1
|
Mualem R, Morales-Quezada L, Farraj RH, Shance S, Bernshtein DH, Cohen S, Mualem L, Salem N, Yehuda RR, Zbedat Y, Waksman I, Biswas S. Econeurobiology and brain development in children: key factors affecting development, behavioral outcomes, and school interventions. Front Public Health 2024; 12:1376075. [PMID: 39391155 PMCID: PMC11465878 DOI: 10.3389/fpubh.2024.1376075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/29/2024] [Indexed: 10/12/2024] Open
Abstract
The Econeurobiology of the brain describes the environment in which an individual's brain develops. This paper explores the complex neural mechanisms that support and evaluate enrichment at various stages of development, providing an overview of how they contribute to plasticity and enhancement of both achievement and health. It explores the deep benefits of enrichment and contrasts them with the negative effects of trauma and stress on brain development. In addition, the paper strongly emphasizes the integration of Gardner's intelligence types into the school curriculum environment. It emphasizes the importance of linking various intelligence traits to educational strategies to ensure a holistic approach to cognitive development. In the field of Econeurobiology, this work explains the central role of the environment in shaping the development of the brain. It examines brain connections and plasticity and reveals the impact of certain environmental factors on brain development in early and mid-childhood. In particular, the six key factors highlighted are an environment of support, nutrition, physical activity, music, sleep, and cognitive strategies, highlighting their potential to improve cognitive abilities, memory, learning, self-regulation, and social and emotional development. This paper also investigates the social determinants of health and education in the context of Econeurobiology. It emphasizes the transformative power of education in society, especially in vulnerable communities facing global challenges in accessing quality education.
Collapse
Affiliation(s)
- Raed Mualem
- Department of Natural and Environmental Sciences, Faculty of Education, Oranim Academic College, Kiryat Tiv'on, Israel
- The Institute for Brain and Rehabilitation Sciences, Nazareth, Israel
- Econeurobiology Research Group, Research Authority, Oranim Academic College, Kiryat Tiv'on, Israel
- Ramat Zevulun High School, Ibtin, Israel
| | - Leon Morales-Quezada
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA, United States
| | - Rania Hussein Farraj
- Econeurobiology Research Group, Research Authority, Oranim Academic College, Kiryat Tiv'on, Israel
| | - Shir Shance
- The Institute for Brain and Rehabilitation Sciences, Nazareth, Israel
- Econeurobiology Research Group, Research Authority, Oranim Academic College, Kiryat Tiv'on, Israel
| | | | - Sapir Cohen
- Econeurobiology Research Group, Research Authority, Oranim Academic College, Kiryat Tiv'on, Israel
| | - Loay Mualem
- Department of Computer Science, Haifa University, Haifa, Israel
| | - Niven Salem
- The Institute for Brain and Rehabilitation Sciences, Nazareth, Israel
| | - Rivka Riki Yehuda
- The Institute for Brain and Rehabilitation Sciences, Nazareth, Israel
| | | | - Igor Waksman
- Bar Ilan University Medical School, Tzfat, Israel
| | - Seema Biswas
- Global Health Research Laboratory, Department of Surgery B, Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
2
|
Suda M, Paul KH, Tripathi U, Minamino T, Tchkonia T, Kirkland JL. Targeting Cell Senescence and Senolytics: Novel Interventions for Age-Related Endocrine Dysfunction. Endocr Rev 2024; 45:655-675. [PMID: 38500373 PMCID: PMC11405506 DOI: 10.1210/endrev/bnae010] [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: 08/23/2023] [Revised: 01/11/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Multiple changes occur in hormonal regulation with aging and across various endocrine organs. These changes are associated with multiple age-related disorders and diseases. A better understanding of responsible underling biological mechanisms could help in the management of multiple endocrine disorders over and above hormone replacement therapy (HRT). Cellular senescence is involved in multiple biological aging processes and pathologies common in elderly individuals. Cellular senescence, which occurs in many older individuals but also across the lifespan in association with tissue damage, acute and chronic diseases, certain drugs, and genetic syndromes, may contribute to such endocrine disorders as osteoporosis, metabolic syndrome, and type 2 diabetes mellitus. Drugs that selectively induce senescent cell removal, "senolytics,", and drugs that attenuate the tissue-destructive secretory state of certain senescent cells, "senomorphics," appear to delay the onset of or alleviate multiple diseases, including but not limited to endocrine disorders such as diabetes, complications of obesity, age-related osteoporosis, and cancers as well as atherosclerosis, chronic kidney disease, neurodegenerative disorders, and many others. More than 30 clinical trials of senolytic and senomorphic agents have already been completed, are underway, or are planned for a variety of indications. Targeting senescent cells is a novel strategy that is distinct from conventional therapies such as HRT, and thus might address unmet medical needs and can potentially amplify effects of established endocrine drug regimens, perhaps allowing for dose decreases and reducing side effects.
Collapse
Affiliation(s)
- Masayoshi Suda
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Karl H Paul
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Utkarsh Tripathi
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan
| | - Tamara Tchkonia
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
3
|
Misiura M, Munkombwe C, Igwe K, Verble DD, Likos KDS, Minto L, Bartlett A, Zetterberg H, Turner JA, Dotson VM, Brickman AM, Hu WT, Wharton W. Neuroimaging correlates of Alzheimer's disease biomarker concentrations in a racially diverse high-risk cohort of middle-aged adults. Alzheimers Dement 2024; 20:5961-5972. [PMID: 39136298 PMCID: PMC11497767 DOI: 10.1002/alz.14051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/24/2024] [Accepted: 05/15/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION In this study, we investigated biomarkers in a midlife, racially diverse, at-risk cohort to facilitate early identification and intervention. We examined neuroimaging measures, including resting state functional magnetic resonance imaging (fMRI), white matter hyperintensity vo (WMH), and hippocampal volumes, alongside cerebrospinal fluid (CSF) markers. METHODS Our data set included 76 cognitively unimpaired, middle-aged, Black Americans (N = 29, F/M = 17/12) and Non-Hispanic White (N = 47, F/M = 27/20) individuals. We compared cerebrospinal fluid phosphorylated tau141 and amyloid beta (Aβ)42 to fMRI default mode network (DMN) subnetwork connectivity, WMH volumes, and hippocampal volumes. RESULTS Results revealed a significant race × Aβ42 interaction in Black Americans: lower Aβ42 was associated with reduced DMN connectivity and increased WMH volumes regions but not in non-Hispanic White individuals. DISCUSSION Our findings suggest that precuneus DMN connectivity and temporal WMHs may be linked to Alzheimer's disease risk pathology during middle age, particularly in Black Americans. HIGHLIGHTS Cerebrospinal fluid (CSF) amyloid beta (Aβ)42 relates to precuneus functional connectivity in Black, but not White, Americans. Higher white matter hyperintensity volume relates to lower CSF Aβ42 in Black Americans. Precuneus may be a hub for early Alzheimer's disease pathology changes detected by functional connectivity.
Collapse
Affiliation(s)
- Maria Misiura
- Department of PsychologyGeorgia State UniversityAtlantaGeorgiaUSA
- Tri‐Institutional Center for Translational Research in Neuroimaging & Data Science, Georgia State University, Georgia Institute of TechnologyEmory UniversityAtlantaGeorgiaUSA
| | | | - Kay Igwe
- Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, and Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Danielle D. Verble
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| | - Kelly D. S. Likos
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| | - Lex Minto
- Department of PsychologyGeorgia State UniversityAtlantaGeorgiaUSA
| | | | - Henrik Zetterberg
- The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and GothenburgUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative Disease, UCL Institute of NeurologyUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCL, Maple HouseLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Jessica A. Turner
- Department of Psychiatry and Mental Health, College of MedicineOhio State UniversityColumbusOhioUSA
| | - Vonetta M. Dotson
- Department of PsychologyGeorgia State UniversityAtlantaGeorgiaUSA
- Gerontology DepartmentGeorgia State UniversityAtlantaGeorgiaUSA
| | - Adam M. Brickman
- Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, and Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - William T. Hu
- Institute for Health, Health Care Policy, and Aging ResearchRutgers UniversityNew BrunswickNew JerseyUSA
| | - Whitney Wharton
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
4
|
Liu M, Zhao W, Zhang W, Tian Z, Zhang Z, Lou Y, Zhang Z, Yang F, Zhu D, Li P. Comparable cognitive impairment was detected in MACS and CS and alleviated after remission of hypercortisolism in MACS. Front Endocrinol (Lausanne) 2024; 15:1373101. [PMID: 39145316 PMCID: PMC11322108 DOI: 10.3389/fendo.2024.1373101] [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: 01/19/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Context Few studies have directly compared the cognitive characteristics of patients with mild autonomous cortisol secretion (MACS) and Cushing's syndrome (CS). The effect of surgical or conservative treatment on cognitive function in patients with MACS is still unclear. Objective To compare the differences in cognitive function between patients with MACS and CS and evaluate the effect of surgery or conservative treatment on cognitive function. Methods We prospectively recruited 59 patients with nonfunctional adrenal adenoma (NFA), 36 patients with MACS, and 20 patients with adrenal CS who completed the global cognition and cognitive subdomains assessments. Seventeen MACS patients were re-evaluated for cognitive function after a 12-month follow-up period; of these, eleven underwent laparoscopic adrenalectomy and six received conservative treatment. Results Patients with MACS and CS performed worse in the global cognition and multiple cognitive domains than those with NFA (all P<0.05). No statistical difference was found in cognitive functions between patients with MACS and CS. Logistic regression analysis showed that patients with MACS (odds ratio [OR]=3.738, 95% confidence intervals [CI]: 1.329-10.515, P=0.012) and CS (OR=6.026, 95% CI: 1.411-25.730, P=0.015) were associated with an increased risk of immediate memory impairment. Visuospatial/constructional, immediate and delayed memory scores of MACS patients were significantly improved at 12 months compared with pre-operation in the surgical treatment group (all P<0.05), whereas there was no improvement in the conservative treatment group. Conclusion Patients with MACS have comparable cognitive impairment as patients with CS. Cognitive function was partially improved in patients with MACS after adrenalectomy. The current data support the inclusion of cognitive function assessment in the clinical management of patients with MACS.
Collapse
Affiliation(s)
- Mengsi Liu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
- Nanjing Drum Tower Hospital, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Wenji Zhao
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Drum Tower Hospital, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Wei Zhang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Drum Tower Hospital, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Zhaoyang Tian
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Drum Tower Hospital, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Zhou Zhang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Drum Tower Hospital, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Yuan Lou
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Drum Tower Hospital, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Ziwei Zhang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Drum Tower Hospital, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Fan Yang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Drum Tower Hospital, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Dalong Zhu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
- Nanjing Drum Tower Hospital, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Ping Li
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Nanjing Drum Tower Hospital, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| |
Collapse
|
5
|
Singh J, Vanlallawmzuali, Singh A, Biswal S, Zomuansangi R, Lalbiaktluangi C, Singh BP, Singh PK, Vellingiri B, Iyer M, Ram H, Udey B, Yadav MK. Microbiota-brain axis: Exploring the role of gut microbiota in psychiatric disorders - A comprehensive review. Asian J Psychiatr 2024; 97:104068. [PMID: 38776563 DOI: 10.1016/j.ajp.2024.104068] [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: 11/22/2023] [Revised: 02/28/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Mental illness is a hidden epidemic in modern science that has gradually spread worldwide. According to estimates from the World Health Organization (WHO), approximately 10% of the world's population suffers from various mental diseases each year. Worldwide, financial and health burdens on society are increasing annually. Therefore, understanding the different factors that can influence mental illness is required to formulate novel and effective treatments and interventions to combat mental illness. Gut microbiota, consisting of diverse microbial communities residing in the gastrointestinal tract, exert profound effects on the central nervous system through the gut-brain axis. The gut-brain axis serves as a conduit for bidirectional communication between the two systems, enabling the gut microbiota to affect emotional and cognitive functions. Dysbiosis, or an imbalance in the gut microbiota, is associated with an increased susceptibility to mental health disorders and psychiatric illnesses. Gut microbiota is one of the most diverse and abundant groups of microbes that have been found to interact with the central nervous system and play important physiological functions in the human gut, thus greatly affecting the development of mental illnesses. The interaction between gut microbiota and mental health-related illnesses is a multifaceted and promising field of study. This review explores the mechanisms by which gut microbiota influences mental health, encompassing the modulation of neurotransmitter production, neuroinflammation, and integrity of the gut barrier. In addition, it emphasizes a thorough understanding of how the gut microbiome affects various psychiatric conditions.
Collapse
Affiliation(s)
- Jawahar Singh
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bathinda, Punjab, India
| | - Vanlallawmzuali
- Department of Biotechnology, Mizoram Central University, Pachhunga University College Campus, Aizawl, Mizoram, India
| | - Amit Singh
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Suryanarayan Biswal
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Ruth Zomuansangi
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - C Lalbiaktluangi
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Bhim Pratap Singh
- Department of Agriculture and Environmental Sciences (AES), National Institute of Food Technology Entrepreneurship and Management (NIFTEM), Sonepat, Haryana, India
| | - Prashant Kumar Singh
- Department of Biotechnology, Pachhunga University College Campus, Mizoram University (A Central University), Aizawl 796001, Mizoram, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab 151401, India
| | - Mahalaxmi Iyer
- Department of Microbiology Central University of Punjab, Bathinda 151401, India
| | - Heera Ram
- Department of Zoology, Jai Narain Vyas University, Jodhpur, Rajasthan 342001, India
| | - Bharat Udey
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Bathinda, Punjab, India
| | - Mukesh Kumar Yadav
- Department of Microbiology Central University of Punjab, Bathinda 151401, India.
| |
Collapse
|
6
|
Lopez M, Siedl A, Phillips KA. Cortisol levels across the lifespan in common marmosets (Callithrix jacchus). Am J Primatol 2024; 86:e23597. [PMID: 38239052 PMCID: PMC10959686 DOI: 10.1002/ajp.23597] [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: 09/11/2023] [Revised: 12/13/2023] [Accepted: 12/28/2023] [Indexed: 02/13/2024]
Abstract
Human aging is associated with senescence of the hypothalamic-pituitary-adrenal (HPA) axis, leading to progressive dysregulation characterized by increased cortisol exposure. This key hormone is implicated in the pathogenesis of many age-related diseases. Common marmosets (Callithrix jacchus) display a wide spectrum of naturally occurring age-related pathologies that compare similarly to humans and are increasingly used as translational models of aging and age-related disease. Whether the marmoset HPA axis also shows senescence with increasing age is unknown. We analyzed hair cortisol concentration (HCC) across the lifespan of 50 captive common marmosets, ranging in age from approximately 2 months-14.5 years, via a cross-sectional design. Samples were processed and analyzed for cortisol using enzyme immunoassay. HCC ranged from 1416 to 15,343 pg/mg and was negatively correlated with age. We found significant main effects of age group (infant, adolescent, adult, aged, very aged) and sex on HCC, and no interaction effects. Infants had significantly higher levels of HCC compared with all other age groups. Females had higher HCC than males. There was no interaction between age and sex. These results suggest marmosets do not show dysregulation of the HPA axis with increasing age, as measured via HCC.
Collapse
Affiliation(s)
- Matthew Lopez
- Department of Psychology, Trinity University, 607 Kings Court, San Antonio Texas, USA
| | - Amaya Siedl
- Department of Psychology, Trinity University, 607 Kings Court, San Antonio Texas, USA
| | - Kimberley A. Phillips
- Department of Psychology, Trinity University, 607 Kings Court, San Antonio Texas, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas USA
| |
Collapse
|
7
|
Suba-Bokodi É, Nagy I, Molnár M. The Impact of Transportation on the Cortisol Level of Dwarf Rabbits Bred to Animal-Assisted Interventions. Animals (Basel) 2024; 14:664. [PMID: 38473049 DOI: 10.3390/ani14050664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/29/2024] [Accepted: 02/17/2024] [Indexed: 03/14/2024] Open
Abstract
(1) Background: the popularity of rabbits has increased during the last decade and become the third most common companion animal in the EU. Rabbits' participation in Animal-Assisted Interventions (AAIs) is growing. It is highly important to ensure the well-being of the animals in AAIs. Whereas the needs and the advantages of people involved in AAI are becoming more and more evident, the needs of animals are not clearly defined, therefore, it is a great field of inquiry. Animals who are used for AAI need to be transported regularly, which itself might be a source of stress. (2) Methods: the stress of rabbits-caused by transportation-was measured in a non-invasive way: cortisol levels were determined from feces, based on their breakdown products. Eighteen animals were involved in the study. Rabbits experienced a 30 min transportation every second day for two weeks (altogether six times) while 126 samples were collected. (3) Results: rabbits could handle the transportation procedure the first time but subsequently the stress hormone metabolites in feces samples increased regardless of the offered treatments (hay, carrot and apple) during the carriage. (4) Conclusions: those owners who use rabbits for Animal-Assisted Interventions need to take into account that transportation itself is a stressful experience for the animals.
Collapse
Affiliation(s)
- Éva Suba-Bokodi
- Institute of Animal Husbandry, Kaposvár Campus, Hungarian University of Agriculture and Life Sciences, 40. Guba S. u., 7400 Kaposvár, Hungary
| | - István Nagy
- Institute of Animal Husbandry, Kaposvár Campus, Hungarian University of Agriculture and Life Sciences, 40. Guba S. u., 7400 Kaposvár, Hungary
| | - Marcell Molnár
- Institute of Animal Husbandry, Kaposvár Campus, Hungarian University of Agriculture and Life Sciences, 40. Guba S. u., 7400 Kaposvár, Hungary
| |
Collapse
|
8
|
Ash H, Goy RW, Spaulding A, Colman RJ, Corbett CJ, Ziegler TE. Cognitive development from infancy to young adulthood in common marmosets (Callithrix jacchus): Effect of age, sex, and hormones on learning and affective state. Dev Psychobiol 2023; 65:e22430. [PMID: 37860906 PMCID: PMC10804839 DOI: 10.1002/dev.22430] [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: 11/11/2022] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/21/2023]
Abstract
Studies looking at individual variability in cognition have increased in recent years. We followed 43 marmosets (21 males, 22 females) from infancy to young adulthood. At 3-months old, marmosets were trained to touch a rewarded stimulus. At 9-, 15-, and 21-months old, they were given visual discrimination and cognitive bias tests, and urine samples were collected to examine hormone levels. Marmosets were significantly more successful learners at 15 months than 9 months. Individuals who were more successful learners at 9 months were also more successful at 15 months, with more male learners than expected at 15 months. At 9 months, learning success was associated with higher cortisol levels. At 15 months, males with higher estradiol levels were more successful learners, whereas at 21 months, females with higher estradiol and cortisol levels tended to be less successful learners and more pessimistic. Nine months, therefore, appears to be an important developmental timepoint for acquiring cognitive control, which has developed by 15 months. Steroids may have differential effects on each sex, with complex interactions between gonadal and adrenal hormones having an influence on cognitive function over the lifespan. This longitudinal study offers new insight into cognition, including its development and biological underpinnings.
Collapse
Affiliation(s)
- Hayley Ash
- Wisconsin National Primate Research Center (WNPRC), University of Wisconsin, Madison WI, United States
| | - Robinson W. Goy
- Wisconsin National Primate Research Center (WNPRC), University of Wisconsin, Madison WI, United States
| | - Abigail Spaulding
- Wisconsin National Primate Research Center (WNPRC), University of Wisconsin, Madison WI, United States
| | - Ricki J. Colman
- Wisconsin National Primate Research Center (WNPRC), University of Wisconsin, Madison WI, United States
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison WI, United States
| | - Cody J. Corbett
- Wisconsin National Primate Research Center (WNPRC), University of Wisconsin, Madison WI, United States
| | - Toni E. Ziegler
- Wisconsin National Primate Research Center (WNPRC), University of Wisconsin, Madison WI, United States
| |
Collapse
|
9
|
Zhang J, Ge P, Liu J, Luo Y, Guo H, Zhang G, Xu C, Chen H. Glucocorticoid Treatment in Acute Respiratory Distress Syndrome: An Overview on Mechanistic Insights and Clinical Benefit. Int J Mol Sci 2023; 24:12138. [PMID: 37569514 PMCID: PMC10418884 DOI: 10.3390/ijms241512138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS), triggered by various pathogenic factors inside and outside the lungs, leads to diffuse lung injury and can result in respiratory failure and death, which are typical clinical critical emergencies. Severe acute pancreatitis (SAP), which has a poor clinical prognosis, is one of the most common diseases that induces ARDS. When SAP causes the body to produce a storm of inflammatory factors and even causes sepsis, clinicians will face a two-way choice between anti-inflammatory and anti-infection objectives while considering the damaged intestinal barrier and respiratory failure, which undoubtedly increases the difficulty of the diagnosis and treatment of SAP-ALI/ARDS. For a long time, many studies have been devoted to applying glucocorticoids (GCs) to control the inflammatory response and prevent and treat sepsis and ALI/ARDS. However, the specific mechanism is not precise, the clinical efficacy is uneven, and the corresponding side effects are endless. This review discusses the mechanism of action, current clinical application status, effectiveness assessment, and side effects of GCs in the treatment of ALI/ARDS (especially the subtype caused by SAP).
Collapse
Affiliation(s)
- Jinquan Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Peng Ge
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jie Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yalan Luo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Haoya Guo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Guixin Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Caiming Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Comprehensive Cancer Center, Monrovia, CA 91016, USA
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
10
|
Lee Y, Lee S, Lee W. Occupational and Environmental Noise Exposure and Extra-Auditory Effects on Humans: A Systematic Literature Review. GEOHEALTH 2023; 7:e2023GH000805. [PMID: 37303697 PMCID: PMC10248481 DOI: 10.1029/2023gh000805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 06/13/2023]
Abstract
Noise is a common harmful factor in our work and the environment. Most studies have investigated the auditory effects of noise exposure; however, few studies have focused on the extra-auditory effects of exposure to occupational or environmental noise. This study aimed to systematically review published studies on the extra-auditory effects of noise exposure. We reviewed literature from PubMed and Google Scholar databases up to July 2022, using the Patient, Intervention, Comparison, and Outcome criteria and Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines to identify studies that reported extra-auditory effects of occupational or environmental noise exposure. Studies were evaluated utilizing validated reporting tools (CONSORT, STROBE) appropriate to study design. A total of 263 articles were identified, of which 36 were finally selected and reviewed. Upon conducting a review of the articles, exposure to noise can elicit a variety of extra-auditory effects on humans. These effects include circulatory effects linked to higher risk of cardiovascular disease and decreased endothelial function, nervous system effects correlated with sleep disturbance, cognitive impairment, and mental health problems, immunological and endocrinal effects connected to increased physiological stress response and metabolic disorders, oncological and respiratory effects associated with an elevated risk of acoustic neuroma and respiratory disorders, gastrointestinal effects linked to an increased risk of gastric or duodenal ulcer, and obstetric effects connected to the risk of preterm birth. Our review suggests that there are numerous extra-auditory effects of noise exposure on human, and further investigations are needed to fully understand these effects.
Collapse
Affiliation(s)
- Yongho Lee
- Department of Occupational and Environmental MedicineGil Medical CenterIncheonRepublic of Korea
| | - Seunghyun Lee
- Department of Occupational and Environmental MedicineGachon University College of MedicineIncheonRepublic of Korea
| | - Wanhyung Lee
- Department of Occupational and Environmental MedicineGil Medical CenterIncheonRepublic of Korea
- Department of Occupational and Environmental MedicineGachon University College of MedicineIncheonRepublic of Korea
| |
Collapse
|
11
|
Ju LS, Morey TE, Seubert CN, Martynyuk AE. Intergenerational Perioperative Neurocognitive Disorder. BIOLOGY 2023; 12:biology12040567. [PMID: 37106766 PMCID: PMC10135810 DOI: 10.3390/biology12040567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023]
Abstract
Accelerated neurocognitive decline after general anesthesia/surgery, also known as perioperative neurocognitive disorder (PND), is a widely recognized public health problem that may affect millions of patients each year. Advanced age, with its increasing prevalence of heightened stress, inflammation, and neurodegenerative alterations, is a consistent contributing factor to the development of PND. Although a strong homeostatic reserve in young adults makes them more resilient to PND, animal data suggest that young adults with pathophysiological conditions characterized by excessive stress and inflammation may be vulnerable to PND, and this altered phenotype may be passed to future offspring (intergenerational PND). The purpose of this narrative review of data in the literature and the authors' own experimental findings in rodents is to draw attention to the possibility of intergenerational PND, a new phenomenon which, if confirmed in humans, may unravel a big new population that may be affected by parental PND. In particular, we discuss the roles of stress, inflammation, and epigenetic alterations in the development of PND. We also discuss experimental findings that demonstrate the effects of surgery, traumatic brain injury, and the general anesthetic sevoflurane that interact to induce persistent dysregulation of the stress response system, inflammation markers, and behavior in young adult male rats and in their future offspring who have neither trauma nor anesthetic exposure (i.e., an animal model of intergenerational PND).
Collapse
Affiliation(s)
- Ling-Sha Ju
- Department of Anesthesiology, College of Medicine, University of Florida, P.O. Box 100254, JHMHC, 1600 SW Archer Road, Gainesville, FL 32610, USA
| | - Timothy E Morey
- Department of Anesthesiology, College of Medicine, University of Florida, P.O. Box 100254, JHMHC, 1600 SW Archer Road, Gainesville, FL 32610, USA
| | - Christoph N Seubert
- Department of Anesthesiology, College of Medicine, University of Florida, P.O. Box 100254, JHMHC, 1600 SW Archer Road, Gainesville, FL 32610, USA
| | - Anatoly E Martynyuk
- Department of Anesthesiology, College of Medicine, University of Florida, P.O. Box 100254, JHMHC, 1600 SW Archer Road, Gainesville, FL 32610, USA
- Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
12
|
Sharvin BL, Aburto MR, Cryan JF. Decoding the neurocircuitry of gut feelings: Region-specific microbiome-mediated brain alterations. Neurobiol Dis 2023; 179:106033. [PMID: 36758820 DOI: 10.1016/j.nbd.2023.106033] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Research in the last decade has unveiled a crucial role for the trillions of microorganisms that reside in the gut in influencing host neurodevelopment across the lifespan via the microbiota-gut-brain axis. Studies have linked alterations in the composition, complexity, and diversity of the gut microbiota to changes in behaviour including abnormal social interactions, cognitive deficits, and anxiety- and depressive-like phenotypes. Moreover, the microbiota has been linked with neurodevelopmental, neuropsychiatric, and neurodegenerative disorders. Interestingly, there appears to be specific brain regions governing the neurocircuitry driving higher cognitive function that are susceptible to influence from manipulations to the host microbiome. This review will aim to elucidate the region-specific effects mediated by the gut microbiota, with a focus on translational animal models and some existing human neuroimaging data. Compelling preclinical evidence suggests disruption to normal microbiota-gut-brain signalling can have detrimental effects on the prefrontal cortex, amygdala, hippocampus, hypothalamus, and striatum. Furthermore, human neuroimaging studies have unveiled a role for the microbiota in mediating functional connectivity and structure of specific brain regions that can be traced back to neurocognition and behavioural output. Understanding these microbiota-mediated changes will aid in identifying unique therapeutic targets for treating neurological disorders associated with these regions.
Collapse
Affiliation(s)
- Brendan L Sharvin
- APC Microbiome, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Maria Rodriguez Aburto
- APC Microbiome, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
13
|
James KA, Stromin JI, Steenkamp N, Combrinck MI. Understanding the relationships between physiological and psychosocial stress, cortisol and cognition. Front Endocrinol (Lausanne) 2023; 14:1085950. [PMID: 36950689 PMCID: PMC10025564 DOI: 10.3389/fendo.2023.1085950] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/14/2023] [Indexed: 03/08/2023] Open
Abstract
Stress is viewed as a state of real or perceived threat to homeostasis, the management of which involves the endocrine, nervous, and immune systems. These systems work independently and interactively as part of the stress response. The scientific stress literature, which spans both animal and human studies, contains heterogeneous findings about the effects of stress on the brain and the body. This review seeks to summarise and integrate literature on the relationships between these systems, examining particularly the roles of physiological and psychosocial stress, the stress hormone cortisol, as controlled by the hypothalamic-pituitary-adrenal (HPA) axis, and the effects of stress on cognitive functioning. Health conditions related to impaired HPA axis functioning and their associated neuropsychiatric symptoms will also be considered. Lastly, this review will provide suggestions of clinical applicability for endocrinologists who are uniquely placed to measure outcomes related to endocrine, nervous and immune system functioning and identify areas of intervention.
Collapse
Affiliation(s)
- Katharine Ann James
- Applied Cognitive Science and Experimental Neuropsychology Team (ACSENT) Laboratory, Department of Psychology, University of Cape Town, Cape Town, South Africa
- Division of Geriatric Medicine, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Juliet Ilena Stromin
- Applied Cognitive Science and Experimental Neuropsychology Team (ACSENT) Laboratory, Department of Psychology, University of Cape Town, Cape Town, South Africa
| | - Nina Steenkamp
- Applied Cognitive Science and Experimental Neuropsychology Team (ACSENT) Laboratory, Department of Psychology, University of Cape Town, Cape Town, South Africa
| | - Marc Irwin Combrinck
- Division of Geriatric Medicine, Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
14
|
Elevated Hippocampal CRMP5 Mediates Chronic Stress-Induced Cognitive Deficits by Disrupting Synaptic Plasticity, Hindering AMPAR Trafficking, and Triggering Cytokine Release. Int J Mol Sci 2023; 24:ijms24054898. [PMID: 36902337 PMCID: PMC10003309 DOI: 10.3390/ijms24054898] [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: 02/08/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Chronic stress is a critical risk factor for developing depression, which can impair cognitive function. However, the underlying mechanisms involved in chronic stress-induced cognitive deficits remain unclear. Emerging evidence suggests that collapsin response mediator proteins (CRMPs) are implicated in the pathogenesis of psychiatric-related disorders. Thus, the study aims to examine whether CRMPs modulate chronic stress-induced cognitive impairment. We used the chronic unpredictable stress (CUS) paradigm to mimic stressful life situations in C57BL/6 mice. In this study, we found that CUS-treated mice exhibited cognitive decline and increased hippocampal CRMP2 and CRMP5 expression. In contrast to CRMP2, CRMP5 levels strongly correlated with the severity of cognitive impairment. Decreasing hippocampal CRMP5 levels through shRNA injection rescued CUS-induced cognitive impairment, whereas increasing CRMP5 levels in control mice exacerbated memory decline after subthreshold stress treatment. Mechanistically, hippocampal CRMP5 suppression by regulating glucocorticoid receptor phosphorylation alleviates chronic stress-induced synaptic atrophy, disruption of AMPA receptor trafficking, and cytokine storms. Our findings show that hippocampal CRMP5 accumulation through GR activation disrupts synaptic plasticity, impedes AMPAR trafficking, and triggers cytokine release, thus playing a critical role in chronic stress-induced cognitive deficits.
Collapse
|
15
|
Liu MS, Tian ZY, Zhang Z, Yang F, Lou Y, Wang YJ, Zeng YJ, Zhang ZW, Zhu DL, Li P. Impaired Cognitive Function in Patients With Autonomous Cortisol Secretion in Adrenal Incidentalomas. J Clin Endocrinol Metab 2023; 108:633-641. [PMID: 36263685 DOI: 10.1210/clinem/dgac603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/02/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Glucocorticoids have potent effects on the central nervous system. However, while patients with Cushing syndrome frequently report impairments in cognitive function, studies investigating cognitive function in patients with autonomous cortisol secretion (ACS) in adrenal incidentalomas (AIs) are scarce. OBJECTIVE The aim of the present study was to evaluate neurocognitive function in patients with ACS. METHODS We prospectively recruited 63 patients with AI, 36 patients with nonfunctional adrenal adenoma (NFA) (46.5 ± 10.5 years), and 27 patients with ACS (48.6 ± 9.1 years); these patients underwent a battery of validated neuropsychological tests. ACS was diagnosed when serum cortisol levels after a 1-mg dexamethasone suppression test (cortisol1 mg DST) ≥ 50 nmol/L. RESULTS Patients with ACS had higher frequency of subjective memory complaints (40.7% vs 13.9%, P < 0.05) and higher proportion of mild cognitive impairment (22.2% vs 2.8%, P < 0.05) than patients with NFA. Furthermore, patients with ACS had worse performance on working memory and the visuospatial/constructional domain than patients with NFA (all P < 0.05). Serum cortisol1 mg DST was negatively correlated with working memory and visuospatial/constructional domains (r = -0.307 and -0.306, respectively, all P < 0.05). Performance on working memory and visuospatial/constructional domains gradually deteriorated with increases in serum cortisol1 mg DST (all P values for trend < 0.05). Multivariate linear regression analysis showed that serum cortisol1 mg DST was a significant risk factor for impairment of working memory and visuospatial/constructional domains (B = -0.006 and -0.043, respectively, all P < 0.05). CONCLUSION This study is the first to report that ACS is accompanied by impaired cognitive function. Consequently, cognitive function assessment should be incorporated into the clinical evaluation of patients with ACS. CLINICAL TRIAL REGISTRATION NUMBER NCT05357456.
Collapse
Affiliation(s)
- Meng-Si Liu
- Department of Endocrinology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing 210008, China
| | - Zhao-Yang Tian
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
- Endocrine and Metabolic Disease Medical Center, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing 210008, China
| | - Zhou Zhang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
- Endocrine and Metabolic Disease Medical Center, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing 210008, China
| | - Fan Yang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
- Endocrine and Metabolic Disease Medical Center, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing 210008, China
| | - Yuan Lou
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
- Endocrine and Metabolic Disease Medical Center, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing 210008, China
| | - Yi-Jie Wang
- Department of Endocrinology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Yang-Jie Zeng
- Department of Endocrinology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing 210008, China
| | - Zi-Wei Zhang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
- Endocrine and Metabolic Disease Medical Center, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing 210008, China
| | - Da-Long Zhu
- Department of Endocrinology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing 210008, China
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
- Endocrine and Metabolic Disease Medical Center, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing 210008, China
| | - Ping Li
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
- Endocrine and Metabolic Disease Medical Center, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing 210008, China
- Department of Endocrinology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| |
Collapse
|
16
|
Ruan J, Hu X, Liu Y, Han Z, Ruan Q. Vulnerability to chronic stress and the phenotypic heterogeneity of presbycusis with subjective tinnitus. Front Neurosci 2022; 16:1046095. [PMID: 36620444 PMCID: PMC9812577 DOI: 10.3389/fnins.2022.1046095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Age-related functional reserve decline and vulnerability of multiple physiological systems and organs, as well as at the cellular and molecular levels, result in different frailty phenotypes, such as physical, cognitive, and psychosocial frailty, and multiple comorbidities, including age-related hearing loss (ARHL) and/or tinnitus due to the decline in auditory reserve. However, the contributions of chronic non-audiogenic cumulative exposure, and chronic audiogenic stress to phenotypic heterogeneity of presbycusis and/or tinnitus remain elusive. Because of the cumulative environmental stressors throughout life, allostasis systems, the hypothalamus-pituitary-adrenal (HPA) and the sympathetic adrenal-medullary (SAM) axes become dysregulated and less able to maintain homeostasis, which leads to allostatic load and maladaptation. Brain-body communication via the neuroendocrine system promotes systemic chronic inflammation, overmobilization of energetic substances (glucose and lipids), and neuroplastic changes via the non-genomic and genomic actions of glucocorticoids, catecholamines, and their receptors. These systemic maladaptive alterations might lead to different frailty phenotypes and physical, cognitive, and psychological comorbidities, which, in turn, cause and exacerbate ARHL and/or tinnitus with phenotypic heterogeneity. Chronic audiogenic stressors, including aging accompanying ontological diseases, cumulative noise exposure, and ototoxic drugs as well as tinnitus, activate the HPA axis and SAM directly and indirectly by the amygdala, promoting allostatic load and maladaptive neuroplasticity in the auditory system and other vulnerable brain regions, such as the hippocampus, amygdala, and medial prefrontal cortex (mPFC). In the auditory system, peripheral deafferentation, central disinhibition, and tonotopic map reorganization may trigger tinnitus. Cross-modal maladaptive neuroplasticity between the auditory and other sensory systems is involved in tinnitus modulation. Persistent dendritic growth and formation, reduction in GABAergic inhibitory synaptic inputs induced by chronic audiogenic stresses in the amygdala, and increased dendritic atrophy in the hippocampus and mPFC, might involve the enhancement of attentional processing and long-term memory storage of chronic subjective tinnitus, accompanied by cognitive impairments and emotional comorbidities. Therefore, presbycusis and tinnitus are multisystem disorders with phenotypic heterogeneity. Stressors play a critical role in the phenotypic heterogeneity of presbycusis. Differential diagnosis based on biomarkers of metabonomics study, and interventions tailored to different ARHL phenotypes and/or tinnitus will contribute to healthy aging and improvement in the quality of life.
Collapse
Affiliation(s)
- Jian Ruan
- Department of Otolaryngology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiuhua Hu
- Laboratory of Aging, Anti-aging & Cognitive Performance, Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital, Fudan University, Shanghai, China,Shanghai Key Laboratory of Clinical Geriatrics, Research Center of Aging and Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuehong Liu
- Department of Otolaryngology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhao Han
- Department of Otolaryngology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qingwei Ruan
- Laboratory of Aging, Anti-aging & Cognitive Performance, Shanghai Institute of Geriatrics and Gerontology, Huadong Hospital, Fudan University, Shanghai, China,Shanghai Key Laboratory of Clinical Geriatrics, Research Center of Aging and Medicine, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China,*Correspondence: Qingwei Ruan,
| |
Collapse
|
17
|
Luthra NS, Clow A, Corcos DM. The Interrelated Multifactorial Actions of Cortisol and Klotho: Potential Implications in the Pathogenesis of Parkinson's Disease. Brain Sci 2022; 12:1695. [PMID: 36552155 PMCID: PMC9775285 DOI: 10.3390/brainsci12121695] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The pathogenesis of Parkinson's disease (PD) is complex, multilayered, and not fully understood, resulting in a lack of effective disease-modifying treatments for this prevalent neurodegenerative condition. Symptoms of PD are heterogenous, including motor impairment as well as non-motor symptoms such as depression, cognitive impairment, and circadian disruption. Aging and stress are important risk factors for PD, leading us to explore pathways that may either accelerate or protect against cellular aging and the detrimental effects of stress. Cortisol is a much-studied hormone that can disrupt mitochondrial function and increase oxidative stress and neuroinflammation, which are recognized as key underlying disease mechanisms in PD. The more recently discovered klotho protein, considered a general aging-suppressor, has a similarly wide range of actions but in the opposite direction to cortisol: promoting mitochondrial function while reducing oxidative stress and inflammation. Both hormones also converge on pathways of vitamin D metabolism and insulin resistance, also implicated to play a role in PD. Interestingly, aging, stress and PD associate with an increase in cortisol and decrease in klotho, while physical exercise and certain genetic variations lead to a decrease in cortisol response and increased klotho. Here, we review the interrelated opposite actions of cortisol and klotho in the pathogenesis of PD. Together they impact powerful and divergent mechanisms that may go on to influence PD-related symptoms. Better understanding of these hormones in PD would facilitate the design of effective interventions that can simultaneously impact the multiple systems involved in the pathogenesis of PD.
Collapse
Affiliation(s)
- Nijee S. Luthra
- Department of Neurology, University of California San Francisco, San Francisco, CA 94127, USA
| | - Angela Clow
- Department of Psychology, School of Social Sciences, University of Westminster, London W1B 2HW, UK
| | - Daniel M. Corcos
- Department of Physical Therapy & Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60208, USA
| |
Collapse
|
18
|
Maganga-Bakita I, Aiken AA, Puracchio MJ, Kentner AC, Hunter RG. Regulatory Effects of Maternal Immune Activation and Environmental Enrichment on Glucocorticoid Receptor and FKBP5 Expression in Stress-sensitive Regions of the Offspring Brain. Neuroscience 2022; 505:51-58. [PMID: 36116554 PMCID: PMC9888218 DOI: 10.1016/j.neuroscience.2022.09.010] [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: 11/16/2021] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 02/02/2023]
Abstract
A mother's exposure to immune challenge during pregnancy is well known to be a detrimental factor to the development of the offspring's brain and an impetus for neuropsychiatric disorders. Previous studies have shown that these adverse events can dysregulate the stress response machinery. Two crucial components of the stress axis considered to be affected have been targets in these studies: the glucocorticoid receptor (GR), and FKBP5 which regulates GR activity. The implementation of interventions such as Environmental Enrichment (EE) have shown positive results in protecting the brain against the consequences associated with gestational insults. In light of this, we investigated the transcriptional regulation of GR and FKBP5 from six stress-sensitive brain regions of the offspring using a rat model of maternal immune activation (MIA). Furthermore, we analyzed the effect of an enriched environment on their expression. We found an increase in FKBP5 in MIA rats in five brain regions. RT-qPCR analysis of MIA's effect on GR yielded insignificant results. However, we found that EE increased GR expression in the medial preoptic area which could be indicative of a positive regulation by EE. This study provides evidence of the impact of both gestational insult and EE on the regulation of stress responsive genes in the developing brain.
Collapse
Affiliation(s)
| | - Ariel A Aiken
- University of Massachusetts Boston, Department of Psychology, Boston, MA, USA
| | - Madeline J Puracchio
- Massachusetts College of Pharmacy and Health Sciences, Department of Psychology, Boston, MA, USA
| | - Amanda C Kentner
- Massachusetts College of Pharmacy and Health Sciences, Department of Psychology, Boston, MA, USA
| | - Richard G Hunter
- University of Massachusetts Boston, Department of Psychology, Boston, MA, USA.
| |
Collapse
|
19
|
Chaudry O, Ndukwe K, Xie L, Figueiredo-Pereira M, Serrano P, Rockwell P. Females exhibit higher GluA2 levels and outperform males in active place avoidance despite increased amyloid plaques in TgF344-Alzheimer's rats. Sci Rep 2022; 12:19129. [PMID: 36352024 PMCID: PMC9646806 DOI: 10.1038/s41598-022-23801-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that is most prevalent in females. While estrogen provides neuroprotection in females, sex mediated differences in the development of AD pathology are not fully elucidated. Therefore, comparing events between sexes in early-stage AD pathology may reveal more effective therapeutic targets of intervention. To address sex differences, we analyzed early-stage 9-month male and female TgF344-AD (Tg-AD) rats, an AD model carrying the APPswe and Presenilin 1 (PS1ΔE9) mutations that develops progressive age-dependent AD pathology similar to humans. Tg-AD females significantly outperformed Tg-AD males in the active place avoidance (aPAT) test that assesses hippocampal-dependent spatial learning and memory. However, comparisons between Tg-AD male or female rats and their WT counterparts showed significant deficits for female but not male rats. Nevertheless, Tg-AD females experienced significantly less hippocampal neuronal loss with higher GluA2 subunit levels than Tg-AD males. Unexpectedly, Tg-AD females displayed higher levels of hippocampal amyloid plaques than Tg-AD males. Thus, we propose that GluA2 may provide a neuroprotective function for Tg-AD females in our rat model by mitigating cognitive impairment independently of amyloid plaques. Elucidating this protective mechanism in AD could lead to new targets for early intervention.
Collapse
Affiliation(s)
- Osama Chaudry
- Department of Biological Sciences, Hunter College CUNY, New York, NY, USA
| | - Kelechi Ndukwe
- Department of Biological Sciences, Hunter College CUNY, New York, NY, USA
- PhD Program in Neuroscience, The Graduate Center CUNY, New York, NY, USA
| | - Lei Xie
- Department of Computer Sciences, Hunter College CUNY, New York, NY, USA
| | | | - Peter Serrano
- Department of Psychology, Hunter College CUNY, New York, NY, USA
| | - Patricia Rockwell
- Department of Biological Sciences, Hunter College CUNY, New York, NY, USA.
| |
Collapse
|
20
|
Puigoriol-Illamola D, Companys-Alemany J, McGuire K, Homer NZM, Leiva R, Vázquez S, Mole DJ, Griñán-Ferré C, Pallàs M. Inhibition of 11β-HSD1 Ameliorates Cognition and Molecular Detrimental Changes after Chronic Mild Stress in SAMP8 Mice. Pharmaceuticals (Basel) 2021; 14:ph14101040. [PMID: 34681264 PMCID: PMC8540242 DOI: 10.3390/ph14101040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
Impaired glucocorticoid (GC) signaling is a significant factor in aging, stress, and neurodegenerative diseases such as Alzheimer's disease. Therefore, the study of GC-mediated stress responses to chronic moderately stressful situations, which occur in daily life, is of huge interest for the design of pharmacological strategies toward the prevention of neurodegeneration. To address this issue, SAMP8 mice were exposed to the chronic mild stress (CMS) paradigm for 4 weeks and treated with RL-118, an 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitor. The inhibition of this enzyme is linked with a reduction in GC levels and cognitive improvement, while CMS exposure has been associated with reduced cognitive performance. The aim of this project was to assess whether RL-118 treatment could reverse the deleterious effects of CMS on cognition and behavioral abilities and to evaluate the molecular mechanisms that compromise healthy aging in SAMP8 mice. First, we confirmed the target engagement between RL-118 and 11β-HSD1. Additionally, we showed that DNA methylation, hydroxymethylation, and histone phosphorylation were decreased by CMS induction, and increased by RL-118 treatment. In addition, CMS exposure caused the accumulation of reactive oxygen species (ROS)-induced damage and increased pro-oxidant enzymes-as well as pro-inflammatory mediators-through the NF-κB pathway and astrogliosis markers, such as GFAP. Of note, these modifications were reversed by 11β-HSD1 inhibition. Remarkably, although CMS altered mTORC1 signaling, autophagy was increased in the SAMP8 RL-118-treated mice. We also showed an increase in amyloidogenic processes and a decrease in synaptic plasticity and neuronal remodeling markers in mice under CMS, which were consequently modified by RL-118 treatment. In conclusion, 11β-HSD1 inhibition through RL-118 ameliorated the detrimental effects induced by CMS, including epigenetic and cognitive disturbances, indicating that GC-excess attenuation shows potential as a therapeutic strategy for age-related cognitive decline and AD.
Collapse
Affiliation(s)
- Dolors Puigoriol-Illamola
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (D.P.-I.); (J.C.-A.); (C.G.-F.)
- Institute of Neuroscience, University of Barcelona (NeuroUB), Passeig Vall d’Hebron 171, 08028 Barcelona, Spain
| | - Júlia Companys-Alemany
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (D.P.-I.); (J.C.-A.); (C.G.-F.)
- Institute of Neuroscience, University of Barcelona (NeuroUB), Passeig Vall d’Hebron 171, 08028 Barcelona, Spain
| | - Kris McGuire
- MRC Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (K.M.); (D.J.M.)
| | - Natalie Z. M. Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, Edinburgh EH16 4TJ, UK;
| | - Rosana Leiva
- Medicinal Chemistry Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (R.L.); (S.V.)
| | - Santiago Vázquez
- Medicinal Chemistry Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (R.L.); (S.V.)
| | - Damian J. Mole
- MRC Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; (K.M.); (D.J.M.)
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (D.P.-I.); (J.C.-A.); (C.G.-F.)
- Institute of Neuroscience, University of Barcelona (NeuroUB), Passeig Vall d’Hebron 171, 08028 Barcelona, Spain
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (D.P.-I.); (J.C.-A.); (C.G.-F.)
- Institute of Neuroscience, University of Barcelona (NeuroUB), Passeig Vall d’Hebron 171, 08028 Barcelona, Spain
- Correspondence: ; Tel.: +34-4024531
| |
Collapse
|
21
|
Peroxiredoxin 6 Knockout Mice Demonstrate Anxiety Behavior and Attenuated Contextual Fear Memory after Receiving Acute Immobilization Stress. Antioxidants (Basel) 2021; 10:antiox10091416. [PMID: 34573048 PMCID: PMC8466988 DOI: 10.3390/antiox10091416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Stress can elicit glucocorticoid release to promote coping mechanisms and influence learning and memory performance. Individual memory performance varies in response to stress, and the underlying mechanism is not clear yet. Peroxiredoxin 6 (PRDX6) is a multifunctional enzyme participating in both physiological and pathological conditions. Several studies have demonstrated the correlation between PRDX6 expression level and stress-related disorders. Our recent finding indicates that lack of the Prdx6 gene leads to enhanced fear memory. However, it is unknown whether PRDX6 is involved in changes in anxiety response and memory performance upon stress. The present study reveals that hippocampal PRDX6 level is downregulated 30 min after acute immobilization stress (AIS) and trace fear conditioning (TFC). In human retinal pigment epithelium (ARPE-19) cells, the PRDX6 expression level decreases after being treated with stress hormone corticosterone. Lack of PRDX6 caused elevated basal H2O2 levels in the hippocampus, basolateral amygdala, and medial prefrontal cortex, brain regions involved in anxiety response and fear memory formation. Additionally, this H2O2 level was still high in the medial prefrontal cortex of the knockout mice under AIS. Anxiety behavior of Prdx6-/- mice was enhanced after immobilization for 30 min. After exposure to AIS before a contextual test, Prdx6-/- mice displayed a contextual fear memory deficit. Our results showed that the memory performance of Prdx6-/- mice was impaired when responding to AIS, accompanied by dysregulated H2O2 levels. The present study helps better understand the function of PRDX6 in memory performance after acute stress.
Collapse
|
22
|
Serum Corticosterone and Insulin Resistance as Early Biomarkers in the hAPP23 Overexpressing Mouse Model of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22136656. [PMID: 34206322 PMCID: PMC8269119 DOI: 10.3390/ijms22136656] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/21/2022] Open
Abstract
Increasing epidemiological evidence highlights the association between systemic insulin resistance and Alzheimer’s disease (AD). As insulin resistance can be caused by high-stress hormone levels and since hypercortisolism appears to be an important risk factor of AD, we aimed to investigate the systemic insulin functionality and circulating stress hormone levels in a mutant humanized amyloid precursor protein (APP) overexpressing (hAPP23+/−) AD mouse model. Memory and spatial learning of male hAPP23+/− and C57BL/6 (wild type, WT) mice were assessed by a Morris Water Maze (MWM) test at the age of 4 and 12 months. The systemic metabolism was examined by intraperitoneal glucose and insulin tolerance tests (GTT, ITT). Insulin and corticosterone levels were determined in serum. In the hippocampus, parietal and occipital cortex of hAPP23+/− brains, amyloid-beta (Aβ) deposits were present at 12 months of age. MWM demonstrated a cognitive decline in hAPP23+/− mice at 12 but not at 4 months, evidenced by increasing total path lengths and deteriorating probe trials compared to WT mice. hAPP23+/− animals presented increased serum corticosterone levels compared to WT mice at both 4 and 12 months. hAPP23+/− mice exhibited peripheral insulin resistance compared to WT mice at 4 months, which stabilized at 12 months of age. Serum insulin levels were similar between genotypes at 4 months of age but were significantly higher in hAPP23+/− mice at 12 months of age. Peripheral glucose homeostasis remained unchanged. These results indicate that peripheral insulin resistance combined with elevated circulating stress hormone levels could be potential biomarkers of the pre-symptomatic phase of AD.
Collapse
|
23
|
Vanderhaeghen T, Beyaert R, Libert C. Bidirectional Crosstalk Between Hypoxia Inducible Factors and Glucocorticoid Signalling in Health and Disease. Front Immunol 2021; 12:684085. [PMID: 34149725 PMCID: PMC8211996 DOI: 10.3389/fimmu.2021.684085] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Glucocorticoid-induced (GC) and hypoxia-induced transcriptional responses play an important role in tissue homeostasis and in the regulation of cellular responses to stress and inflammation. Evidence exists that there is an important crosstalk between both GC and hypoxia effects. Hypoxia is a pathophysiological condition to which cells respond quickly in order to prevent metabolic shutdown and death. The hypoxia inducible factors (HIFs) are the master regulators of oxygen homeostasis and are responsible for the ability of cells to cope with low oxygen levels. Maladaptive responses of HIFs contribute to a variety of pathological conditions including acute mountain sickness (AMS), inflammation and neonatal hypoxia-induced brain injury. Synthetic GCs which are analogous to the naturally occurring steroid hormones (cortisol in humans, corticosterone in rodents), have been used for decades as anti-inflammatory drugs for treating pathological conditions which are linked to hypoxia (i.e. asthma, ischemic injury). In this review, we investigate the crosstalk between the glucocorticoid receptor (GR), and HIFs. We discuss possible mechanisms by which GR and HIF influence one another, in vitro and in vivo, and the therapeutic effects of GCs on HIF-mediated diseases.
Collapse
Affiliation(s)
- Tineke Vanderhaeghen
- Centre for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Rudi Beyaert
- Centre for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Claude Libert
- Centre for Inflammation Research, Flanders Institute for Biotechnology (VIB), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
24
|
Lin YF, Wang LY, Chen CS, Li CC, Hsiao YH. Cellular senescence as a driver of cognitive decline triggered by chronic unpredictable stress. Neurobiol Stress 2021; 15:100341. [PMID: 34095365 PMCID: PMC8163993 DOI: 10.1016/j.ynstr.2021.100341] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
When an individual is under stress, the undesired effect on the brain often exceeds expectations. Additionally, when stress persists for a long time, it can trigger serious health problems, particularly depression. Recent studies have revealed that depressed patients have a higher rate of brain aging than healthy subjects and that depression increases dementia risk later in life. However, it remains unknown which factors are involved in brain aging triggered by chronic stress. The most critical change during brain aging is the decline in cognitive function. In addition, cellular senescence is a stable state of cell cycle arrest that occurs because of damage and/or stress and is considered a sign of aging. We used the chronic unpredictable stress (CUS) model to mimic stressful life situations and found that, compared with nonstressed control mice, CUS-treated C57BL/6 mice exhibited depression-like behaviors and cognitive decline. Additionally, the protein expression of the senescence marker p16INK4a was increased in the hippocampus, and senescence-associated β-galactosidase (SA-β-gal)-positive cells were found in the hippocampal dentate gyrus (DG) in CUS-treated mice. Furthermore, the levels of SA-β-gal or p16INK4a were strongly correlated with the severity of memory impairment in CUS-treated mice, whereas clearing senescent cells using the pharmacological senolytic cocktail dasatinib plus quercetin (D + Q) alleviated CUS-induced cognitive deficits, suggesting that targeting senescent cells may be a promising candidate approach to study chronic stress-induced cognitive decline. Our findings open new avenues for stress-related research and provide new insight into the association of chronic stress-induced cellular senescence with cognitive deficits.
Collapse
Affiliation(s)
- Yu-Fen Lin
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Li-Yun Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Sheng Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Chun Li
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Hsin Hsiao
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
25
|
Down-regulation of MST1 in hippocampus protects against stress-induced depression-like behaviours and synaptic plasticity impairments. Brain Behav Immun 2021; 94:196-209. [PMID: 33607238 DOI: 10.1016/j.bbi.2021.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 01/26/2021] [Accepted: 02/05/2021] [Indexed: 12/15/2022] Open
Abstract
Depression is a common mental disorder, and its main environmental risk factor is chronic stress. The activation of mammalian STE20-like kinase 1 (MST1), a key factor involved in the underlying pathophysiology of stress, can trigger synaptic plasticity impairment, neuronal dysfunction and neuroinflammation. However, it is unclear whether down-regulation of MST1 in the hippocampus protects against stress-induced behavioural dysfunctions. In this study, three mouse models were used to assess the role of MST1 in stress. Various behavioural tests, in vivo electrophysiological recordings, Western blotting, Golgi staining and immunofluorescence assay were used. The data showed that the level of phospho-MST1 (T183) was significantly increased in the hippocampus of mice subjected to chronic unpredictable mild stress (CUMS) and that mice with MST1 overexpression showed depression-like behaviours. Importantly, the impairment of cognitive functions and the hippocampal synaptic plasticity induced by CUMS were significantly improved by MST1 knockdown, suggesting that MST1 down-regulation effectively protected against stress-induced behavioural dysfunctions. Moreover, MST1 knockdown suppressed CUMS-induced microglial activation, reduced the abnormal expression of inflammatory cytokines and impeded the activation of p38, implying that the antidepressant-like effects of MST1 knockdown were associated with inhibiting the p38 pathway. These findings suggest that hippocampal MST1 is an essential regulator of stress, which can be an ideal target for the development of antidepressants in the future.
Collapse
|
26
|
Pakdeepak K, Chokchaisiri R, Govitrapong P, Tocharus C, Suksamrarn A, Tocharus J. 5,6,7,4'-Tetramethoxyflavanone alleviates neurodegeneration in a dexamethasone-induced neurodegenerative mouse model through promotion of neurogenesis via the Raf/ERK1/2 pathway. Phytother Res 2021; 35:2536-2544. [PMID: 33319436 DOI: 10.1002/ptr.6983] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 12/27/2022]
Abstract
Adult neurogenesis plays an important role in improving cognitive functions. Neurogenesis generates new neurons, a process mediated by neural stem cell proliferation, migration, and differentiation. Long-term exposure to high levels of glucocorticoid results in the suppression of neurogenesis pathways and leads to the onset of cognitive impairment. The induction of neurogenesis by a potent bioactive compound is considered the most promising treatment for neurodegenerative disorders. 5,6,7,4'-Tetramethoxyflavanone (TMF) is a flavonoid compound isolated from Chromolaena odorata (L.) R. M. King & H. Rob. Previous study showed that TMF improved cognitive impairment by attenuating Aβ production and pTau expression, thereby increased cell survival and promoted synaptic plasticity. The aim of this study was to investigate the effect of TMF on dexamethasone (DEX)-suppressed neurogenesis in mice. Mice received DEX for 28 days before being treated with TMF for additional 30 days. Mice were randomly divided into four groups: control, TMF, DEX, and DEX + TMF. TMF promoted neurogenesis by increasing BrdU-positive cells, Prox1, doublecortin, and Nestin expression. TMF also upregulated the expression of Raf and extracellular-signal-regulated kinase (ERK)1/2, which are pivotal for neurogenesis signaling. In conclusion, TMF promoted neurogenesis-related protein expression in the proliferation, differentiation, and maturation phases via Raf/ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Kanet Pakdeepak
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Graduate School, Chiang Mai University, Chiang Mai, Thailand
| | | | | | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Functional Food Research Center for Well-being, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
27
|
Bonaterra GA, Mierau O, Hofmann J, Schwarzbach H, Aziz-Kalbhenn H, Kolb C, Kinscherf R. In Vitro Effects of St. John's Wort Extract Against Inflammatory and Oxidative Stress and in the Phagocytic and Migratory Activity of Mouse SIM-A9 Microglia. Front Pharmacol 2021; 11:603575. [PMID: 33628177 PMCID: PMC7898673 DOI: 10.3389/fphar.2020.603575] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/10/2020] [Indexed: 12/28/2022] Open
Abstract
Introduction: Herbal medicinal plants as Hypericum perforatum L., known as St. John’s wort (SJW) have been in use for a long time. SJW that is specifically used for the treatment of depressive disorders. Inflammatory cytokines derived from microglia play an important role in the regulation of the synthesis and reuptake of glutamate and influence synaptic function, morphology and neuronal plasticity. The present study was performed to investigate, whether STW3-VI, a special SJW extract has protective effects on mouse SIM-A9 microglia against cytotoxic and proinflammatory effects of ROS, glutamate, NMDA or cortisol. Additionally, we investigated the effects of SJW on migratory and phagocytic properties of microglia. Results: Pre-treatment (48 h) of microglia with STW3-VI (5 or 10 μg/ml)—in contrast to desipramine—inhibited the H2O2-induced TNF-α release by 20–40%. Pre-treatment (48 h) of microglia with STW3-VI (5 or 10 μg/ml) delayed the 3 or 4 mM H2O2-induced intracellular ROS level by 26.9 and 44.4%, respectively. Furthermore, pre-treatment (48 h) of microglia with STW3-VI (5 μg/ml) - in contrast to desipramine - lowered the glutamate-induced cytotoxicity by 13.2%. Besides, pre-treatment (48 h) of microglia with STW3-VI (5 or 10 μg/ml) or desipramine (5 µM) inhibited the NMDA-induced decrease of the viability by 16.5–28.8% or 12%, respectively. Finally, pre-treatment (48 h) of microglia with STW3-VI (5 or 10 μg/ml)—in contrast to desipramine - reduced the cortisol-induced cytotoxicity by 15.5 and 12.9%. Treatment of microglia with STW3-VI (10 or 100 μg/ml) increased the migratory and the phagocytic capacities by 100 and 40%. Conclusion: Our data provide evidence that STW3-VI—in contrast to desipramine - protects microglia from oxidative stress, NMDA- or glutamate-induced cytotoxicity, and has anti-inflammatory properties that are accompanied by improvement of their migratory and phagocytic capacity. These protective (particularly the anti-inflammatory) properties may be beneficial in the treatment of depressive disorders.
Collapse
Affiliation(s)
- Gabriel A Bonaterra
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Olga Mierau
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Johanna Hofmann
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Hans Schwarzbach
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Heba Aziz-Kalbhenn
- Bayer Consumer Health Division, Phytomedicines Supply and Development Center, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Christiane Kolb
- Bayer Consumer Health Division, Phytomedicines Supply and Development Center, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Ralf Kinscherf
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| |
Collapse
|
28
|
Barlampa D, Bompoula MS, Bargiota A, Kalantaridou S, Mastorakos G, Valsamakis G. Hypothalamic Inflammation as a Potential Pathophysiologic Basis for the Heterogeneity of Clinical, Hormonal, and Metabolic Presentation in PCOS. Nutrients 2021; 13:520. [PMID: 33562540 PMCID: PMC7915850 DOI: 10.3390/nu13020520] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among women of reproductive age. It is a heterogeneous condition characterized by reproductive, endocrine, metabolic, and psychiatric abnormalities. More than one pathogenic mechanism is involved in its development. On the other hand, the hypothalamus plays a crucial role in many important functions of the body, including weight balance, food intake, and reproduction. A high-fat diet with a large amount of long-chain saturated fatty acids can induce inflammation in the hypothalamus. Hypothalamic neurons can sense extracellular glucose concentrations and participate, with a feedback mechanism, in the regulation of whole-body glucose homeostasis. When consumed nutrients are rich in fat and sugar, and these regulatory mechanisms can trigger inflammatory pathways resulting in hypothalamic inflammation. The latter has been correlated with metabolic diseases, obesity, and depression. In this review, we explore whether the pattern and the expansion of hypothalamic inflammation, as a result of a high-fat and -sugar diet, may contribute to the heterogeneity of the clinical, hormonal, and metabolic presentation in PCOS via pathophysiologic mechanisms affecting specific areas of the hypothalamus. These mechanisms could be potential targets for the development of effective therapies for the treatment of PCOS.
Collapse
Affiliation(s)
- Danai Barlampa
- Unit of Endocrinology, Aretaieion University Hospital, Medical School of Athens, Ethnikon and Kapodistriakon University of Athens, Athens, Vasilisis Sofia Avenue 76, 115 28 Athens, Greece; (D.B.); (G.V.)
| | - Maria Sotiria Bompoula
- Reproductive Endocrinology Unit, 3nd University Department of Obs & Gynae, Attikon University Hospital, Medical School of Athens, Ethnikon and Kapodistriakon University of Athens, 12462 Athens, Greece; (M.S.B.); (S.K.)
| | - Alexandra Bargiota
- Department of Endocrinology and Metabolic Disorders, University Hospital of Larissa, Medical School of Larissa, University of Thessaly, 41334 Larissa, Greece;
| | - Sophia Kalantaridou
- Reproductive Endocrinology Unit, 3nd University Department of Obs & Gynae, Attikon University Hospital, Medical School of Athens, Ethnikon and Kapodistriakon University of Athens, 12462 Athens, Greece; (M.S.B.); (S.K.)
| | - George Mastorakos
- Unit of Endocrinology, Aretaieion University Hospital, Medical School of Athens, Ethnikon and Kapodistriakon University of Athens, Athens, Vasilisis Sofia Avenue 76, 115 28 Athens, Greece; (D.B.); (G.V.)
| | - Georgios Valsamakis
- Unit of Endocrinology, Aretaieion University Hospital, Medical School of Athens, Ethnikon and Kapodistriakon University of Athens, Athens, Vasilisis Sofia Avenue 76, 115 28 Athens, Greece; (D.B.); (G.V.)
- Reproductive Endocrinology Unit, 3nd University Department of Obs & Gynae, Attikon University Hospital, Medical School of Athens, Ethnikon and Kapodistriakon University of Athens, 12462 Athens, Greece; (M.S.B.); (S.K.)
- Department of Endocrinology and Metabolic Disorders, University Hospital of Larissa, Medical School of Larissa, University of Thessaly, 41334 Larissa, Greece;
| |
Collapse
|
29
|
Dionysopoulou S, Charmandari E, Bargiota A, Vlahos NF, Mastorakos G, Valsamakis G. The Role of Hypothalamic Inflammation in Diet-Induced Obesity and Its Association with Cognitive and Mood Disorders. Nutrients 2021; 13:498. [PMID: 33546219 PMCID: PMC7913301 DOI: 10.3390/nu13020498] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/24/2021] [Accepted: 01/30/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is often associated with cognitive and mood disorders. Recent evidence suggests that obesity may cause hypothalamic inflammation. Our aim was to investigate the hypothesis that there is a causal link between obesity-induced hypothalamic inflammation and cognitive and mood disorders. Inflammation may influence hypothalamic inter-connections with regions important for cognition and mood, while it may cause dysregulation of the Hypothalamic-Pituitary-Adrenal (HPA) axis and influence monoaminergic systems. Exercise, healthy diet, and glucagon-like peptide receptor agonists, which can reduce hypothalamic inflammation in obese models, could improve the deleterious effects on cognition and mood.
Collapse
Affiliation(s)
- Sofia Dionysopoulou
- Division of Endocrinology, Metabolism and Diabetes, Hippocratio General Hospital, 11527 Athens, Greece;
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece;
- Division of Endocrinology and Metabolism, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Alexandra Bargiota
- Department of Endocrinology and Metabolic Diseases, University Hospital of Larisa, Medical School of Larisa, University of Thessaly, 41334 Larisa, Greece;
| | - Nikolaos F Vlahos
- 2nd Department of Obstetrics and Gynecology, Areteion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - George Mastorakos
- Endocrine Unit, Areteion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Georgios Valsamakis
- Department of Endocrinology and Metabolic Diseases, University Hospital of Larisa, Medical School of Larisa, University of Thessaly, 41334 Larisa, Greece;
- Endocrine Unit, Areteion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| |
Collapse
|
30
|
Pervin M, Unno K, Konishi T, Nakamura Y. L-Arginine Exerts Excellent Anti-Stress Effects on Stress-Induced Shortened Lifespan, Cognitive Decline and Depression. Int J Mol Sci 2021; 22:E508. [PMID: 33419170 PMCID: PMC7825557 DOI: 10.3390/ijms22020508] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 12/23/2022] Open
Abstract
The anti-stress potential of dietary L-arginine (Arg) was assessed in psychosocially stress-loaded senescence-accelerated (SAMP10) mice. Although this strain of mouse is sensitive to stress, daily administration of Arg at 3 mg/kg significantly suppressed aging-related cognitive decline and behavioral depression at nine months of age and counteracted stress-induced shortened lifespan. To investigate the mechanism of the anti-stress effect of Arg in the brain, early changes in oxidative damage and gene expression levels were measured using SAMP10 mice that were stress-loaded for three days. Increased lipid peroxidation in the brains of stressed mice was significantly lowered by Arg intake. Several genes associated with oxidative stress response and neuronal excitotoxic cell death, including Nr4a1, Arc, and Cyr61, remarkably increased in response to psychosocial stress; however, their expression was significantly suppressed in mice that ingested Arg even under stress conditions. In contrast, the genes that maintain mitochondrial functions and neuronal survival, including Hba-a2 and Hbb-b2, were significantly increased in mice that ingested Arg. These results indicate that Arg reduces oxidative damage and enhances mitochondrial functions in the brain. We suggest that the daily intake of Arg plays important roles in reducing stress-induced brain damage and slowing aging.
Collapse
Affiliation(s)
- Monira Pervin
- Tea Science Center, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan;
| | - Keiko Unno
- Tea Science Center, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan;
| | - Tomokazu Konishi
- Faculty of Bioresource Sciences, Akita Prefectural University, Shimoshinjo Nakano, Akita 010-0195, Japan;
| | - Yoriyuki Nakamura
- Tea Science Center, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan;
| |
Collapse
|
31
|
Lyons CE, Bartolomucci A. Stress and Alzheimer's disease: A senescence link? Neurosci Biobehav Rev 2020; 115:285-298. [PMID: 32461080 PMCID: PMC7483955 DOI: 10.1016/j.neubiorev.2020.05.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 04/11/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022]
Abstract
Chronic stress has been shown to promote numerous aging-related diseases, and to accelerate the aging process itself. Of particular interest is the impact of stress on Alzheimer's disease (AD), the most prevalent form of dementia. The vast majority of AD cases have no known genetic cause, making it vital to identify the environmental factors involved in the onset and progression of the disease. Age is the greatest risk factor for AD, and measures of biological aging such as shorter telomere length, significantly increase likelihood for developing AD. Stress is also considered a crucial contributor to AD, as indicated by a formidable body of research, although the mechanisms underlying this association remain unclear. Here we review human and animal literature on the impact of stress on AD and discuss the mechanisms implicated in the interaction. In particular we will focus on the burgeoning body of research demonstrating that senescent cells, which accumulate with age and actively drive a number of aging-related diseases, may be a key mechanism through which stress drives AD.
Collapse
Affiliation(s)
- Carey E Lyons
- Department of Integrative Biology and Physiology, University of Minnesota, United States; Graduate Program in Neuroscience, University of Minnesota, United States.
| | | |
Collapse
|
32
|
Meszaros K, Patocs A. Glucocorticoids Influencing Wnt/β-Catenin Pathway; Multiple Sites, Heterogeneous Effects. Molecules 2020; 25:molecules25071489. [PMID: 32218328 PMCID: PMC7181001 DOI: 10.3390/molecules25071489] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoid hormones are vital; their accurate operation is a necessity at all ages and in all life situations. Glucocorticoids regulate diverse physiological processes and they use many signaling pathways to fulfill their effect. As the operation of these hormones affects many organs, the excess of glucocorticoids is actually detrimental to the whole human body. The endogenous glucocorticoid excess is a relatively rare condition, but a significant proportion of adult people uses glucocorticoid medication for the treatment of chronic illnesses, therefore they are exposed to the side effects of long-term glucocorticoid treatment. Our review summarizes the adverse effects of glucocorticoid excess affecting bones, adipose tissue, brain and skin, focusing on those effects which involve the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
| | - Attila Patocs
- Hereditary Tumours Research Group, 1089 Budapest, Hungary;
- Department of Laboratory Medicine, Semmelweis University, 1089 Budapest, Hungary
- Department of Molecular Genetics, National Institute of Oncology, 1122 Budapest, Hungary
- Correspondence: ; Tel.: +36-1-266-0926; Fax: +36-1-266-0816
| |
Collapse
|
33
|
Cernackova A, Durackova Z, Trebaticka J, Mravec B. Neuroinflammation and depressive disorder: The role of the hypothalamus. J Clin Neurosci 2020; 75:5-10. [PMID: 32217047 DOI: 10.1016/j.jocn.2020.03.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/13/2020] [Accepted: 03/02/2020] [Indexed: 01/07/2023]
Abstract
Data accumulated over the last two decades has demonstrated that hypothalamic inflammation plays an important role in the etiopathogenesis of the most prevalent diseases, such as cardiovascular diseases, metabolic syndrome, and even cancer. Recent findings indicate that hypothalamic inflammation is also associated with stress exposure and certain psychiatric diseases, such as depressive disorder. Mechanistic studies have shown that intense and/or chronic stress exposure is accompanied by the synthesis of inflammatory molecules in the hypothalamus, altered hypothalamic-pituitary-adrenal axis activity, and development of glucocorticoid resistance. Consequently, these factors might play a role in the etiopathogenesis of psychiatric disorders. We propose that hypothalamic inflammation represents an interconnection between somatic diseases and depressive disorder. These assumptions are discussed in this mini-review in the light of available data from studies focusing on hypothalamic inflammation.
Collapse
Affiliation(s)
- Alena Cernackova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Slovakia; Institute of Experimental Endocrinology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Zdenka Durackova
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University in Bratislava, Slovakia
| | - Jana Trebaticka
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, Comenius University and Child University Hospital, Bratislava, Slovakia
| | - Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Slovakia; Institute of Experimental Endocrinology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
34
|
Puigoriol-Illamola D, Leiva R, Vázquez-Carrera M, Vázquez S, Griñán-Ferré C, Pallàs M. 11β-HSD1 Inhibition Rescues SAMP8 Cognitive Impairment Induced by Metabolic Stress. Mol Neurobiol 2019; 57:551-565. [PMID: 31399953 DOI: 10.1007/s12035-019-01708-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022]
Abstract
Ageing and obesity have been shown to increase the risk of cognitive decline and Alzheimer's disease (AD). Besides, elevated glucocorticoid (GCs) levels cause metabolic stress and have been associated with the neurodegenerative process. Direct pieces of evidence link the reduction of GCs caused by the inhibition of 11β-HSD type 1 (11β-HSD1) with cognitive improvement. In the present study, we investigated the beneficial effects of 11β-HSD1 inhibitor (i) RL-118 after high-fat diet (HFD) treatment in the senescence-accelerated mouse prone 8 (SAMP8). We found an improvement in glucose intolerance induced by HFD in mice treated with RL-118, a significant reduction in 11β-HSD1 and glucocorticoid receptor (GR) protein levels. Furthermore, specific modifications in the FGF21 activation after treatment with 11β-HSD1i, RL-118, which induced changes in SIRT1/PGC1α/AMPKα pathway, were found. Oxidative stress (OS) and reactive oxygen species (ROS), as well as inflammatory markers and microglial activation, were significantly diminished in HFD mice treated with 11β-HSD1i. Remarkably, treatment with 11β-HSD1i altered PERK pathway in both diet groups, increasing autophagy only in HFD mice group. After RL-118 treatment, a decrease in glycogen synthase kinase 3 (GSK3β) activation, Tau hyperphosphorylation, BACE1 protein levels and the product β-CTF were found. Increases in the non-amyloidogenic secretase ADAM10 protein levels and the product sAPPα were found in both treated mice, regardless of the diet. Consequently, beneficial effects on social behaviour and cognitive performance were found in treated mice. Thus, our results support the therapeutic strategy of selective 11β-HSD1i for the treatment of age-related cognitive decline and AD.
Collapse
Affiliation(s)
- Dolors Puigoriol-Illamola
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028, Barcelona, Spain.,Institute of Neuroscience, University of Barcelona (NeuroUB), Barcelona, Spain
| | - Rosana Leiva
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Department de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain.,Institute of Biomedicine, University of Barcelona (IBUB), Barcelona, Spain
| | - Manel Vázquez-Carrera
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028, Barcelona, Spain.,Institute of Biomedicine, University of Barcelona (IBUB), Barcelona, Spain.,Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain.,Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Santiago Vázquez
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Department de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, and Institute of Biomedicine (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain.,Institute of Biomedicine, University of Barcelona (IBUB), Barcelona, Spain
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028, Barcelona, Spain.,Institute of Neuroscience, University of Barcelona (NeuroUB), Barcelona, Spain
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av Joan XXIII 27-31, 08028, Barcelona, Spain. .,Institute of Neuroscience, University of Barcelona (NeuroUB), Barcelona, Spain.
| |
Collapse
|
35
|
Timmermans S, Souffriau J, Libert C. A General Introduction to Glucocorticoid Biology. Front Immunol 2019; 10:1545. [PMID: 31333672 PMCID: PMC6621919 DOI: 10.3389/fimmu.2019.01545] [Citation(s) in RCA: 350] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
Glucocorticoids (GCs) are steroid hormones widely used for the treatment of inflammation, autoimmune diseases, and cancer. To exert their broad physiological and therapeutic effects, GCs bind to the GC receptor (GR) which belongs to the nuclear receptor superfamily of transcription factors. Despite their success, GCs are hindered by the occurrence of side effects and glucocorticoid resistance (GCR). Increased knowledge on GC and GR biology together with a better understanding of the molecular mechanisms underlying the GC side effects and GCR are necessary for improved GC therapy development. We here provide a general overview on the current insights in GC biology with a focus on GC synthesis, regulation and physiology, role in inflammation inhibition, and on GR function and plasticity. Furthermore, novel and selective therapeutic strategies are proposed based on recently recognized distinct molecular mechanisms of the GR. We will explain the SEDIGRAM concept, which was launched based on our research results.
Collapse
Affiliation(s)
- Steven Timmermans
- Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jolien Souffriau
- Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Claude Libert
- Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
36
|
Ouanes S, Popp J. High Cortisol and the Risk of Dementia and Alzheimer's Disease: A Review of the Literature. Front Aging Neurosci 2019; 11:43. [PMID: 30881301 PMCID: PMC6405479 DOI: 10.3389/fnagi.2019.00043] [Citation(s) in RCA: 263] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/13/2019] [Indexed: 01/09/2023] Open
Abstract
Introduction: Cortisol effects on the brain are exerted through two distinct receptors, inducing complex and even opposite effects on the cerebral structures implicated in the various cognitive functions. High cortisol may also have deleterious effects on the brain structures and contribute to neurodegeneration, in particular Alzheimer’s disease (AD), via different mechanisms. Objective: To examine the interrelationships between cortisol, cognitive impairment and AD. Methods: Review of the literature. Results: Clinical studies found that elevated cortisol was associated with poorer overall cognitive functioning, as well as with poorer episodic memory, executive functioning, language, spatial memory, processing speed, and social cognition; while in animals, glucocorticoid administration resulted in cognitive impairment and abnormal behavior. In cognitively healthy subjects, higher cortisol levels have been associated with an increased risk of cognitive decline and AD. Subjects with dementia and Mild Cognitive Impairment (MCI) due to AD have been found to have higher CSF cortisol levels than cognitively healthy controls. Elevated CSF cortisol may also be associated with a more rapid cognitive decline in MCI due to AD. Elevated cortisol levels have been also found in delirium. High cortisol may mediate the impact of stressful life events, high neuroticism, depression, sleep disturbances, as well as cardiovascular risk factors on cognitive performance, neurodegeneration, and cognitive decline. High cortisol may also exert neurotoxic effects on the hippocampus, and promote oxidative stress and amyloid β peptide toxicity. Further possible underlying mechanisms include the interactions of cortisol with inflammatory mediators, neurotransmitters, and growth factors. Conclusion: Elevated cortisol levels may exert detrimental effects on cognition and contribute to AD pathology. Further studies are needed to investigate cortisol-reducing and glucocorticoidreceptor modulating interventions to prevent cognitive decline.
Collapse
Affiliation(s)
- Sami Ouanes
- Department of Psychiatry, Hospital of Cery, University Hospital of Lausanne, Lausanne, Switzerland.,Department of Psychiatry, Hamad Medical Corporation, Doha, Qatar
| | - Julius Popp
- Department of Psychiatry, Hospital of Cery, University Hospital of Lausanne, Lausanne, Switzerland.,Geriatric Psychiatry, Department of Mental Health and Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
37
|
Qian X, Wang ZR, Zheng JJ, Ding JQ, Zhong JG, Zhang TY, Li W, Zhang M. Baicalein improves cognitive deficits and hippocampus impairments in temporal lobe epilepsy rats. Brain Res 2019; 1714:111-118. [PMID: 30817901 DOI: 10.1016/j.brainres.2019.02.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/20/2019] [Accepted: 02/23/2019] [Indexed: 11/16/2022]
Abstract
Temporal lobe epilepsy (TLE) is a chronic neurological disorder that is a refractory disease. Baicalein possesses various pharmacological activities, including neuroprotection in neurodegenerative disease. However, whether baicalein is protective in the treatment of TLE is not determined. Therefore, the present study investigated the role of baicalein in the treatment of TLE. Baicalein was injected intraperitoneally to TLE rats for two weeks after the onset of spontaneous recurrent seizures (SRS). Rats were observed for the occurrence of SRS, and cognitive and hippocampus injuries were evaluated. Oxidative stress and inflammatory cytokines were measured. Corticosterone and its receptor, actin-associated protein F-actin and cofilin-1 were investigated in the brains of epileptic rats. Baicalein significantly improved cognition and reduced hippocampus damage and mossy fibre sprouting in TLE rats without obvious SRS suppression. Baicalein produced excellent anti-oxidative and anti-inflammatory effects in TLE rats. Baicalein restored the disruption of the glucocorticoid signal pathway and actin-associated protein in TLE rats. These results suggest that the neuroprotective effects of baicalein on cognition and the hippocampus are associated with the suppression of oxidative stress and inflammation and the regulation of the glucocorticoid pathway and actin-associated protein in TLE rats. This evidence supports the use of baicalein as an adjuvant agent for epilepsy treatment.
Collapse
Affiliation(s)
- Xu Qian
- Department of Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University and Key Laboratory of Molecular Target & Clinical Pharmacology, Guangzhou, Guangdong, China
| | - Zhao-Rui Wang
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Jing-Jun Zheng
- Department of Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University and Key Laboratory of Molecular Target & Clinical Pharmacology, Guangzhou, Guangdong, China
| | - Ji-Qiang Ding
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Jia-Gui Zhong
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Teng-Yue Zhang
- Department of Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University and Key Laboratory of Molecular Target & Clinical Pharmacology, Guangzhou, Guangdong, China
| | - Wei Li
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China.
| | - Mei Zhang
- Department of Clinical Pharmacology, School of Pharmaceutical Science, Guangzhou Medical University and Key Laboratory of Molecular Target & Clinical Pharmacology, Guangzhou, Guangdong, China.
| |
Collapse
|
38
|
Altered hippocampal function with preserved cognitive performance in treatment-naive major depressive disorder. Neuroreport 2019; 30:46-52. [PMID: 30422941 DOI: 10.1097/wnr.0000000000001163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The hippocampus is implicated in the pathophysiology of major depressive disorder (MDD), with evidence that morphological changes occur with disease progression. It was hypothesized that treatment-naive patients with depression would show performance deficits in hippocampus-dependent memory trials, with concurrent hippocampal activation deficits on functional magnetic resonance imaging, compared with control participants. Thirteen treatment-naive patients with MDD and 13 control participants completed a hippocampus-dependent memory functional magnetic resonance imaging process-dissociation task. On behavioural measures of habit memory and guessing, there were no significant differences between groups. Functional magnetic resonance imaging analysis indicated that compared with the control group, the MDD group showed increased activation in the parahippocampal gyrus and hippocampus on habit memory and nonitem trials. These alterations in hippocampal functioning with preserved cognitive performance on a test of hippocampus-dependent memory in MDD may be indicative of a compensatory mechanism.
Collapse
|
39
|
Alegría-Herrera E, Herrera-Ruiz M, Román-Ramos R, Zamilpa A, Santillán-Urquiza MA, Aguilar MI, Avilés-Flores M, Fuentes-Mata M, Jiménez-Ferrer E. Effect of Ocimum basilicum, Ocimum selloi, and Rosmarinic Acid on Cerebral Vascular Damage in a Chronic Hypertension Model. Biol Pharm Bull 2019; 42:201-211. [PMID: 30713252 DOI: 10.1248/bpb.b18-00574] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The main objective of treatment against hypertension is not only to reduce blood pressure levels, but also to reduce vascular risk in general. In the present work, administering angiotensin II (AGII; 0.2 µg/kg intraperitoneally (i.p.) for 12 weeks) activates the hypothalamic-pituitary-adrenal (HPA) axis, which caused an increase in corticosterone levels, as well as in proinflammatory cytokines (interleukin 1β (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-α)) and macrophage chemotactic protein 1 (MCP-1), and decreased anti-inflammatory cytokines (interleukin 10 (IL-10) and interleukin 4 (IL-4)). On observing the behavior in the different models, an anxiogenic effect (elevated plus maze (EPM)) and cognitive impairment (water Morris maze (WMM)) was observed in animals with AGII. By administering organic extracts from Ocimum basilicum (Oba-EtOAc) and Ocimum selloi (Ose-EtOAc), and some doses of rosmarinic acid (RA) (6 weeks per os (p.o.)), the damage caused by AGII was stopped by re-establishing corticosterone serum levels and by decreasing the proinflammatory cytokines and MCP-1.
Collapse
Affiliation(s)
- Elian Alegría-Herrera
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS)
- Doctorado en Ciencias Biológicas y de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa
- Departamento Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa
| | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS)
| | - Rubén Román-Ramos
- Departamento Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS)
| | | | - María Isabel Aguilar
- Laboratorio 111, Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México
| | | | | | | |
Collapse
|
40
|
Baker JD, Ozsan I, Rodriguez Ospina S, Gulick D, Blair LJ. Hsp90 Heterocomplexes Regulate Steroid Hormone Receptors: From Stress Response to Psychiatric Disease. Int J Mol Sci 2018; 20:ijms20010079. [PMID: 30585227 PMCID: PMC6337637 DOI: 10.3390/ijms20010079] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 01/30/2023] Open
Abstract
The hypothalamus-pituitary-adrenal (HPA) axis directly controls the stress response. Dysregulation of this neuroendocrine system is a common feature among psychiatric disorders. Steroid hormone receptors, like glucocorticoid receptor (GR), function as transcription factors of a diverse set of genes upon activation. This activity is regulated by molecular chaperone heterocomplexes. Much is known about the structure and function of these GR/heterocomplexes. There is strong evidence suggesting altered regulation of steroid receptor hormones by chaperones, particularly the 51 kDa FK506-binding protein (FKBP51), may work with environmental factors to increase susceptibility to various psychiatric illnesses including post-traumatic stress disorder (PTSD), major depressive disorder (MDD), and anxiety. This review highlights the regulation of steroid receptor dynamics by the 90kDa heat shock protein (Hsp90)/cochaperone heterocomplexes with an in depth look at how the structural regulation and imbalances in cochaperones can cause functional effects on GR activity. Links between the stress response and circadian systems and the development of novel chaperone-targeting therapeutics are also discussed.
Collapse
Affiliation(s)
- Jeremy D Baker
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Ilayda Ozsan
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Santiago Rodriguez Ospina
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Danielle Gulick
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| | - Laura J Blair
- USF Health Byrd Institute, Morsani College of Medicine, Department of Molecular Medicine, University of South Florida, 4001 East Fowler Ave, Tampa, FL 33613, USA.
| |
Collapse
|
41
|
Bahramzadeh Zoeram S, Elahdadi Salmani M, Lashkarbolouki T, Goudarzi I. Hippocampal orexin receptor blocking prevented the stress induced social learning and memory deficits. Neurobiol Learn Mem 2018; 157:12-23. [PMID: 30458283 DOI: 10.1016/j.nlm.2018.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 10/14/2018] [Accepted: 11/16/2018] [Indexed: 01/09/2023]
Abstract
Stress as a homeostatic challenge leads to the malfunction of learning and memory processes, namely social learning and memory. The orexin system is involved in stress responses through connections to the hypothalamic-pituitary axis (HPA). In addition, the hippocampus, a structure vulnerable to stress-induced changes, expresses orexin receptors 1 and 2 (OXr1 and OXr2) in various sub-regions. The present study is aimed at assessing the effects of hippocampal orexin receptor blockade on social learning and memory impairments and anxiety development following stress. Male Wistar rats (220-250 g) underwent cannula implantation in the hippocampus. Acute (two mild electric shocks, 5.5 mA) and chronic stresses (ten days of restraint, 6 h daily) were applied with or without injection of orexin receptor antagonists (SB-334867 or TCS OX 29). Sociability and social novelty in animals were assessed in a three-chamber social maze at the end of stress application. Anxiety and exploratory behavior of animals were then examined, with 20 min intervals, using the open field (OF) and elevated plus maze (EPM) tests, respectively. Cisterna Magna cerebro-spinal fluid (CSF) was drained, before sacrifice, for orexin (OX) assay and trunk blood was collected to measure the plasma corticosterone (CRT). Neither the acute nor the chronic stress could affect the sociability. The acute but not chronic stress prevented the animal from sniffing the familiar caged rat in the novelty session, a response which was reversed following the blockade of both OXRs. Furthermore, acute but not chronic stress, led to increased anxiety and immobility behavior which were both impeded by blocking the orexin receptor (OXR). Conversely, OX content in CSF increased due to chronic restraint stress, an effect that was reversed by orexin blockade. Finally, elevated plasma CRT was recorded in response to both acute and chronic stresses. The observed increase in plasma CRT in chronically-stressed rats was abolished following inhibition of OXRs, however a similar effect was not seen in the acute-stress group. Our results identify hippocampal OXRs as potential candidates capable of preventing acute stress-induced impairments of social novelty and anxiety behavior, and chronic stress-induced plasma CRT and CSF orexin, changes. OXR manipulation may improve adaptation to stress pathophysiology.
Collapse
Affiliation(s)
| | | | | | - Iran Goudarzi
- School of Biology, Damghan University, Damghan, Iran
| |
Collapse
|
42
|
Roat-Shumway S, Wroolie TE, Watson K, Schatzberg AF, Rasgon NL. Cognitive effects of mifepristone in overweight, euthymic adults with depressive disorders. J Affect Disord 2018; 239:242-246. [PMID: 30025313 DOI: 10.1016/j.jad.2018.07.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/17/2018] [Accepted: 07/07/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Previous studies have shown that individuals with mood disorders have a higher prevalence of both hypercortisolemia and insulin resistance. Insulin resistance is posited to contribute to the cognitive deficits observed in individuals who have depression. However, the mechanistic relationship between cortisol and insulin within the central nervous system remains to be further elucidated. This study aimed to evaluate the effects of the antiglucocorticoid agent, mifepristone, on metabolic function and cognitive performance in individuals receiving treatment for depressive disorders who were euthymic at baseline. METHODS Participants were administered a 600 mg/day dose of mifepristone for 28 days. Oral glucose tolerance tests (OGTTs) and cognitive assessments measuring verbal memory and executive functioning were administered at baseline and after 28 days of treatment. RESULTS Improvements in attention and verbal learning were associated with reduction of fasting plasma glucose (FPG) in response to mifepristone treatment. LIMITATIONS Limitations include the open-label design of this study and a small sample size. CONCLUSIONS The findings from this study suggest that improvement in fasting plasma glucose levels, upon administration of mifepristone, is associated with the improvement in early input of verbal information. Further studies are warranted in order to better evaluate the use of mifepristone or other antiglucocorticoid agents in treatment of mood disorders characterized by metabolic dysfunction.
Collapse
Affiliation(s)
- S Roat-Shumway
- Stanford University School of Medicine, Department of Psychiatry & Behavioral Sciences, 401 Quarry Road, Stanford, CA 94305-5723, United States
| | - T E Wroolie
- Stanford University School of Medicine, Department of Psychiatry & Behavioral Sciences, 401 Quarry Road, Stanford, CA 94305-5723, United States
| | - K Watson
- Stanford University School of Medicine, Department of Psychiatry & Behavioral Sciences, 401 Quarry Road, Stanford, CA 94305-5723, United States
| | - A F Schatzberg
- Stanford University School of Medicine, Department of Psychiatry & Behavioral Sciences, 401 Quarry Road, Stanford, CA 94305-5723, United States
| | - N L Rasgon
- Stanford University School of Medicine, Department of Psychiatry & Behavioral Sciences, 401 Quarry Road, Stanford, CA 94305-5723, United States.
| |
Collapse
|
43
|
Ali SA, Begum T, Reza F. Hormonal Influences on Cognitive Function. Malays J Med Sci 2018; 25:31-41. [PMID: 30914845 PMCID: PMC6422548 DOI: 10.21315/mjms2018.25.4.3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/10/2018] [Indexed: 11/09/2022] Open
Abstract
This article examines how hormonal changes may affect the neuronal networking and mechanisms of cognitive function. Hormones are the chemical regulators of the human body and function critically to maintain various processes, such as growth, emotions and even cognition. Numerous studies have examined the relationship between hormonal effects and cognitive function; these studies have investigated different factors, such as aging, pregnancy, post-natal states, emotions and stress. Different types of hormones produce different outcomes for the human body and mind. Hormones may also contribute to both positive and negative outcomes, depending on whether the hormone levels are too low or too high. To investigate the hormonal effects on cognitive function, the sources of localisation must be localised, so that the neuronal network can be realised. Furthermore, cognitive function does not rely on a specific brain region but is determined by the neuronal network interactions. Thus, it is worthwhile to know the neural mechanisms behind cognitive functions that are affected by hormones.
Collapse
Affiliation(s)
- Siti Atiyah Ali
- Department of Neurosciences, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Tahamina Begum
- Department of Neurosciences, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Faruque Reza
- Department of Neurosciences, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
44
|
A general neurologist's perspective on the urgent need to apply resilience thinking to the prevention and treatment of Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 3:498-506. [PMID: 29124107 PMCID: PMC5671621 DOI: 10.1016/j.trci.2017.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The goal of this article was to look at the problem of Alzheimer's disease (AD) through the lens of a socioecological resilience-thinking framework to help expand our view of the prevention and treatment of AD. This serious and complex public health problem requires a holistic systems approach. We present the view that resilience thinking, a theoretical framework that offers multidisciplinary approaches in ecology and natural resource management to solve environmental problems, can be applied to the prevention and treatment of AD. Resilience thinking explains a natural process that occurs in all complex systems in response to stressful challenges. The brain is a complex system, much like an ecosystem, and AD is a disturbance (allostatic overload) within the ecosystem of the brain. Resilience thinking gives us guidance, direction, and ideas about how to comprehensively prevent and treat AD and tackle the AD epidemic.
Collapse
|
45
|
Kataja EL, Karlsson L, Huizink AC, Tolvanen M, Parsons C, Nolvi S, Karlsson H. Pregnancy-related anxiety and depressive symptoms are associated with visuospatial working memory errors during pregnancy. J Affect Disord 2017; 218:66-74. [PMID: 28458118 DOI: 10.1016/j.jad.2017.04.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/29/2017] [Accepted: 04/07/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cognitive deficits, especially in memory and concentration, are often reported during pregnancy. Similar cognitive dysfunctions can also occur in depression and anxiety. To date, few studies have investigated the associations between cognitive deficits and psychiatric symptoms during pregnancy. This field is of interest because maternal cognitive functioning, and particularly its higher-order aspects are related to maternal well-being and caregiving behavior, as well as later child development. METHODS Pregnant women (N =230), reporting low (n =87), moderate (n =97), or high (n =46) levels of depressive, general anxiety and/or pregnancy-related anxiety symptoms (assessed repeatedly with EPDS, SCL-90/anxiety subscale, PRAQ-R2, respectively) were tested in mid-pregnancy for their cognitive functions. A computerized neuropsychological test battery was used. RESULTS Pregnant women with high or moderate level of psychiatric symptoms had significantly more errors in visuospatial working memory/executive functioning task than mothers with low symptom level. Depressive symptoms throughout pregnancy and concurrent pregnancy-related anxiety symptoms were significant predictors of the performance in the task. General anxiety symptoms were not related to visuospatial working memory. LIMITATIONS Cognitive functions were evaluated only at one time-point during pregnancy precluding causal conclusions. CONCLUSIONS Maternal depressive symptoms and pregnancy-related anxiety symptoms were both associated with decrements in visuospatial working memory/executive functioning. Depressive symptoms seem to present more stable relationship with cognitive deficits, while pregnancy-related anxiety was associated only concurrently. Future studies could investigate, how stable these cognitive differences are, and whether they affect maternal ability to deal with demands of pregnancy and later parenting.
Collapse
Affiliation(s)
- E-L Kataja
- Department of Psychology, University of Turku, Finland; FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Finland.
| | - L Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Finland; Department of Child Psychiatry, Turku University Hospital and University of Turku, Finland
| | - A C Huizink
- Department of Clinical Developmental Psychology, VU University Amsterdam, Netherlands
| | - M Tolvanen
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Finland; Department of Community Dentistry, University of Turku, Finland
| | - C Parsons
- Department of Psychiatry, University of Oxford, UK; Interacting Minds Center, Department of Clinical Medicine, Aarhus University, Denmark
| | - S Nolvi
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Finland
| | - H Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Finland; Department of Psychiatry, University of Turku, Finland
| |
Collapse
|
46
|
Pesce M, Tatangelo R, La Fratta I, Rizzuto A, Campagna G, Turli C, Ferrone A, Franceschelli S, Speranza L, Verrocchio MC, De Lutiis MA, Felaco M, Grilli A. Memory Training Program Decreases the Circulating Level of Cortisol and Pro-inflammatory Cytokines in Healthy Older Adults. Front Mol Neurosci 2017; 10:233. [PMID: 28790890 PMCID: PMC5522887 DOI: 10.3389/fnmol.2017.00233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 07/06/2017] [Indexed: 12/31/2022] Open
Abstract
Aging cognitive decline has been associated to impairment of the Hypothalamus Pituitary Adrenals (HPA) axis activity and a higher level of the systemic inflammation. However, little is known about the molecules driving this process at peripheral level. In addition, the cognitive function is to some extent modifiable with Memory Training (MT) programs, even among older adults and beyond. The study aims to evaluate whether MT could contribute to ameliorate cognitive performance and modulate the HPA axis activity as well the low level inflammation in the aging phenotype. Whether the phosphatase WIP-1, a negative regulator for inflammation, is involved in this process was also investigated. We recruited 31 young adults (19-28, years of age) and 62 older adults aged over 60. Thirty-two older adults were submitted to 6-months of MT program (EG), and 28 older adults were no treated and used as Control Group (CG). Global cognitive functioning (MMSE score), verbal and visual memory, and attention were assessed at baseline (T0) and after 6-months (T1). At the same time, plasmatic level of Cortisol (C), IL-1β, IL-18, IL-6, and the expression of WIP-1 mRNA and protein in ex vivo Peripheral Blood Mononuclear Cells were analyzed in young adults at T0, as well in older adults at T0 and T1. Together, the results suggest that MT improves the global cognitive functionality, verbal and visual memory, as well as the level of attention. At the same time we observed a decrease of the plasmatic level of C, of the cytokines, and an increase of the expression of mRNA and protein of WIP-1. The analysis of correlations highlighted that the level of the mRNA of WIP-1 was positively associated to the MMSE score, and negatively to the C and cytokine levels. In conclusion, we purpose the MT as tool that could help support successful aging through the improving of memory, attention and global cognitive function performance. Furthermore, this approach could participate to maintain lower the peripheral levels of the C and pro-inflammatory cytokines. The WIP-1 as a potential new target of the pathophysiology of aging is theorized.
Collapse
Affiliation(s)
| | - Raffaella Tatangelo
- School of Medicine and Health Science, University G. D’AnnunzioChieti, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ouanes S, Castelao E, von Gunten A, Vidal PM, Preisig M, Popp J. Personality, Cortisol, and Cognition in Non-demented Elderly Subjects: Results from a Population-Based Study. Front Aging Neurosci 2017; 9:63. [PMID: 28352228 PMCID: PMC5348534 DOI: 10.3389/fnagi.2017.00063] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/01/2017] [Indexed: 12/29/2022] Open
Abstract
Certain personality traits, in particular higher neuroticism, have been associated, on one hand, with elevated cortisol levels, and on the other hand, with poorer cognitive performance. At the same time, several studies highlighted the association between high cortisol and poor cognitive functioning. Here, we hypothesized that increased cortisol may be associated with poorer cognition and with certain personality traits (mainly high neuroticism), and that personality might explain the association between cortisol and cognition. A cross-sectional analysis was conducted using data from Colaus/PsyColaus, a population-based study involving residents of Lausanne, Switzerland. Salivary cortisol samples (upon waking, 30 min after waking, at 11 am and at 8 pm) along with cognitive and personality measures were obtained from 643 non-demented participants aged at least 65. Personality traits were assessed using the NEO Five-Factor Inventory (NEO-FFI). We examined the links between the cortisol Area under the Curve (AUC), the Clinical Dementia Rating Sum of Boxes (CDRSOB) and the NEO-FFI scores. No association was found between personality traits and the CDRSOB or the MMSE score, controlling for age, sex, depression, education and BMI. However, the executive functioning domain z-score was negatively associated with agreeableness (p = 0.005; slope = -0.107 [-0.181; -0.033]) and openness (p = 0.029; slope = -0.081 [-0.154; -0.008]) after controlling for age, sex, depression, education and BMI. The CDRSOB score was positively associated with the cortisol AUC after controlling for age, sex, BMI, education and depression, (p = 0.003; slope = 0.686 [0.240; 1.333]). This association remained significant after controlling for personality traits and for the interaction between personality traits and the cortisol AUC (p = 0.006; slope = 0.792 [0.233; 1.352]. High agreeableness and openness might be associated with poorer executive performance in later life. Increased cortisol may be associated with both specific personality traits (high extraversion, low openness) and worse cognitive performance. Increased salivary cortisol does not mediate the relationship between personality traits and cognitive impairment.
Collapse
Affiliation(s)
- Sami Ouanes
- Department of Psychiatry, University Hospital of Lausanne Lausanne, Switzerland
| | - Enrique Castelao
- Department of Psychiatry, University Hospital of Lausanne Lausanne, Switzerland
| | - Armin von Gunten
- Department of Psychiatry, University Hospital of Lausanne Lausanne, Switzerland
| | - Pedro M Vidal
- Department of Internal Medicine, University Hospital of Lausanne Lausanne, Switzerland
| | - Martin Preisig
- Department of Psychiatry, University Hospital of Lausanne Lausanne, Switzerland
| | - Julius Popp
- Department of Psychiatry, University Hospital of Lausanne Lausanne, Switzerland
| |
Collapse
|
48
|
Ouanes S, Castelao E, Gebreab S, von Gunten A, Preisig M, Popp J. Life events, salivary cortisol, and cognitive performance in nondemented subjects: a population-based study. Neurobiol Aging 2017; 51:1-8. [DOI: 10.1016/j.neurobiolaging.2016.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 11/15/2016] [Accepted: 11/19/2016] [Indexed: 02/01/2023]
|