kodja S, Mortaud S, Pillon D. When pharmaceutical drugs become environmental pollutants: Potential neural effects and underlying mechanisms. ENVIRONMENTAL RESEARCH 2022; 205:112495. [PMID: 34883077 DOI: 10.1016/j.envres.2021.112495] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/12/2021] [Accepted: 12/01/2021] [Indexed: 06/13/2023]
Abstract
Pharmaceutical drugs have become consumer products, with a daily use for some of them. The volume of production and consumption of drugs is such that they have become environmental pollutants. Their transfer to wastewater through urine, feces or rinsing in case of skin use, associated with partial elimination by wastewater treatment plants generalize pollution in the hydrosphere, including drinking water, sediments, soils, the food chain and plants. Here, we review the potential effects of environmental exposure to three classes of pharmaceutical drugs, i.e. antibiotics, antidepressants and non-steroidal anti-inflammatory drugs, on neurodevelopment. Experimental studies analyzing their underlying modes of action including those related to endocrine disruption, and molecular mechanisms including epigenetic modifications are presented. In addition, the contribution of brain imaging to the assessment of adverse effects of these three classes of pharmaceuticals is approached.
Collapse
Affiliation(s)
- Pascal Vaudin
- Physiologie de La Reproduction et des Comportements, CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380, Nouzilly, France.
| | - Corinne Augé
- UMR 1253, IBrain, University of Tours, INSERM, 37000, Tours, France
| | - Nathalie Just
- Physiologie de La Reproduction et des Comportements, CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380, Nouzilly, France
| | - Sakina Mhaouty-Kodja
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Stéphane Mortaud
- Immunologie et Neurogénétique Expérimentales et Moléculaires, UMR7355, CNRS, Université D'Orléans, 45000, Orléans, France
| | - Delphine Pillon
- Physiologie de La Reproduction et des Comportements, CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380, Nouzilly, France
| |
Collapse
|
39
|
Abstract
Addison's disease typically results from the autoimmune destruction of the adrenal cortex and requires lifelong replacement with glucocorticoids and mineralocorticoids. Main symptoms are non-specific and, therefore, often overlooked or misleading. Patients are frequently not diagnosed until experiencing a life-threatening adrenal crisis. This article highlights the essential clinical characteristics, diagnostic aspects and principles of management of adrenal insufficiency.
Collapse
|
40
|
Koulouris N, Dimakou K, Gourgoulianis K, Tzanakis N, Rapti A, Gaga M, Georgatou N, Steiropoulos P, Karachristos C, Gogali A, Kalafatakis K, Kostikas K. Self-perceived quality of sleep among COPD patients in Greece: the SLEPICO study. Sci Rep 2022; 12:540. [PMID: 35017591 PMCID: PMC8752730 DOI: 10.1038/s41598-021-04610-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/01/2021] [Indexed: 01/03/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a leading cause of morbidity and mortality worldwide accompanied by a substantial social and economic burden for the patient and the society. Poor sleep quality among COPD patients is frequently unnoticed and unaddressed by physicians and patients themselves, although it is a major source of further deterioration of these patients' quality of life. The aim of the present study was to record the quality of sleep in COPD patients among the Greek population and correlate these findings with various features of these patients, using the COPD and Asthma Sleep Impact Scale (CASIS). This was a cross-sectional observational study. Forty different variables (demographics, vital sign measurements, COPD-related medical history parameters, comorbidities, CASIS questionnaire results, COPD assessment test, COPD severity based on spirometry measurements, COPD stage based on the ABCD assessment approach, inhaled COPD treatment report) were collected from 3454 nation-wide COPD patients (Greece). The study sample consisted of COPD patients, mainly male (73%) with a median age of 69 years and a median BMI of 27.2. More than half of COPD patients (60.6%) suffered from moderate disease severity and 23.8% from severe disease, while less than half (42.1%) suffered from at least one exacerbation of the disease over the last year prior study enrollment. About 14% reported frequent to very frequent issues affecting their sleep quality, between a fourth and a third of them reported occasional night sleep disturbances, and at least half of them reported no or very infrequent problems in their night sleep. Our study indicates that the COPD assessment test (CAT) and the spirometry-based disease severity can predict the poorness in the quality of sleep (F2,3451 = 1397.5, p < 0.001, adj. R2 = 0.45) as assessed by CASIS score, and that the latter also correlates with age (ρ = 0.122, p < 0.001) and disease duration (ρ = 0.104, p < 0.001). On the contrary, there appears to be no correlation between sleep quality and number of exacerbations. Finally, untreated patients with COPD suffer from poorer quality of sleep compared to treated subjects, independently of the use of inhaled corticosteroids (F2,3451 = 21.65, p < 0.001). The results of the SLEPICO study show that increased age, prolonged disease duration, and especially CAT score ≥ 10, and severe COPD stage, might act as important indicators for deterioration in the quality of sleep, with potential consequences in the daily routine of those patients, thus urging potentially for further pharmacological interventions or modifications.
Collapse
Affiliation(s)
- Nikolaos Koulouris
- First Department of Pulmonary Medicine and Intensive Care Unit, National and Kapodistrian University of Athens, Medical School, 115 27, Athens, Greece
| | - Katerina Dimakou
- 5Th Respiratory Medicine Department, General Hospital for Chest Diseases of Athens "SOTIRIA", Athens, Greece
| | - Konstantinos Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Nikolaos Tzanakis
- Department of Respiratory Medicine, University General Hospital of Heraklion, Medical School, University of Crete, 71003, Heraklion, Greece
| | - Aggeliki Rapti
- 2Nd Respiratory Medicine Department, General Hospital for Chest Diseases of Athens "SOTIRIA", Athens, Greece
| | - Mina Gaga
- 7Th Respiratory Medicine Department, Athens "Sotiria" Chest Diseases Hospital, Athens, Greece
| | | | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, University General Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | - Christos Karachristos
- Department of Pulmonary Medicine, General Hospital of Thessaloniki "Georgios Papanikolaou", G. Papanikolaou Ave, 57010, Exohi, Greece
| | - Athena Gogali
- Respiratory Medicine Department, University Hospital of Ioannina, Ioannina, Greece
| | - Konstantinos Kalafatakis
- Department of Informatics and Telecommunications, School of Informatics and Telecommunications, University of Ioannina, Arta, Greece
| | | |
Collapse
|
41
|
Chai Y, Li Q, Wang Y, Tao E, Asakawa T. The Value of HPA Axis Hormones as Biomarkers for Screening and Early Diagnosis of Postpartum Depression: Updated Information About Methodology. Front Endocrinol (Lausanne) 2022; 13:916611. [PMID: 35903273 PMCID: PMC9315198 DOI: 10.3389/fendo.2022.916611] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/25/2022] [Indexed: 12/30/2022] Open
Abstract
Because of the high prevalence of postpartum depression (PPD) and the suffering involved, early diagnosis is urgent; however, current screening tools and diagnosis are inadequate. In addition to conventional methods such as the Edinburgh Postnatal Depression Scale and clinical interviews, several hormones in the hypothalamic-pituitary-adrenal (HPA) axis, such as corticotrophin-releasing hormone, adrenocorticotropic hormone, and cortisol, have been considered because of their critical roles in stress regulation in the mothers. The study designs are complicated, however, and so the effectiveness of these hormones as biomarkers for PPD is still controversial. Such inconsistency may have resulted from the variation in methodology between studies. The methodology problems in the investigation of PPD and HPA axis hormones have not been reported extensively. We therefore sought to summarize the methodological problems of studies published in the past decade, including the strengths and weaknesses of the examinations and the technological difficulties involved. Our findings suggest that (a) suitable samples and appropriate detection methods would reduce heterogeneity among trials; (b) the cutoff value of the scale test should be carefully selected for determining the performance of biomarker tests; (c) evaluation methods and criteria should be chosen with consideration of the tools feasible for use in local hospitals and population; and (d) the cost of diagnosis should be reduced. We hope that these findings provide insight for future investigations of HPA axis hormones as biomarkers for screening and early diagnosis of PPD.
Collapse
Affiliation(s)
- Yujuan Chai
- Department of Biomedical Engineering, Shenzhen University, Shenzhen, China
| | - Qihang Li
- Department of Biomedical Engineering, Shenzhen University, Shenzhen, China
| | - Yang Wang
- Greater Bay Area International Institute for Innovation, Shenzhen University, Shenzhen, China
| | - Enxiang Tao
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Tetsuya Asakawa
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Institute of Neurology, The Third People’s Hospital of Shenzhen, Shenzhen, China
- Research Base of Traditional Chinese Medicine Syndrome, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- *Correspondence: Tetsuya Asakawa, ; orcid.org/0000-0002-2300-3509
| |
Collapse
|
42
|
Grant AD, Wilbrecht L, Kriegsfeld LJ. Adolescent Development of Biological Rhythms in Female Rats: Estradiol Dependence and Effects of Combined Contraceptives. Front Physiol 2021; 12:752363. [PMID: 35615288 PMCID: PMC9126190 DOI: 10.3389/fphys.2021.752363] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/11/2021] [Indexed: 01/23/2023] Open
Abstract
Adolescence is a period of continuous development, including the maturation of endogenous rhythms across systems and timescales. Although, these dynamic changes are well-recognized, their continuous structure and hormonal dependence have not been systematically characterized. Given the well-established link between core body temperature (CBT) and reproductive hormones in adults, we hypothesized that high-resolution CBT can be applied to passively monitor pubertal development and disruption with high fidelity. To examine this possibility, we used signal processing to investigate the trajectory of CBT rhythms at the within-day (ultradian), daily (circadian), and ovulatory timescales, their dependence on estradiol (E2), and the effects of hormonal contraceptives. Puberty onset was marked by a rise in fecal estradiol (fE2), followed by an elevation in CBT and circadian power. This time period marked the commencement of 4-day rhythmicity in fE2, CBT, and ultradian power marking the onset of the estrous cycle. The rise in circadian amplitude was accelerated by E2 treatment, indicating a role for this hormone in rhythmic development. Contraceptive administration in later adolescence reduced CBT and circadian power and resulted in disruption to 4-day cycles that persisted after discontinuation. Our data reveal with precise temporal resolution how biological rhythms change across adolescence and demonstrate a role for E2 in the emergence and preservation of multiscale rhythmicity. These findings also demonstrate how hormones delivered exogenously in a non-rhythmic pattern can disrupt rhythmic development. These data lay the groundwork for a future in which temperature metrics provide an inexpensive, convenient method for monitoring pubertal maturation and support the development of hormone therapies that better mimic and support human chronobiology.
Collapse
Affiliation(s)
- Azure D. Grant
- The Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| | - Linda Wilbrecht
- The Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
- Department of Psychology, University of California, Berkeley, Berkeley, CA, United States
| | - Lance J. Kriegsfeld
- The Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
- Department of Psychology, University of California, Berkeley, Berkeley, CA, United States
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, United States
- Graduate Group in Endocrinology, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
43
|
Udeh-Momoh C, Watermeyer T. Female specific risk factors for the development of Alzheimer's disease neuropathology and cognitive impairment: Call for a precision medicine approach. Ageing Res Rev 2021; 71:101459. [PMID: 34508876 DOI: 10.1016/j.arr.2021.101459] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/13/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) includes a long asymptomatic stage, which precedes the formal diagnosis of dementia. AD biomarker models provide a framework for precision medicine approaches during this stage. However, such approaches have ignored the possible influence of sex on cognition and brain health, despite female sex noted as a major risk factor. Since AD-related changes may emerge in midlife, intervention efforts are being redirected around this period. Midlife coincides with several endocrinological changes, such as the menopausal transition experienced by women. In this narrative review, we discuss evidence for sex-differences in AD neuropathological burden and outline key endocrinological mechanisms for both sexes, focussing on hormonal events throughout the lifespan that may influence female susceptibility to AD neuropathology and dementia onset. We further consider common non-modifiable (genetic) and modifiable (lifestyle and health) risk factors, highlighting possible sex-dependent differential effects for the AD disease course. Finally, we evaluate the studies selected for this review demonstrating sex-differences in cognitive, pathological and health factors, summarising the state of sex differences in AD risk factors. We further provide recommendations for targeted research on female-specific risk factors, to inform personalised strategies for AD-prevention and the promotion of female brain health.
Collapse
|
44
|
Zhang T. A Modeling and Machine Learning Pipeline to Rationally Design Treatments to Restore Neuroendocrine Disorders in Heterogeneous Individuals. Front Genet 2021; 12:656508. [PMID: 34567056 PMCID: PMC8458900 DOI: 10.3389/fgene.2021.656508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 08/11/2021] [Indexed: 11/16/2022] Open
Abstract
Heterogeneity among individual patients presents a fundamental challenge to effective treatment, since a treatment protocol working for a portion of the population often fails in others. We hypothesize that a computational pipeline integrating mathematical modeling and machine learning could be used to address this fundamental challenge and facilitate the optimization of individualized treatment protocols. We tested our hypothesis with the neuroendocrine systems controlled by the hypothalamic–pituitary–adrenal (HPA) axis. With a synergistic combination of mathematical modeling and machine learning (ML), this integrated computational pipeline could indeed efficiently reveal optimal treatment targets that significantly contribute to the effective treatment of heterogeneous individuals. What is more, the integrated pipeline also suggested quantitative information on how these key targets should be perturbed. Based on such ML revealed hints, mathematical modeling could be used to rationally design novel protocols and test their performances. We believe that this integrated computational pipeline, properly applied in combination with other computational, experimental and clinical research tools, can be used to design novel and improved treatment against a broad range of complex diseases.
Collapse
Affiliation(s)
- Tongli Zhang
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
45
|
Ding Y, Wei Z, Yan H, Guo W. Efficacy of Treatments Targeting Hypothalamic-Pituitary-Adrenal Systems for Major Depressive Disorder: A Meta-Analysis. Front Pharmacol 2021; 12:732157. [PMID: 34566653 PMCID: PMC8461240 DOI: 10.3389/fphar.2021.732157] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022] Open
Abstract
Abnormal hypothalamic-pituitary-adrenal (HPA) axis has been implicated in major depressive disorder (MDD). A number of studies have attempted to use HPA-modulating medications to treat depression. However, their results are inconsistent. The efficacy of these drugs for MDD remains uncertain. The aims of this meta-analysis were to determine the effect and safety profile of HPA-targeting medications for MDD. World of Science and PubMed databases were comprehensively searched up to March 2021. All randomized controlled trials (RCTs) and open-label trials exploring antiglucocorticoid and related medications in patients with depression were included. Standardized mean differences (SMDs) and risk ratios (RRs) with 95% confidence intervals (CIs) were calculated for continuous or dichotomous outcomes, respectively. In the meta-analysis, we identified 16 RCTs and seven open-label studies that included 2972 subjects. Pooling the change data that assessed the efficacy across all included HPA-targeting medications for depression showed a significant difference between interventions and controls with very small heterogeneity after influence analysis (SMD = 0.138, 95%CI = 0.052, 0.224, p = 0.002; I2 = 20.7%, p = 0.212). No obvious publication bias was observed (p = 0.127). Effectiveness remained significant in patients with MDD (SMD = 0.136, 95%CI = 0.049, 0.223, p = 0.002). Subgroup analysis showed a significant difference favoring mifepristone and vasopressin 1B (V1B) receptor antagonist treatment. Adverse events were reported by 14 studies and our analysis of high-quality studies showed a significant difference in favor of controls (RR = 1.283, 95%CI = 1.134, 1.452, p = 0). Our study suggested that patients with MDD may benefit from mifepristone and V1B receptor antagonist treatments that have tolerable side effects. HPA-based medications are promising for depression treatment. However, additional high-quality RCTs, including head-to-head trials, are needed. Systematic Review Registration:https://www.crd.york.ac.uk/PROSPERO/, identifier registration number: CRD42021247279
Collapse
Affiliation(s)
- Yudan Ding
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zirou Wei
- Mental Health Center, The Second Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Haohao Yan
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wenbin Guo
- National Clinical Research Center for Mental Disorders, Department of Psychiatry, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
46
|
Oda S, Ashida K, Uchiyama M, Sakamoto S, Hasuzawa N, Nagayama A, Wang L, Nagata H, Sakamoto R, Kishimoto J, Todaka K, Ogawa Y, Nakanishi Y, Nomura M. An Open-label Phase I/IIa Clinical Trial of 11β-HSD1 Inhibitor for Cushing's Syndrome and Autonomous Cortisol Secretion. J Clin Endocrinol Metab 2021; 106:e3865-e3880. [PMID: 34143883 DOI: 10.1210/clinem/dgab450] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitors demonstrate antimetabolic and antisarcopenic effects in Cushing's syndrome (CS) and autonomous cortisol secretion (ACS) patients. OBJECTIVE To confirm the efficacy and safety of S-707106 (11β-HSD1 inhibitor) administered to CS and ACS patients. DESIGN A 24-week single-center, open-label, single-arm, dose-escalation, investigator-initiated clinical trial on a database. SETTING Kyushu University Hospital, Kurume University Hospital, and related facilities. PATIENTS Sixteen patients with inoperable or recurrent CS and ACS, with mildly impaired glucose tolerance. INTERVENTION Oral administration of 200 mg S-707106 after dinner, daily, for 24 weeks. In patients with insufficient improvement in oral glucose tolerance test results at 12 weeks, an escalated dose of S-707106 (200 mg twice daily) was administered for the residual 12 weeks. MAIN OUTCOME MEASURES The rate of participants responding to glucose tolerance impairment, defined as those showing a 25% reduction in the area under the curve (AUC) of plasma glucose during the 75-g oral glucose tolerance test at 24 weeks. RESULTS S-707106 administration could not achieve the primary endpoint of this clinical trial (>20% of responsive participants). AUC glucose decreased by -7.1% [SD, 14.8 (90% CI -14.8 to -1.0), P = 0.033] and -2.7% [14.5 (-10.2 to 3.4), P = 0.18] at 12 and 24 weeks, respectively. S-707106 administration decreased AUC glucose significantly in participants with a high body mass index. Body fat percentage decreased by -2.5% [1.7 (-3.3 to -1.8), P < 0.001] and body muscle percentage increased by 2.4% [1.6 (1.7 to 3.1), P < 0.001]. CONCLUSIONS S-707106 is an effective insulin sensitizer and antisarcopenic and antiobesity medication for these patients.
Collapse
Affiliation(s)
- Satoko Oda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka-city, Japan
| | - Kenji Ashida
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka-city, Japan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume-city, Japan
| | - Makiko Uchiyama
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka-city, Japan
| | - Shohei Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka-city, Japan
| | - Nao Hasuzawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka-city, Japan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume-city, Japan
| | - Ayako Nagayama
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume-city, Japan
| | - Lixiang Wang
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka-city, Japan
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume-city, Japan
| | - Hiromi Nagata
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka-city, Japan
| | - Ryuichi Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka-city, Japan
| | - Junji Kishimoto
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka-city, Japan
| | - Koji Todaka
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka-city, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka-city, Japan
| | - Yoichi Nakanishi
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka-city, Japan
| | - Masatoshi Nomura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka-city, Japan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kurume University School of Medicine, Kurume-city, Japan
| |
Collapse
|
47
|
Younes N, Bourdeau I, Lacroix A. Latent Adrenal Insufficiency: From Concept to Diagnosis. Front Endocrinol (Lausanne) 2021; 12:720769. [PMID: 34512551 PMCID: PMC8429826 DOI: 10.3389/fendo.2021.720769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/09/2021] [Indexed: 11/22/2022] Open
Abstract
Primary adrenal insufficiency (PAI) is a rare disease and potentially fatal if unrecognized. It is characterized by destruction of the adrenal cortex, most frequently of autoimmune origin, resulting in glucocorticoid, mineralocorticoid, and adrenal androgen deficiencies. Initial signs and symptoms can be nonspecific, contributing to late diagnosis. Loss of zona glomerulosa function may precede zona fasciculata and reticularis deficiencies. Patients present with hallmark manifestations including fatigue, weight loss, abdominal pain, melanoderma, hypotension, salt craving, hyponatremia, hyperkalemia, or acute adrenal crisis. Diagnosis is established by unequivocally low morning serum cortisol/aldosterone and elevated ACTH and renin concentrations. A standard dose (250 µg) Cosyntropin stimulation test may be needed to confirm adrenal insufficiency (AI) in partial deficiencies. Glucocorticoid and mineralocorticoid substitution is the hallmark of treatment, alongside patient education regarding dose adjustments in periods of stress and prevention of acute adrenal crisis. Recent studies identified partial residual adrenocortical function in patients with AI and rare cases have recuperated normal hormonal function. Modulating therapies using rituximab or ACTH injections are in early stages of investigation hoping it could maintain glucocorticoid residual function and delay complete destruction of adrenal cortex.
Collapse
Affiliation(s)
| | | | - Andre Lacroix
- Division of Endocrinology, Department of Medicine and Research Center, Centre Hospitalier de l’Université de Montréal (CHUM), Montréal, QC, Canada
| |
Collapse
|
48
|
Liu PY, Lawrence-Sidebottom D, Piotrowska K, Zhang W, Iranmanesh A, Auchus RJ, Veldhuis JD, Van Dongen HPA. Clamping Cortisol and Testosterone Mitigates the Development of Insulin Resistance during Sleep Restriction in Men. J Clin Endocrinol Metab 2021; 106:e3436-e3448. [PMID: 34043794 PMCID: PMC8660069 DOI: 10.1210/clinem/dgab375] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Indexed: 01/04/2023]
Abstract
CONTEXT Sleep loss in men increases cortisol and decreases testosterone, and sleep restriction by 3 to 4 hours/night induces insulin resistance. OBJECTIVE We clamped cortisol and testosterone and determined the effect on insulin resistance. METHODS This was a randomized double-blind, in-laboratory crossover study in which 34 healthy young men underwent 4 nights of sleep restriction of 4 hours/night under 2 treatment conditions in random order: dual hormone clamp (cortisol and testosterone fixed), or matching placebo (cortisol and testosterone not fixed). Fasting blood samples, and an additional 23 samples for a 3-hour oral glucose tolerance test (OGTT), were collected before and after sleep restriction under both treatment conditions. Cytokines and hormones were measured from the fasting samples. Overall insulin sensitivity was determined from the OGTT by combining complementary measures: homeostasis model assessment of insulin resistance of the fasting state; Matsuda index of the absorptive state; and minimal model of both fasting and absorptive states. RESULTS Sleep restriction alone induced hyperinsulinemia, hyperglycemia, and overall insulin resistance (P < 0.001 for each). Clamping cortisol and testosterone alleviated the development of overall insulin resistance (P = 0.046) and hyperinsulinemia (P = 0.014) by 50%. Interleukin-6, high-sensitivity C-reactive protein, peptide YY, and ghrelin did not change, whereas tumor necrosis factor-α and leptin changed in directions that would have mitigated insulin resistance with sleep restriction alone. CONCLUSION Fixing cortisol-testosterone exposure mitigates the development of insulin resistance and hyperinsulinemia, but not hyperglycemia, from sustained sleep restriction in men. The interplay between cortisol and testosterone may be important as a mechanism by which sleep restriction impairs metabolic health.
Collapse
Affiliation(s)
- Peter Y Liu
- Division of Endocrinology, The Lundquist Institute at Harbor UCLA Medical Center, Torrance, CA, USA
- David Geffen School of Medicine, University of California—Los Angeles, Los Angeles, CA, USA
| | - Darian Lawrence-Sidebottom
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
- Neuroscience Graduate Program, Washington State University, Pullman, WA, USA
| | - Katarzyna Piotrowska
- Division of Endocrinology, The Lundquist Institute at Harbor UCLA Medical Center, Torrance, CA, USA
| | - Wenyi Zhang
- Division of Endocrinology, The Lundquist Institute at Harbor UCLA Medical Center, Torrance, CA, USA
| | - Ali Iranmanesh
- Endocrinology Service, VA Medical Center, Salem, VA, USA
| | - Richard J Auchus
- Division of Metabolism, Diabetes, and Endocrinology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Johannes D Veldhuis
- Endocrine Research Unit, Mayo School of Graduate Medical Education, Center for Translational Science Activities, Mayo Clinic, Rochester, MN, USA
| | - Hans P A Van Dongen
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| |
Collapse
|
49
|
Williams SJ, Meadows R, Coveney CM. Desynchronised times? Chronobiology, (bio)medicalisation and the rhythms of life itself. SOCIOLOGY OF HEALTH & ILLNESS 2021; 43:1501-1517. [PMID: 34254324 DOI: 10.1111/1467-9566.13324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/26/2021] [Accepted: 06/07/2021] [Indexed: 06/13/2023]
Abstract
This paper takes a critical look at the role of chronobiology in society today, with particular reference to its entanglements with health and medicine and whether or not this amounts to the (bio)medicalisation of our bodily rhythms. What we have here, we show, is a complex unfolding storyline, within and beyond medicine. On the one hand, the promises and problems of these circadian, infradian and ultradian rhythms for our health and well-being are now increasingly emphasised. On the other hand, a variety of new rhythmic interventions and forms of governance are now emerging within and beyond medicine, from chronotherapies and chronopharmacology to biocompatible school and work schedules, and from chronodiets to the optimisation of all we do according to our 'chronotypes'. Conceptualising these developments, we suggest challenges us to think within and beyond medicalisation to wider processes of biomedicalisation and the biopolitics of our body clocks: a vital new strand of chronopolitics today indeed which implicates us all in sickness and in health as the very embodiment of these rhythms of life itself. The paper concludes with a call for further research on these complex unfolding relations between chronobiology, health and society in these desynchronised times of ours.
Collapse
Affiliation(s)
| | - Robert Meadows
- Department of Sociology, University of Surrey, Surrey, UK
| | - Catherine M Coveney
- Social and Policy Studies, School of Social Sciences and Humanities, Loughborough University, London, UK
| |
Collapse
|
50
|
O'Byrne NA, Yuen F, Butt WZ, Liu PY. Sleep and Circadian Regulation of Cortisol: A Short Review. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2021; 18:178-186. [PMID: 35128146 DOI: 10.1016/j.coemr.2021.03.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The central circadian pacemaker (CCP) located in the suprachiasmatic nucleus (SCN) of the hypothalamus drives the 24-hour pattern in cortisol, which functions as the main central synchronizing signal that coordinates peripheral clocks in organs that control whole body metabolism. A superimposed pulsatile pattern of cortisol allows rapid responses that fine tune the body's reaction to changes in the environment. In addition to coordinating metabolic processes to predictable environmental events, cortisol is the main catabolic signal which acts with testosterone, the quintessential male anabolic hormone, to maintain catabolic-anabolic homeostasis in men. Sleep restriction, when sufficiently substantial, increases late afternoon/early evening cortisol, but does not alter 24-hour cortisol; whereas even maximal acute circadian misalignment only slightly delays the cortisol rhythm. Prolonged circadian misalignment decreases overall cortisol exposure. The implications of these regulatory changes on health and disease requires further evaluation.
Collapse
Affiliation(s)
- Nora A O'Byrne
- The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Fiona Yuen
- The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Waleed Z Butt
- The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Peter Y Liu
- The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA.,Department of Medicine, Division of Endocrinology, David Geffen School of Medicine at UCLA
| |
Collapse
|