1
|
Singh P, Vasundhara B, Das N, Sharma R, Kumar A, Datusalia AK. Metabolomics in Depression: What We Learn from Preclinical and Clinical Evidences. Mol Neurobiol 2024:10.1007/s12035-024-04302-5. [PMID: 38898199 DOI: 10.1007/s12035-024-04302-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
Depression is one of the predominant common mental illnesses that affects millions of people of all ages worldwide. Random mood changes, loss of interest in routine activities, and prevalent unpleasant senses often characterize this common depreciated mental illness. Subjects with depressive disorders have a likelihood of developing cardiovascular complications, diabesity, and stroke. The exact genesis and pathogenesis of this disease are still questionable. A significant proportion of subjects with clinical depression display inadequate response to antidepressant therapies. Hence, clinicians often face challenges in predicting the treatment response. Emerging reports have indicated the association of depression with metabolic alterations. Metabolomics is one of the promising approaches that can offer fresh perspectives into the diagnosis, treatment, and prognosis of depression at the metabolic level. Despite numerous studies exploring metabolite profiles post-pharmacological interventions, a quantitative understanding of consistently altered metabolites is not yet established. The article gives a brief discussion on different biomarkers in depression and the degree to which biomarkers can improve treatment outcomes. In this review article, we have systemically reviewed the role of metabolomics in depression along with current challenges and future perspectives.
Collapse
Affiliation(s)
- Pooja Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, 226002, India
| | - Boosani Vasundhara
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, 226002, India
| | - Nabanita Das
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, 226002, India
| | - Ruchika Sharma
- Centre for Precision Medicine and Centre, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | - Anoop Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, 226002, India.
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, 226002, India.
| |
Collapse
|
2
|
Agranyoni O, Sur D, Amidror S, Shidlovsky N, Bagaev A, Yissachar N, Pinhasov A, Navon-Venezia S. Colon impairments and inflammation driven by an altered gut microbiota leads to social behavior deficits rescued by hyaluronic acid and celecoxib. BMC Med 2024; 22:182. [PMID: 38685001 PMCID: PMC11059729 DOI: 10.1186/s12916-024-03323-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/27/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The exact mechanisms linking the gut microbiota and social behavior are still under investigation. We aimed to explore the role of the gut microbiota in shaping social behavior deficits using selectively bred mice possessing dominant (Dom) or submissive (Sub) behavior features. Sub mice exhibit asocial, depressive- and anxiety-like behaviors, as well as systemic inflammation, all of which are shaped by their impaired gut microbiota composition. METHODS An age-dependent comparative analysis of the gut microbiota composition of Dom and Sub mice was performed using 16S rRNA sequencing, from early infancy to adulthood. Dom and Sub gastrointestinal (GI) tract anatomy, function, and immune profiling analyses were performed using histology, RT-PCR, flow cytometry, cytokine array, and dextran-FITC permeability assays. Short chain fatty acids (SCFA) levels in the colons of Dom and Sub mice were quantified using targeted metabolomics. To support our findings, adult Sub mice were orally treated with hyaluronic acid (HA) (30 mg/kg) or with the non-steroidal anti-inflammatory agent celecoxib (16 mg/kg). RESULTS We demonstrate that from early infancy the Sub mouse gut microbiota lacks essential bacteria for immune maturation, including Lactobacillus and Bifidobacterium genera. Furthermore, from birth, Sub mice possess a thicker colon mucin layer, and from early adulthood, they exhibit shorter colonic length, altered colon integrity with increased gut permeability, reduced SCFA levels and decreased regulatory T-cells, compared to Dom mice. Therapeutic intervention in adult Sub mice treated with HA, celecoxib, or both agents, rescued Sub mice phenotypes. HA treatment reduced Sub mouse gut permeability, increased colon length, and improved mouse social behavior deficits. Treatment with celecoxib increased sociability, reduced depressive- and anxiety-like behaviors, and increased colon length, and a combined treatment resulted in similar effects as celecoxib administered as a single agent. CONCLUSIONS Overall, our data suggest that treating colon inflammation and decreasing gut permeability can restore gut physiology and prevent social deficits later in life. These findings provide critical insights into the importance of early life gut microbiota in shaping gut immunity, functionality, and social behavior, and may be beneficial for the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Oryan Agranyoni
- Department of Molecular Biology and the Dr. Miriam and Sheldon G. School of Medicine, Ariel University, Ariel, Israel
| | - Debpali Sur
- Department of Molecular Biology and the Dr. Miriam and Sheldon G. School of Medicine, Ariel University, Ariel, Israel
| | - Sivan Amidror
- The Goodman Faculty of Life Sciences, Bar-Ilan Institute of Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, Israel
| | - Nuphar Shidlovsky
- The Goodman Faculty of Life Sciences, Bar-Ilan Institute of Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, Israel
| | - Anastasia Bagaev
- Department of Molecular Biology and the Dr. Miriam and Sheldon G. School of Medicine, Ariel University, Ariel, Israel
| | - Nissan Yissachar
- The Goodman Faculty of Life Sciences, Bar-Ilan Institute of Nanotechnology and Advanced Materials, Bar Ilan University, Ramat Gan, Israel
| | - Albert Pinhasov
- Department of Molecular Biology and the Dr. Miriam and Sheldon G. School of Medicine, Ariel University, Ariel, Israel.
| | - Shiri Navon-Venezia
- Department of Molecular Biology and the Dr. Miriam and Sheldon G. School of Medicine, Ariel University, Ariel, Israel.
| |
Collapse
|
3
|
Sur D, Agranyoni O, Kirby M, Cohen N, Bagaev A, Karandasheva K, Shmerkin E, Gorobets D, Savita BK, Avneri R, Divon MS, Lax E, Michaelevski I, Pinhasov A. Nurture outpaces nature: fostering with an attentive mother alters social dominance in a mouse model of stress sensitivity. Mol Psychiatry 2023; 28:3816-3828. [PMID: 37845494 DOI: 10.1038/s41380-023-02273-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 10/18/2023]
Abstract
Maternal care is critical for epigenetic programming during postnatal brain development. Stress is recognized as a critical factor that may affect maternal behavior, yet owing to high heterogeneity in stress response, its impact varies among individuals. We aimed here to understand the connection between inborn stress vulnerability, maternal care, and early epigenetic programming using mouse populations that exhibit opposite poles of the behavioral spectrum (social dominance [Dom] and submissiveness [Sub]) and differential response to stress. In contrast to stress-resilient Dom dams, stress-vulnerable Sub dams exhibit significantly lower maternal attachment, serum oxytocin, and colonic Lactobacillus reuteri populations. Sub offspring showed a reduced hippocampal expression of key methylation genes at postnatal day (PND) 7 and a lack of developmentally-dependent increase in 5-methylcytosine (5-mC) at PND 21. In addition, Sub pups exhibit significant hypermethylation of gene promoters connected with glutamatergic synapses and behavioral responses. We were able to reverse the submissive endophenotype through cross-fostering Sub pups with Dom dams (Sub/D). Thus, Sub/D pups exhibited elevated hippocampal expression of DNMT3A at PND 7 and increased 5-mC levels at PND 21. Furthermore, adult Sub/D offspring exhibited increased sociability, social dominance, and hippocampal glutamate and monoamine levels resembling the neurochemical profile of Dom mice. We postulate that maternal inborn stress vulnerability governs epigenetic patterning sculpted by maternal care and intestinal microbiome diversity during early developmental stages and shapes the array of gene expression patterns that may dictate neuronal architecture with a long-lasting impact on stress sensitivity and the social behavior of offspring.
Collapse
Affiliation(s)
- Debpali Sur
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Oryan Agranyoni
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Michael Kirby
- Dr. Miriam and Sheldon G. Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Naamah Cohen
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Anastasia Bagaev
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Kristina Karandasheva
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Elena Shmerkin
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Denis Gorobets
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Brajesh Kumar Savita
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Raphael Avneri
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Mali-Salmon Divon
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
- Dr. Miriam and Sheldon G. Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Elad Lax
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Izhak Michaelevski
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel
| | - Albert Pinhasov
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ramat HaGolan St 65, 4077625, Ariel, Israel.
- Dr. Miriam and Sheldon G. Adelson School of Medicine, Ariel University, Ariel, Israel.
| |
Collapse
|
4
|
Gao J, Zhao L, Li D, Li Y, Wang H. Enriched environment ameliorates postsurgery sleep deprivation-induced cognitive impairments through the AMPA receptor GluA1 subunit. Brain Behav 2023; 13:e2992. [PMID: 37095708 PMCID: PMC10275526 DOI: 10.1002/brb3.2992] [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: 01/03/2023] [Revised: 02/25/2023] [Accepted: 03/20/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND As a common postsurgery complication, sleep deprivation (SD) can severely deteriorate the cognitive function of patients. Enriched environment (EE) exposure can increase children's cognitive ability, and whether EE exposure could be utilized to alleviate postsurgery SD-induced cognitive impairments is investigated in this study. METHODS Open inguinal hernia repair surgery without skin/muscle retraction was performed on Sprague-Dawley male rats (9-week-old), which were further exposed to EE or standard environment (SE). Elevated plus maze (EPM), novel object recognition (NOR), object location memory (OLM), and Morris Water Maze assays were utilized to monitor cognitive functions. Cresyl violet acetate staining in the Cornusammonis 3 (CA3) region of rat hippocampus was used to detect neuron loss. The relative expression of brain-derived neurotrophic factor (BDNF) and synaptic glutamate receptor 1 (GluA1) subunits in the hippocampus were detected with quantitative reverse transcription polymerase chain reaction (RT-qPCR), Western blots, enzyme-linked immunosorbent assay (ELISA), and immunofluorescence. RESULTS EE restored normal levels of time spent in the center, time in distal open arms, open/total arms ratio, and total distance traveled in the EPM test; EE restored normal levels of recognition index in the NOR and OLM test; EE restored normal levels of time in the target quadrant, escape latencies, and platform site crossings in the Morris Water Maze test. EE exposure decreased neuron loss in the CA3 region of the hippocampus with increased BDNF and phosphorylated (p)-GluA1 (ser845) expression. CONCLUSION EE ameliorates postsurgery SD-induced cognitive impairments, which may be mediated by the axis of BDNF/GluA1. EE exposure could be considered as an aid in promoting cognitive function in postsurgery SD.
Collapse
Affiliation(s)
- Jie Gao
- Department of Anesthesiologythe Third Central Clinical College of Tianjin Medical University, Nankai University Affinity the Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary DiseaseTianjinChina
- Department of AnesthesiologyTianjin Haihe HospitalTianjinChina
| | - Lina Zhao
- Department of Anesthesiologythe Third Central Clinical College of Tianjin Medical University, Nankai University Affinity the Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary DiseaseTianjinChina
| | - Dedong Li
- Department of Anesthesiologythe Third Central Clinical College of Tianjin Medical University, Nankai University Affinity the Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary DiseaseTianjinChina
| | - Yun Li
- Department of Anesthesiologythe Third Central Clinical College of Tianjin Medical University, Nankai University Affinity the Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary DiseaseTianjinChina
| | - Haiyun Wang
- Department of Anesthesiologythe Third Central Clinical College of Tianjin Medical University, Nankai University Affinity the Third Central Hospital, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary DiseaseTianjinChina
| |
Collapse
|
5
|
Ping A, Yang M, Xu S, Li Q, Feng Y, Gao K, Wang S, Duan K. Correlations between GRIN2B and GRIN3A gene polymorphisms and postpartum depressive symptoms in Chinese parturients undergoing cesarean section: A prospective cohort study. J Psychosom Res 2023; 168:111210. [PMID: 36898314 DOI: 10.1016/j.jpsychores.2023.111210] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/12/2023]
Abstract
OBJECTIVE To investigate the association of postpartum depressive symptoms (PDS) and self-harm ideation with n-methyl-d-aspartate (NMDA) receptor GRIN2B and GRIN3A gene polymorphisms and other risk factors in women undergoing cesarean section. METHODS A total of 362 parturients undergoing cesarean section under lumbar anesthesia were selected and their postpartum depression level was assessed by the Edinburgh Postpartum Depression Scale (EPDS) at 42 days postpartum, with an EPDS score of 9/10 as the cut-off value. Three GRIN2B SNP loci (rs1805476, rs3026174, rs4522263) and five GRIN3A SNP loci (rs1983812, rs2050639, rs2050641, rs3739722, rs10989563) were selected for genotype detection. The role of each SNP, linkage disequilibrium and haplotypes in the development of postpartum depression was analyzed. Logistic regression analysis was performed for related risk factors. RESULTS PDS incidence was 16.85%, and self-harm ideation incidence was 13.54%. Univariate analysis showed that GRIN2B rs1805476, rs3026174 and rs4522263 gene polymorphisms were associated with PDS (p < 0.05), with GRIN2B rs4522263 gene also associated with maternal self-harm ideation. GRIN3A rs1983812, rs2050639, rest rs2050641, rs3739722 and rs10989563 alleles were not associated with PDS. Logistic regression analysis indicated that high pregnancy stress, as well as rs1805476 and rs4522263 alleles were PDS risk factors following cesarean delivery. GRIN2B (TTG p = 0.002) and GRIN3A (TGTTC p = 0.002) haplotypes were associated with the lower PDS incidence and higher PDS incidence respectively. CONCLUSION GRIN2B rs1805476 GG genotype, rs4522263 CC genotype and high stress during pregnancy were risk factors for PDS, whilst a significantly higher incidence of self-harm ideation was evident in parturients carrying GRIN2B rs4522263 CC genotype.
Collapse
Affiliation(s)
- Anqi Ping
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Mi Yang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Shouyu Xu
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Qiuwen Li
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Yunfei Feng
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Kai Gao
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Saiying Wang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China.
| | - Kaiming Duan
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China.
| |
Collapse
|
6
|
Becker M, Abaev K, Pinhasov A, Ornoy A. S-Adenosyl-Methionine alleviates sociability aversion and reduces changes in gene expression in a mouse model of social hierarchy. Behav Brain Res 2022; 427:113866. [DOI: 10.1016/j.bbr.2022.113866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 02/06/2023]
|
7
|
Spatial working memory is disparately interrelated with social status through different developmental stages in rats. Behav Brain Res 2022; 416:113547. [PMID: 34437940 DOI: 10.1016/j.bbr.2021.113547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/15/2021] [Accepted: 08/21/2021] [Indexed: 02/02/2023]
Abstract
Social life necessitates cognitive competence to meet the dynamic demands of social development. The formation of dominance hierarchy is a general phenomenon in social groups. As an essential element of executive and cognitive function, working memory could influence and be influenced by social status in a dominance hierarchy. However, the direction and degree of the association between them through different developmental stages remain unclear. To address this issue and clarify the "cause or consequence" problem, we investigated the spatial working memory performance in a Y-maze and Morris water maze in home-caged sibling Wistar rats (N = 26 cages, three rats/cage) through three stages of their life: before (week 7), during (week 10), and after (week 20) assumed timings of the social dominance hierarchy formation (SDHF). We used the social dominance tube test during the assumed time of hierarchy formation (weeks 9-11) to measure the relative dominance status in each cage. Here, we found that higher working memory index before SDHF could be predictive of later acquisition of higher social status. Working memory performance declined for all animals during SDHF, in which agonistic conflicts are increased. However, living within an established hierarchical social network for several weeks deteriorated the working memory performance of dominant and middle-ranked animals, while the performance of subordinates improved and got significantly better than higher-ranked animals. In conclusion, while working memory and social status were correlated positively before dominance hierarchy formation, there was a trade-off between them after the formation of it. In contrast to the common view, these results highlight the adverse effect of higher social status on cognitive behavior.
Collapse
|
8
|
Shao S, Yao D, Li S, Li J, Si Y, Zhang H, Zhu Z, Song D, Li H. N-Cadherin Regulates GluA1-Mediated Depressive-Like Behavior in Adolescent Female Rat Offspring following Prenatal Stress. Neuroendocrinology 2022; 112:493-509. [PMID: 34348318 DOI: 10.1159/000518383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/06/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND The incidence of depression is twice higher in women than in men, and gender differences in the prevalence rates first emerge around puberty. Prenatal stress (PS) induces gender-dependent depressive-like behavior in adolescent offspring, but the neuro-physiological mechanisms remain unclear. Our study aimed to investigate the possible neuro-physiological mechanisms of gender-dependent depressive-like behavior in PS adolescent offspring and further explored the possibility of treating depression in adolescent female rats. METHODS The pregnant rats were exposed to restraint stress in the third trimester for 7 days. The depressive-like behavior and the expression of N-cadherin and AMPARs in the hippocampus of adolescent offspring rats were assessed. 10 mg/kg AMPAR antagonist CNQX and 10 mg/kg N-cadherin antagonist ADH-1 were intraperitoneally injected into female adolescent offspring, respectively; 0.2 µg AMPAR agonist CX546 was administered to the dentate gyrus of male adolescent offspring to determine the role of N-cadherin-AMPARs in depressive-like behavior of the offspring following PS. RESULTS We found that PS increased N-cadherin expression, which upregulated GluA1 expression in the dentate gyrus, mediating depressive-like behavior in adolescent female rat offspring by reducing PSD-95. In addition, ADH-1 and CNQX improved depressive-like behavior in adolescent female offspring following PS. Furthermore, injection of the CX546 into the dentate gyrus induced depressive-like behavior in PS male offspring. CONCLUSION The gender-dependent expression of N-cadherin-GluA1 pathway in adolescent offspring in the dentate gyrus was the key factor in gender differences of depressive-like behavior following PS.
Collapse
Affiliation(s)
- Shuya Shao
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dan Yao
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Senya Li
- The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Li
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yufang Si
- Key Laboratory of Resource Biology and Biotechnology in Western China, Maternal and Infant Health Research Institute and Medical College, Northwestern University, Xi'an, China
| | - Huiping Zhang
- The Affiliated Children Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhongliang Zhu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Maternal and Infant Health Research Institute and Medical College, Northwestern University, Xi'an, China
| | - Dongli Song
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, California, USA
| | - Hui Li
- Department of Neonatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
9
|
Murlanova K, Begmatova D, Weber-Stadlbauer U, Meyer U, Pletnikov M, Pinhasov A. Double trouble: Prenatal immune activation in stress sensitive offspring. Brain Behav Immun 2022; 99:3-8. [PMID: 34547401 DOI: 10.1016/j.bbi.2021.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 12/15/2022] Open
Abstract
Viral infections during pregnancy are associated with increased incidence of psychiatric disorders in offspring. The pathological outcomes of viral infection appear to be caused by the deleterious effects of innate immune response-associated factors on development of the fetus, which predispose the offspring to pathological conditions in adulthood. The negative impact of viral infections varies substantially between pregnancies. Here, we explored whether differential stress sensitivity underlies the high heterogeneity of immune reactivity and whether this may influence the pathological consequences of maternal immune activation. Using mouse models of social dominance (Dom) and submissiveness (Sub), which possess innate features of stress resilience and vulnerability, respectively, we identified differential immune reactivity to the synthetic analogue of viral double-stranded RNA, Poly(I:C), in Sub and Dom nulliparous and pregnant females. More specifically, we found that Sub females showed an exacerbated pro- and anti-inflammatory cytokine response to Poly(I:C) as compared with Dom females. Sub offspring born to Sub mothers (stress sensitive offspring) showed enhanced locomotory response to the non-competitive NMDA antagonist, MK-801, which was potentiated by prenatal Poly(I:C) exposure. Our findings suggest that inherited stress sensitivity may lead to functional changes in glutamatergic signaling, which in turn is further exacerbated by prenatal exposure to viral-like infection. The maternal immunome seems to play a crucial role in these observed phenomena.
Collapse
Affiliation(s)
- Kateryna Murlanova
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel, Israel; Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Dilorom Begmatova
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Zurich, Switzerland
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Zurich, Switzerland
| | - Mikhail Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Albert Pinhasov
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel, Israel.
| |
Collapse
|
10
|
Multidimensional nature of dominant behavior: Insights from behavioral neuroscience. Neurosci Biobehav Rev 2021; 132:603-620. [PMID: 34902440 DOI: 10.1016/j.neubiorev.2021.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/29/2021] [Accepted: 12/09/2021] [Indexed: 12/17/2022]
Abstract
Social interactions for many species of animals are critical for survival, wellbeing, and reproduction. Optimal navigation of a social system increases chances for survival and reproduction, therefore there is strong incentive to fit into social structures. Social animals rely heavily on dominant-submissive behaviors in establishment of stable social hierarchies. There is a link between extreme manifestation of dominance/submissiveness and behavioral deviations. To understand neural substrates affiliated with a specific hierarchical rank, there is a real need for reliable animal behavioral models. Different paradigms have been consolidated over time to study the neurobiology of social rank behavior in a standardized manner using rodent models to unravel the neural pathways and substrates involved in normal and abnormal intraspecific social interactions. This review summarizes and discusses the commonly used behavioral tests and new directions for the assessment of dominance in rodents. We discuss the hierarchy inheritable nature and other critical issues regarding hierarchical rank manifestation which may help in designing social-rank-related studies that serve as promising pre-clinical tools in behavioral psychiatry.
Collapse
|
11
|
Anti-Cancer Effects of Cyclic Peptide ALOS4 in a Human Melanoma Mouse Model. Int J Mol Sci 2021; 22:ijms22179579. [PMID: 34502483 PMCID: PMC8430629 DOI: 10.3390/ijms22179579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022] Open
Abstract
We examined the effects of ALOS4, a cyclic peptide discovered previously by phage library selection against integrin αvβ3, on a human melanoma (A375) xenograft model to determine its abilities as a potential anti-cancer agent. We found that ALOS4 promoted healthy weight gain in A375-engrafted nude mice and reduced melanoma tumor mass and volume. Despite these positive changes, examination of the tumor tissue did not indicate any significant effects on proliferation, mitotic index, tissue vascularization, or reduction of αSMA or Ki-67 tumor markers. Modulation in overall expression of critical downstream αvβ3 integrin factors, such as FAK and Src, as well as reductions in gene expression of c-Fos and c-Jun transcription factors, indirectly confirmed our suspicions that ALOS4 is likely acting through an integrin-mediated pathway. Further, we found no overt formulation issues with ALOS4 regarding interaction with standard inert laboratory materials (polypropylene, borosilicate glass) or with pH and temperature stability under prolonged storage. Collectively, ALOS4 appears to be safe, chemically stable, and produces anti-cancer effects in a human xenograft model of melanoma. We believe these results suggest a role for ALOS4 in an integrin-mediated pathway in exerting its anti-cancer effects possibly through immune response modulation.
Collapse
|
12
|
Agranyoni O, Meninger-Mordechay S, Uzan A, Ziv O, Salmon-Divon M, Rodin D, Raz O, Koman I, Koren O, Pinhasov A, Navon-Venezia S. Gut microbiota determines the social behavior of mice and induces metabolic and inflammatory changes in their adipose tissue. NPJ Biofilms Microbiomes 2021; 7:28. [PMID: 33741982 PMCID: PMC7979825 DOI: 10.1038/s41522-021-00193-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
The link between the gut microbiota and social behavior has been demonstrated, however the translational impact of a certain microbiota composition on stable behavioral patterns is yet to be elucidated. Here we employed an established social behavior mouse model of dominance (Dom) or submissiveness (Sub). A comprehensive 16S rRNA gene sequence analysis of Dom and Sub mice revealed a significantly different gut microbiota composition that clearly distinguishes between the two behavioral modes. Sub mice gut microbiota is significantly less diverse than that of Dom mice, and their taxa composition uniquely comprised the genera Mycoplasma and Anaeroplasma of the Tenericutes phylum, in addition to the Rikenellaceae and Clostridiaceae families. Conversely, the gut microbiota of Dom mice includes the genus Prevotella of the Bacteriodetes phylum, significantly less abundant in Sub mice. In addition, Sub mice show lower body weight from the age of 2 weeks and throughout their life span, accompanied with lower epididymis white adipose tissue (eWAT) mass and smaller adipocytes together with substantially elevated expression of inflammation and metabolic-related eWAT adipokines. Finally, fecal microbiota transplantation into germ-free mice show that Sub-transplanted mice acquired Sub microbiota and adopted their behavioral and physiological features, including depressive-like and anti-social behaviors alongside reduced eWAT mass, smaller adipocytes, and a Sub-like eWAT adipokine profile. Our findings demonstrate the critical role of the gut microbiome in determining dominance vs. submissiveness and suggest an association between gut microbiota, the eWAT metabolic and inflammatory profile, and the social behavior mode.
Collapse
Affiliation(s)
- Oryan Agranyoni
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel
| | | | - Atara Uzan
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Oren Ziv
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Mali Salmon-Divon
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel
- The Dr. Miriam and Sheldon G. Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Dmitry Rodin
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel
| | - Olga Raz
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel
| | - Igor Koman
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Albert Pinhasov
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel.
- The Dr. Miriam and Sheldon G. Adelson School of Medicine, Ariel University, Ariel, Israel.
| | - Shiri Navon-Venezia
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel.
- The Dr. Miriam and Sheldon G. Adelson School of Medicine, Ariel University, Ariel, Israel.
| |
Collapse
|
13
|
Murlanova K, Michaelevski I, Kreinin A, Terrillion C, Pletnikov M, Pinhasov A. Link between temperament traits, brain neurochemistry and response to SSRI: insights from animal model of social behavior. J Affect Disord 2021; 282:1055-1066. [PMID: 33601678 DOI: 10.1016/j.jad.2020.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/14/2020] [Accepted: 11/01/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dominant-submissive relationships depend upon functionality of the neural circuits involving monoaminergic neurotransmission. Behavioral profiles of selectively bred dominant (Dom) and submissive (Sub) mice have been proposed to mimic hyperthymic- or depressive-like temperaments observed in patients with affective disorders. These mice differentially respond to psychotropic agents and stressful stimuli, however, the mechanisms underlying these differences remain unclear. To address these mechanisms, we analyzed the brain monoamine content and responses to paroxetine (PXT) in Dom and Sub mice. METHODS The behavioral effects of PXT (3 mg/kg, single injection) were assessed with the Elevated Plus Maze (EPM) and Forced Swim Test (FST). Monoamine tissue content was analyzed by HPLC-ECD. RESULTS Compared to Dom, Sub mice had decreased levels of serotonin (5-HT) in the brainstem (BS), reduced levels of norepinephrine (NE) in the prefrontal cortex (PFC), hippocampus (HPC), and striatum (STR) and elevated levels of dopamine (DA) in PFC, HPC, STR and BS. In EPM, PXT administration increased locomotion and exploration in Dom mice, with no effect in Sub mice. In FST, PXT disrupted immobility in Dom mice only. The PXT-produced differences in regional monoamine content were strain-dependent and consistent with the behavioral alterations. LIMITATIONS Chronic PXT treatment, in vivo monoamine assays and sex-dependent analysis were out of the scope of this study and will be performed in the future in order to provide an in-depth evaluation of the neurochemical mechanisms underlying temperament-dependent responses to SSRIs. CONCLUSIONS Our findings suggest neurochemical mechanisms that underlie temperament-based response to antidepressant treatment.
Collapse
Affiliation(s)
- Kateryna Murlanova
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; Department of Psychiatry and Behavioral Sciences and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; Department of Physiology and Biophysics, The Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
| | - Izhak Michaelevski
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; Adelson School of Medicine, Ariel University, Ariel 4070000, Israel
| | - Anatoly Kreinin
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
| | - Chantelle Terrillion
- Department of Psychiatry and Behavioral Sciences and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Mikhail Pletnikov
- Department of Psychiatry and Behavioral Sciences and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; Department of Physiology and Biophysics, The Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
| | - Albert Pinhasov
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel; Adelson School of Medicine, Ariel University, Ariel 4070000, Israel.
| |
Collapse
|
14
|
Yoshino Y, Roy B, Kumar N, Shahid Mukhtar M, Dwivedi Y. Molecular pathology associated with altered synaptic transcriptome in the dorsolateral prefrontal cortex of depressed subjects. Transl Psychiatry 2021; 11:73. [PMID: 33483466 PMCID: PMC7822869 DOI: 10.1038/s41398-020-01159-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023] Open
Abstract
Disrupted synaptic plasticity is the hallmark of major depressive disorder (MDD), with accompanying changes at the molecular and cellular levels. Often, the maladaptive molecular changes at the synapse are the result of global transcriptional reprogramming dictated by activity-dependent synaptic modulation. Thus far, no study has directly studied the transcriptome-wide expression changes locally at the synapse in MDD brain. Here, we have examined altered synaptic transcriptomics and their functional relevance in MDD with a focus on the dorsolateral prefrontal cortex (dlPFC). RNA was isolated from total fraction and purified synaptosomes of dlPFC from well-matched 15 non-psychiatric controls and 15 MDD subjects. Transcriptomic changes in synaptic and total fractions were detected by next-generation RNA-sequencing (NGS) and analyzed independently. The ratio of synaptic/total fraction was estimated to evaluate a shift in gene expression ratio in MDD subjects. Bioinformatics and network analyses were used to determine the biological relevance of transcriptomic changes in both total and synaptic fractions based on gene-gene network, gene ontology (GO), and pathway prediction algorithms. A total of 14,005 genes were detected in total fraction. A total of 104 genes were differentially regulated (73 upregulated and 31 downregulated) in MDD group based on 1.3-fold change threshold and p < 0.05 criteria. In synaptosomes, out of 13,236 detectable genes, 234 were upregulated and 60 were downregulated (>1.3-fold, p < 0.05). Several of these altered genes were validated independently by a quantitative polymerase chain reaction (qPCR). GO revealed an association with immune system processes and cell death. Moreover, a cluster of genes belonged to the nervous system development, and psychological disorders were discovered using gene-gene network analysis. The ratio of synaptic/total fraction showed a shift in expression of 119 genes in MDD subjects, which were primarily associated with neuroinflammation, interleukin signaling, and cell death. Our results suggest not only large-scale gene expression changes in synaptosomes, but also a shift in the expression of genes from total to synaptic fractions of dlPFC of MDD subjects with their potential role in immunomodulation and cell death. Our findings provide new insights into the understanding of transcriptomic regulation at the synapse and their possible role in MDD pathogenesis.
Collapse
Affiliation(s)
- Yuta Yoshino
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Bhaskar Roy
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Nilesh Kumar
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - M Shahid Mukhtar
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
15
|
Becker M, Pinhasov A, Ornoy A. Animal Models of Depression: What Can They Teach Us about the Human Disease? Diagnostics (Basel) 2021; 11:123. [PMID: 33466814 PMCID: PMC7830961 DOI: 10.3390/diagnostics11010123] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/28/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Depression is apparently the most common psychiatric disease among the mood disorders affecting about 10% of the adult population. The etiology and pathogenesis of depression are still poorly understood. Hence, as for most human diseases, animal models can help us understand the pathogenesis of depression and, more importantly, may facilitate the search for therapy. In this review we first describe the more common tests used for the evaluation of depressive-like symptoms in rodents. Then we describe different models of depression and discuss their strengths and weaknesses. These models can be divided into several categories: genetic models, models induced by mental acute and chronic stressful situations caused by environmental manipulations (i.e., learned helplessness in rats/mice), models induced by changes in brain neuro-transmitters or by specific brain injuries and models induced by pharmacological tools. In spite of the fact that none of the models completely resembles human depression, most animal models are relevant since they mimic many of the features observed in the human situation and may serve as a powerful tool for the study of the etiology, pathogenesis and treatment of depression, especially since only few patients respond to acute treatment. Relevance increases by the fact that human depression also has different facets and many possible etiologies and therapies.
Collapse
Affiliation(s)
- Maria Becker
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | - Albert Pinhasov
- Department of Molecular Biology and Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | - Asher Ornoy
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel
| |
Collapse
|
16
|
Dhuriya YK, Sharma D. Neuronal Plasticity: Neuronal Organization is Associated with Neurological Disorders. J Mol Neurosci 2020; 70:1684-1701. [PMID: 32504405 DOI: 10.1007/s12031-020-01555-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/13/2020] [Indexed: 12/18/2022]
Abstract
Stimuli from stressful events, attention in the classroom, and many other experiences affect the functionality of the brain by changing the structure or reorganizing the connections between neurons and their communication. Modification of the synaptic transmission is a vital mechanism for generating neural activity via internal or external stimuli. Neuronal plasticity is an important driving force in neuroscience research, as it is the basic process underlying learning and memory and is involved in many other functions including brain development and homeostasis, sensorial training, and recovery from brain injury. Indeed, neuronal plasticity has been explored in numerous studies, but it is still not clear how neuronal plasticity affects the physiology and morphology of the brain. Thus, unraveling the molecular mechanisms of neuronal plasticity is essential for understanding the operation of brain functions. In this timeline review, we discuss the molecular mechanisms underlying different forms of synaptic plasticity and their association with neurodegenerative/neurological disorders as a consequence of alterations in neuronal plasticity.
Collapse
Affiliation(s)
- Yogesh Kumar Dhuriya
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR) Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, India
| | - Divakar Sharma
- Department of Biochemistry, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India. .,CRF, Mass Spectrometry Laboratory, Kusuma School of Biological Sciences (KSBS), Indian Institute of Technology-Delhi (IIT-D), Delhi, 110016, India.
| |
Collapse
|
17
|
Link between personality and response to THC exposure. Behav Brain Res 2019; 379:112361. [PMID: 31734264 DOI: 10.1016/j.bbr.2019.112361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/29/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
The effects of cannabis reported by users range from experiences of euphoria and anxiolytic effects to paranoia, anxiety, and increased risk of depression. Attempts to reconcile the apparent contradictions in user response have not been conclusive. Here, we utilized selectively-bred stress-resilient socially dominant (Dom) and stress-sensitive socially submissive (Sub) mice to elucidate this contradiction. Following short-term, repeated treatment with delta-9-tetrahydrocannabinol (THC) at two different doses (1.5 mg/kg and 15 mg/kg), Sub mice presented significant place-aversion in a Conditioned Place Preference paradigm at a high dose, whereas Dom mice displayed no place preference or aversion. Forced Swim test conducted after 6-week of washout period, revealed differential impact of the two THC doses depending upon behavioral pattern. Specifically, the low dose alleviated depressive-like behavior in Sub mice, while the high dose produced the opposite effect in Dom mice. Interestingly, corticosterone concentration in serum was elevated at the high dose regardless of the mice-population tested. We conclude here that differences in dominance behavior and stress vulnerability are involved in the regulation of cannabis response among users and should be considered when prescribing THC-containing medications to patients.
Collapse
|
18
|
Social defeat-induced Cingulate gyrus immediate-early gene expression and anxiolytic-like effect depend upon social rank. Brain Res Bull 2018; 143:97-105. [PMID: 30343051 DOI: 10.1016/j.brainresbull.2018.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 12/22/2022]
Abstract
Social hierarchy is considered to impart an adaptive advantage to the species by reducing long-term conflict between conspecifics. While social stratification is frequently established via stress-inducing stimuli, the subsequent integration of individuals into the hierarchy may attenuate anxiety. Presently, we hypothesized that repeated reinforcement of murine social hierarchy in the dominant-submissive relationship (DSR) food-competition test would engender divergent neuroplastic changes mediating both social and anxiety-like behavior among selectively-bred Dominant (Dom) and Submissive (Sub) mice. Two weeks of repeated respective social victory or defeat reduced serum corticosterone levels of both Dom and Sub mice, whereas socially-defeated Sub mice demonstrated markedly greater exploration of the open arms of the elevated plus maze (EPM). At the same time, social victory led to markedly greater expression of the immediate-early genes (IEGs) c-Jun and EGR-1 in the lateral septal nucleus (LSN) among Dom mice, in contrast with defeated Sub counterparts which demonstrated four-fold greater IEG expression in the cingulate gyrus (Cg). These findings point towards involvement of the Cg in the anxiety-like effect among Sub mice after repeated social defeat, and suggest stabilization of the social hierarchy to attenuate the stress-inducing nature of social interaction, particularly for subordinates. Further study of the potentially anxiolytic-like effects of Cg activity should shed light upon the functional significance of the Cg in social interaction, social hierarchical sorting and anxiety.
Collapse
|
19
|
Social dominance predicts hippocampal glucocorticoid receptor recruitment and resilience to prenatal adversity. Sci Rep 2018; 8:9595. [PMID: 29941995 PMCID: PMC6018627 DOI: 10.1038/s41598-018-27988-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/12/2018] [Indexed: 12/19/2022] Open
Abstract
The developing fetus is highly sensitive to prenatal stress, which may alter Hypothalamic-Pituitary-Adrenal (HPA) axis programming and increase the risk of behavioral disorders. There is high variability among the human population, wherein many offspring of stressed pregnancies display resilience to adversity, while the remainder displays vulnerability. In order to identify biological substrates mediating between resilience or vulnerability to prenatal adversity, we exposed stress-resistant Dominant (Dom) and stress-sensitive Submissive (Sub) mice to mild prenatal restraint stress (PRS, 45 min on gestational days (GD) 15, 16 and 17). We hypothesized that PRS would differentially alter prenatal programming of limbic regions regulating the HPA axis and affect among Dom and Sub offspring. Indeed, PRS increased Sub offspring’s serum corticosterone, and exaggerated their anxiety- and depressive-like behavior, while Dom offspring remained resilient to the hormonal and behavioral consequences of PRS. Moreover, PRS exposure markedly facilitated glucocorticoid receptor (GR) recruitment to the hippocampus among Dom mice in response to restraint stress, which may be responsible for their resilience to stressful challenge. These findings suggest proclivity to adaptive or maladaptive prenatal programming of hippocampal GR recruitment to be inheritable and predictable by social dominance or submissiveness.
Collapse
|
20
|
Borovok N, Nesher E, Reichenstein M, Tikhonova T, Levin Y, Pinhasov A, Michaelevski I. Effect of social interactions on hippocampal protein expression in animal dominant and submissive model of behavioral disorders. Proteomics Clin Appl 2017; 11. [DOI: 10.1002/prca.201700089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 06/07/2017] [Accepted: 06/26/2017] [Indexed: 02/02/2023]
Affiliation(s)
- Natalia Borovok
- Department of Biochemistry and Molecular Biology; Tel Aviv University; Tel-Aviv Israel
| | | | - Michal Reichenstein
- Department of Biochemistry and Molecular Biology; Tel Aviv University; Tel-Aviv Israel
| | | | - Yishai Levin
- de Botton Institute for Protein Profiling; The Nancy & Stephen Grand Israel National Center for Personalized Medicine; Weizmann Institute of Science; Rehovot Israel
| | - Albert Pinhasov
- Department of Molecular Biology; Ariel University; Ariel Israel
| | - Izhak Michaelevski
- Department of Molecular Biology; Ariel University; Ariel Israel
- Department of Biochemistry and Molecular Biology; Tel Aviv University; Tel-Aviv Israel
- Sagol School of Neuroscience; Tel Aviv University; Tel Aviv Israel
| |
Collapse
|
21
|
Luan X, Tao J, Zhang J, Xie Y, Zhang X, Su H, He J. Increased BDNF may not be associated with cognitive impairment in heroin-dependent patients. Medicine (Baltimore) 2017; 96:e6582. [PMID: 28403087 PMCID: PMC5403084 DOI: 10.1097/md.0000000000006582] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A growing number of evidence suggests that brain-derived neurotrophic factor (BDNF) plays an important part in modulating the activities on the basis of hippocampus neural plasticity, such as learning and memory. Heroin addiction has a series of cognitive impairments that may be associated with BDNF. In this study, we explored the association of BDNF with cognitive function in heroin-dependent patients.We enrolled 86 heroin-dependent patients and 238 normal control subjects and examined their cognition by the repeatable battery for the assessment of neuropsychological status (RBANS) and serum BDNF levels in 2 groups.BDNF levels were significantly higher in patients than controls (P < .001). Cognitive scores of the RBANS showed that attention and language index (P < .05) were significantly lower in heroin-dependent patients than control groups. Unfortunately, we found no positive association between BDNF and cognitive function in patients, except that BDNF was positively associated with visuospatial/constructional index in control groups.Our findings suggest that BDNF may not be involved in the pathophysiology of heroin dependence, but more studies about cognitive impairment in heroin addiction are needed.
Collapse
Affiliation(s)
- Xiaoqian Luan
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou
| | - Jingyan Tao
- Department of Rehabilitation Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou
| | - Jie Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai
| | - Ying Xie
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou
| | - Xiangyang Zhang
- Beijing HuiLongGuan Hospital, Peking University, Beijing, PR China
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Hang Su
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai
| | - Jincai He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou
| |
Collapse
|
22
|
The Role of Neural Plasticity in Depression: From Hippocampus to Prefrontal Cortex. Neural Plast 2017; 2017:6871089. [PMID: 28246558 PMCID: PMC5299163 DOI: 10.1155/2017/6871089] [Citation(s) in RCA: 357] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/04/2017] [Indexed: 12/19/2022] Open
Abstract
Neural plasticity, a fundamental mechanism of neuronal adaptation, is disrupted in depression. The changes in neural plasticity induced by stress and other negative stimuli play a significant role in the onset and development of depression. Antidepressant treatments have also been found to exert their antidepressant effects through regulatory effects on neural plasticity. However, the detailed mechanisms of neural plasticity in depression still remain unclear. Therefore, in this review, we summarize the recent literature to elaborate the possible mechanistic role of neural plasticity in depression. Taken together, these findings may pave the way for future progress in neural plasticity studies.
Collapse
|
23
|
Kandimalla R, Thirumala V, Reddy PH. Is Alzheimer's disease a Type 3 Diabetes? A critical appraisal. Biochim Biophys Acta Mol Basis Dis 2016; 1863:1078-1089. [PMID: 27567931 DOI: 10.1016/j.bbadis.2016.08.018] [Citation(s) in RCA: 371] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/07/2016] [Accepted: 08/17/2016] [Indexed: 12/20/2022]
Abstract
Recently researchers proposed the term 'Type-3-Diabetes' for Alzheimer's disease (ad) because of the shared molecular and cellular features among Type-1-Diabetes, Type-2-Diabetes and insulin resistance associated with memory deficits and cognitive decline in elderly individuals. Recent clinical and basic studies on patients with diabetes and AD revealed previously unreported cellular and pathological among diabetes, insulin resistance and AD. These studies are also strengthened by various basic biological studies that decipher the effects of insulin in the pathology of AD through cellular and molecular mechanisms. For instance, insulin is involved in the activation of glycogen synthase kinase 3β, which in turn causes phosphorylation of tau, which involved in the formation of neurofibrillary tangles. Interestingly, insulin also plays a crucial role in the formation amyloid plaques. In this review, we discussed significant shared mechanisms between AD and diabetes and we also provided therapeutic avenues for diabetes and AD. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.
Collapse
Affiliation(s)
- Ramesh Kandimalla
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States.
| | - Vani Thirumala
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; BSA Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Departments of Cell Biology & Biochemistry, Neuroscience & Pharmacology and Neurology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States
| |
Collapse
|
24
|
Nicotine Significantly Improves Chronic Stress-Induced Impairments of Cognition and Synaptic Plasticity in Mice. Mol Neurobiol 2016; 54:4644-4658. [DOI: 10.1007/s12035-016-0012-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/03/2016] [Indexed: 12/30/2022]
|
25
|
Borovok N, Nesher E, Levin Y, Reichenstein M, Pinhasov A, Michaelevski I. Dynamics of Hippocampal Protein Expression During Long-term Spatial Memory Formation. Mol Cell Proteomics 2015; 15:523-41. [PMID: 26598641 DOI: 10.1074/mcp.m115.051318] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Indexed: 01/08/2023] Open
Abstract
Spatial memory depends on the hippocampus, which is particularly vulnerable to aging. This vulnerability has implications for the impairment of navigation capacities in older people, who may show a marked drop in performance of spatial tasks with advancing age. Contemporary understanding of long-term memory formation relies on molecular mechanisms underlying long-term synaptic plasticity. With memory acquisition, activity-dependent changes occurring in synapses initiate multiple signal transduction pathways enhancing protein turnover. This enhancement facilitates de novo synthesis of plasticity related proteins, crucial factors for establishing persistent long-term synaptic plasticity and forming memory engrams. Extensive studies have been performed to elucidate molecular mechanisms of memory traces formation; however, the identity of plasticity related proteins is still evasive. In this study, we investigated protein turnover in mouse hippocampus during long-term spatial memory formation using the reference memory version of radial arm maze (RAM) paradigm. We identified 1592 proteins, which exhibited a complex picture of expression changes during spatial memory formation. Variable linear decomposition reduced significantly data dimensionality and enriched three principal factors responsible for variance of memory-related protein levels at (1) the initial phase of memory acquisition (165 proteins), (2) during the steep learning improvement (148 proteins), and (3) the final phase of the learning curve (123 proteins). Gene ontology and signaling pathways analysis revealed a clear correlation between memory improvement and learning phase-curbed expression profiles of proteins belonging to specific functional categories. We found differential enrichment of (1) neurotrophic factors signaling pathways, proteins regulating synaptic transmission, and actin microfilament during the first day of the learning curve; (2) transcription and translation machinery, protein trafficking, enhancement of metabolic activity, and Wnt signaling pathway during the steep phase of memory formation; and (3) cytoskeleton organization proteins. Taken together, this study clearly demonstrates dynamic assembly and disassembly of protein-protein interaction networks depending on the stage of memory formation engrams.
Collapse
Affiliation(s)
- Natalia Borovok
- From the ‡Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Elimelech Nesher
- §Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
| | - Yishai Levin
- ¶de Botton Institute for Protein Profiling, The Nancy & Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michal Reichenstein
- From the ‡Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Albert Pinhasov
- §Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
| | - Izhak Michaelevski
- From the ‡Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel-Aviv 6997801, Israel; ‖Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
26
|
Nesher E, Koman I, Gross M, Tikhonov T, Bairachnaya M, Salmon-Divon M, Levin Y, Gerlitz G, Michaelevski I, Yadid G, Pinhasov A. Synapsin IIb as a functional marker of submissive behavior. Sci Rep 2015; 5:10287. [PMID: 25998951 PMCID: PMC4441117 DOI: 10.1038/srep10287] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 04/09/2015] [Indexed: 12/22/2022] Open
Abstract
Dominance and submissiveness are important functional elements of the social hierarchy. By employing selective breeding based on a social interaction test, we developed mice with strong and stable, inheritable features of dominance and submissiveness. In order to identify candidate genes responsible for dominant and submissive behavior, we applied transcriptomic and proteomic studies supported by molecular, behavioral and pharmacological approaches. We clearly show here that the expression of Synapsin II isoform b (Syn IIb) is constitutively upregulated in the hippocampus and striatum of submissive mice in comparison to their dominant and wild type counterparts. Moreover, the reduction of submissive behavior achieved after mating and delivery was accompanied by a marked reduction of Syn IIb expression. Since submissiveness has been shown to be associated with depressive-like behavior, we applied acute SSRI (Paroxetine) treatment to reduce submissiveness in studied mice. We found that reduction of submissive behavior evoked by Paroxetine was paired with significantly decreased Syn IIb expression. In conclusion, our findings indicate that submissiveness, known to be an important element of depressive-like behavioral abnormalities, is strongly linked with changes in Syn IIb expression.
Collapse
Affiliation(s)
- Elimelech Nesher
- 1] Department of Molecular Biology. Ariel University, Ariel. Israel [2] Faculty of Life Sciences. Bar-Ilan University, Ramat Gan. Israel
| | - Igor Koman
- Department of Molecular Biology. Ariel University, Ariel. Israel
| | - Moshe Gross
- Department of Molecular Biology. Ariel University, Ariel. Israel
| | - Tatiana Tikhonov
- Department of Molecular Biology. Ariel University, Ariel. Israel
| | | | | | - Yishai Levin
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine. Weizmann Institute of Science, Rehovot. Israel
| | - Gabi Gerlitz
- Department of Molecular Biology. Ariel University, Ariel. Israel
| | - Izhak Michaelevski
- 1] Department of Biochemistry and Molecular Biology. Tel-Aviv University, Tel-Aviv. Israel [2] Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv. Israel
| | - Gal Yadid
- Faculty of Life Sciences. Bar-Ilan University, Ramat Gan. Israel
| | - Albert Pinhasov
- Department of Molecular Biology. Ariel University, Ariel. Israel
| |
Collapse
|