1
|
Tanabe M, Saito Y, Takasaki A, Nakano K, Yamamoto S, Suzuki C, Kawamura N, Hattori A, Oikawa M, Nagashima S, Yanagi S, Yamaguchi T, Fukuda T. Role of immature choroid plexus in the pathology of model mice and human iPSC-derived organoids with autism spectrum disorder. Cell Rep 2024; 44:115133. [PMID: 39731733 DOI: 10.1016/j.celrep.2024.115133] [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: 11/20/2023] [Revised: 10/22/2024] [Accepted: 12/11/2024] [Indexed: 12/30/2024] Open
Abstract
During gestation, the choroid plexus (ChP) produces protein-rich cerebrospinal fluid and matures prior to brain development. It is assumed that ChP dysfunction has a profound effect on developmental neuropsychiatric disorders, such as autism spectrum disorder (ASD). However, the mechanisms linking immature ChP to the onset of ASD remain unclear. Here, we find that ChP-specific CAMDI-knockout mice develop an immature ChP alongside decreased multiciliogenesis and expression of differentiation marker genes following disruption of the cerebrospinal fluid barrier. These mice exhibit ASD-like behaviors, including anxiety and impaired socialization. Additionally, the administration of metformin, an FDA-approved drug, before the social critical period achieves ChP maturation and restores social behaviors. Furthermore, both the ASD model mice and organoids derived from patients with ASD developed an immature ChP. These results propose the involvement of an immature ChP in the pathogenesis of ASD and suggest the targeting of functional maturation of the ChP as a therapeutic strategy for ASD.
Collapse
Affiliation(s)
- Motoi Tanabe
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Yuga Saito
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Ayaka Takasaki
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Keita Nakano
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Shunta Yamamoto
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Chikako Suzuki
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Nao Kawamura
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Aki Hattori
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Mami Oikawa
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Shun Nagashima
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Shigeru Yanagi
- Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Tomoyuki Yamaguchi
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Toshifumi Fukuda
- Laboratory of Regenerative Medicine, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan.
| |
Collapse
|
2
|
Wang J, Bai L, Ye Y, Chen X, Hu X, Peng Y. Sex differences in mortality risk and U-shaped relationship with klotho levels: A long-term cohort study. Exp Gerontol 2024; 198:112643. [PMID: 39613274 DOI: 10.1016/j.exger.2024.112643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUNDS It remains unclear whether there are sex differences in the correlation between klotho and mortality risk. The purpose of our study is to investigate the relationship between klotho levels and all-cause mortality, specifically examining potential sex disparities. METHODS The study utilized data from the National Health and Nutrition Examination Survey. The study employed the Cox proportional hazards model and Kaplan-Meier survival curves to conduct multivariate analyses investigating the associations between klotho and mortality. The impact of klotho level on mortality was assessed by a restricted cubic spline curve. RESULTS Our study included 13,748 participants. The hazard ratios (HR) for all-cause mortality were found to be higher in males compared to females. The risk of all-cause mortality decreased until the klotho level reached 959 pg/mL in all-cause mortality, then increased thereafter in females (P value for nonlinearity = 0.01, P overall = 0.02); no curved relationship was observed in males. CONCLUSION Klotho levels and mortality follow a U-shaped curve, with sex differences in the relationship. Males with higher klotho levels have a higher risk of all-cause death than females.
Collapse
Affiliation(s)
- Junwen Wang
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Street, Chengdu 610041, China
| | - Lin Bai
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Street, Chengdu 610041, China
| | - Yuyang Ye
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Street, Chengdu 610041, China
| | - Xuefeng Chen
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Street, Chengdu 610041, China
| | - Xinru Hu
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Street, Chengdu 610041, China
| | - Yong Peng
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Street, Chengdu 610041, China.
| |
Collapse
|
3
|
Sadanandan J, Sathyanesan M, Newton SS. Aging alters the expression of trophic factors and tight junction proteins in the mouse choroid plexus. Fluids Barriers CNS 2024; 21:77. [PMID: 39334352 PMCID: PMC11438291 DOI: 10.1186/s12987-024-00574-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The choroid plexus (CP) is an understudied tissue in the central nervous system and is primarily implicated in cerebrospinal fluid (CSF) production. CP also produces numerous neurotrophic factors (NTF) which circulate to different brain regions. Regulation of NTFs in the CP during natural aging is largely unknown. Here, we investigated the age and gender-specific transcription of NTFs along with the changes in the tight junctional proteins (TJPs) and the water channel protein Aquaporin (AQP1). METHODS Male and female mice were used for our study. Age-related transcriptional changes were analyzed using quantitative PCR at three different time points: mature adult, middle-aged, and aged. Transcriptional changes during aging were further confirmed with digital droplet PCR. Additionally, we used immunohistochemical analysis (IHC) for the evaluation of in vivo protein expression. We further investigated the cellular phenotype of these NTFS, TJP, and water channel proteins in the mouse CP by co-labeling them with the classical vascular marker, Isolectin B4, and epithelial cell marker, Plectin. RESULTS Aging significantly altered NTF gene expression in the CP. Brain-derived neurotrophic factor (BDNF), Midkine (MDK), VGF, Insulin-like growth factor (IGF1), IGF2, Klotho (KL), Erythropoietin (EPO), and its receptor (EPOR) were reduced in the aged CP of males and females. Vascular endothelial growth factor (VEGF) transcription was gender-specific; in males, gene expression was unchanged in the aged CP, while females showed an age-dependent reduction. Age-dependent changes in VEGF localization were evident, from vasculature to epithelial cells. IGF2 and klotho localized in the basolateral membrane of the CP and showed an age-dependent reduction in epithelial cells. Water channel protein AQP1 localized in the tip of epithelial cells and showed an age-related reduction in mRNA and protein levels. TJP's JAM, CLAUDIN1, CLAUDIN2 and CLAUDIN5 were reduced in aged mice. CONCLUSIONS Our study highlights transcriptional level changes in the CP during aging. The age-related transcriptional changes exhibit similarities as well as gene-specific differences in the CP of males and females. Altered transcription of the water channel protein AQP1 and TJPs could be involved in reduced CSF production during aging. Importantly, reduction in the neurotrophic factors and longevity factor Klotho can play a role in regulating brain aging.
Collapse
Affiliation(s)
- Jayanarayanan Sadanandan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Monica Sathyanesan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Samuel S Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.
| |
Collapse
|
4
|
Seitz-Holland J, Alemán-Gómez Y, Cho KIK, Pasternak O, Cleusix M, Jenni R, Baumann PS, Klauser P, Conus P, Hagmann P, Do KQ, Kubicki M, Dwir D. Matrix metalloproteinase 9 (MMP-9) activity, hippocampal extracellular free water, and cognitive deficits are associated with each other in early phase psychosis. Neuropsychopharmacology 2024; 49:1140-1150. [PMID: 38431757 PMCID: PMC11109110 DOI: 10.1038/s41386-024-01814-5] [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: 07/13/2023] [Revised: 12/18/2023] [Accepted: 01/29/2024] [Indexed: 03/05/2024]
Abstract
Increasing evidence points toward the role of the extracellular matrix, specifically matrix metalloproteinase 9 (MMP-9), in the pathophysiology of psychosis. MMP-9 is a critical regulator of the crosstalk between peripheral and central inflammation, extracellular matrix remodeling, hippocampal development, synaptic pruning, and neuroplasticity. Here, we aim to characterize the relationship between plasma MMP-9 activity, hippocampal microstructure, and cognition in healthy individuals and individuals with early phase psychosis. We collected clinical, blood, and structural and diffusion-weighted magnetic resonance imaging data from 39 individuals with early phase psychosis and 44 age and sex-matched healthy individuals. We measured MMP-9 plasma activity, hippocampal extracellular free water (FW) levels, and hippocampal volumes. We used regression analyses to compare MMP-9 activity, hippocampal FW, and volumes between groups. We then examined associations between MMP-9 activity, FW levels, hippocampal volumes, and cognitive performance assessed with the MATRICS battery. All analyses were controlled for age, sex, body mass index, cigarette smoking, and years of education. Individuals with early phase psychosis demonstrated higher MMP-9 activity (p < 0.0002), higher left (p < 0.05) and right (p < 0.05) hippocampal FW levels, and lower left (p < 0.05) and right (p < 0.05) hippocampal volume than healthy individuals. MMP-9 activity correlated positively with hippocampal FW levels (all participants and individuals with early phase psychosis) and negatively with hippocampal volumes (all participants and healthy individuals). Higher MMP-9 activity and higher hippocampal FW levels were associated with slower processing speed and worse working memory performance in all participants. Our findings show an association between MMP-9 activity and hippocampal microstructural alterations in psychosis and an association between MMP-9 activity and cognitive performance. Further, more extensive longitudinal studies should examine the therapeutic potential of MMP-9 modulators in psychosis.
Collapse
Affiliation(s)
- Johanna Seitz-Holland
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Yasser Alemán-Gómez
- Connectomics Lab, Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Medical Image Analysis Laboratory, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kang Ik K Cho
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martine Cleusix
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philipp S Baumann
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Paul Klauser
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philippe Conus
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Patric Hagmann
- Connectomics Lab, Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Marek Kubicki
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniella Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
5
|
Sadanandan J, Sathyanesan M, Newton SS. Regulation of trophic factors in the choroid plexus of aged mice. RESEARCH SQUARE 2024:rs.3.rs-4123786. [PMID: 38562722 PMCID: PMC10984084 DOI: 10.21203/rs.3.rs-4123786/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background The choroid plexus (CP) is an understudied tissue in the central nervous system (CNS), primarily implicated in cerebrospinal fluid (CSF) production. Additionally, CP produces numerous neurotrophic factors (NTF), which circulate to different regions of the brain. Regulation of NTF in the CP during natural aging has yet to be discovered. Here, we investigated the age and gender-specific transcription of NTFs along with the changes in the tight junctional proteins (TJPs) and water channel protein Aquaporin (AQP1). Methods We used male and female mice for our study. We analyzed neurotrophic factor gene expression patterns using quantitative and digital droplet PCR at three different time points: mature adult, middle-aged, and aged. Additionally, we used immunohistochemical analysis (IHC) to evaluate in vivo protein expression. We further investigated the cellular phenotype of these NTFS, TJP and water channel proteins in the mouse CP by co-labeling them with the classical vascular marker, Isolectin B4, and epithelial cell marker, plectin. Results Aging significantly altered the NTF's gene expression in the CP Brain-derived neurotrophic factor (BDNF), Midkine, VGF, Insulin-like growth factor (IGF1), IGF2, klotho, Erythropoietin, and its receptor were reduced in the aged CP of males and females. Vascular endothelial growth factor (VEGF) transcription was gender-specific; in males, gene expression is unchanged in the aged CP while females showed an age-dependent reduction. Age-dependent changes in VEGF localization were evident, from vasculature to epithelial cells. IGF2 and klotho localized in the basolateral membrane of the CP and showed an age-dependent reduction in epithelial cells. Water channel protein AQP1 localized in the tip of epithelial cells and showed an age-related reduction in mRNA and protein levels. TJP's JAM, CLAUDIN1, CLAUDIN2, and CLAUDIN5 were reduced in aged mice. Conclusions Our study highlights transcriptional level changes in the CP during aging. The age-related transcriptional changes exhibit similarities as well as gene-specific differences in the CP of males and females. Altered transcription of the water channel protein AQP1 and TJPs could be involved in reduced CSF production during aging. Importantly, reduction in the neurotrophic factors and longevity factor Klotho can play a role in regulating brain aging.
Collapse
|
6
|
Bannai D, Reuter M, Hegde R, Hoang D, Adhan I, Gandu S, Pong S, Raymond N, Zeng V, Chung Y, He G, Sun D, van Erp TGM, Addington J, Bearden CE, Cadenhead K, Cornblatt B, Mathalon DH, McGlashan T, Jeffries C, Stone W, Tsuang M, Walker E, Woods SW, Cannon TD, Perkins D, Keshavan M, Lizano P. Linking enlarged choroid plexus with plasma analyte and structural phenotypes in clinical high risk for psychosis: A multisite neuroimaging study. Brain Behav Immun 2024; 117:70-79. [PMID: 38169244 DOI: 10.1016/j.bbi.2023.12.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/04/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Choroid plexus (ChP) enlargement exists in first-episode and chronic psychosis, but whether enlargement occurs before psychosis onset is unknown. This study investigated whether ChP volume is enlarged in individuals with clinical high-risk (CHR) for psychosis and whether these changes are related to clinical, neuroanatomical, and plasma analytes. METHODS Clinical and neuroimaging data from the North American Prodrome Longitudinal Study 2 (NAPLS2) was used for analysis. 509 participants (169 controls, 340 CHR) were recruited. Conversion status was determined after 2-years of follow-up, with 36 psychosis converters. The lateral ventricle ChP was manually segmented from baseline scans. A subsample of 31 controls and 53 CHR had plasma analyte and neuroimaging data. RESULTS Compared to controls, CHR (d = 0.23, p = 0.017) and non-converters (d = 0.22, p = 0.03) demonstrated higher ChP volumes, but not in converters. In CHR, greater ChP volume correlated with lower cortical (r = -0.22, p < 0.001), subcortical gray matter (r = -0.21, p < 0.001), and total white matter volume (r = -0.28,p < 0.001), as well as larger lateral ventricle volume (r = 0.63,p < 0.001). Greater ChP volume correlated with makers functionally associated with the lateral ventricle ChP in CHR [CCL1 (r = -0.30, p = 0.035), ICAM1 (r = 0.33, p = 0.02)], converters [IL1β (r = 0.66, p = 0.004)], and non-converters [BMP6 (r = -0.96, p < 0.001), CALB1 (r = -0.98, p < 0.001), ICAM1 (r = 0.80, p = 0.003), SELE (r = 0.59, p = 0.026), SHBG (r = 0.99, p < 0.001), TNFRSF10C (r = 0.78, p = 0.001)]. CONCLUSIONS CHR and non-converters demonstrated significantly larger ChP volumes compared to controls. Enlarged ChP was associated with neuroanatomical alterations and analyte markers functionally associated with the ChP. These findings suggest that the ChP may be a key an important biomarker in CHR.
Collapse
Affiliation(s)
- Deepthi Bannai
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Martin Reuter
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA; Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Rachal Hegde
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Dung Hoang
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Iniya Adhan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Swetha Gandu
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Sovannarath Pong
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nick Raymond
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Victor Zeng
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yoonho Chung
- Department of Psychology, Yale University, New Haven, CT, USA
| | - George He
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Daqiang Sun
- Semel Institute for Neuroscience and Human Behavior and Department of Psychology, UCLA, Los Angeles, CA, USA
| | - Theo G M van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, UC Irvine, Irvine, CA, USA
| | - Jean Addington
- Hotchkins Brain Institute, Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Carrie E Bearden
- Semel Institute for Neuroscience and Human Behavior and Department of Psychology, UCLA, Los Angeles, CA, USA
| | | | | | | | | | - Clark Jeffries
- Renaissance Computing Institute, University of North Carolina, Chapel Hill, NC, USA
| | - William Stone
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Ming Tsuang
- Department of Psychiatry, UCSD, San Diego, CA, USA
| | - Elaine Walker
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Scott W Woods
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Tyrone D Cannon
- Department of Psychology, Yale University, New Haven, CT, USA; Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Diana Perkins
- Renaissance Computing Institute, University of North Carolina, Chapel Hill, NC, USA; Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Matcheri Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Paulo Lizano
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Division of Translational Neuroscience, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
7
|
Drapšin M, Dočkal T, Houdek P, Sládek M, Semenovykh K, Sumová A. Circadian clock in choroid plexus is resistant to immune challenge but dampens in response to chronodisruption. Brain Behav Immun 2024; 117:255-269. [PMID: 38280534 DOI: 10.1016/j.bbi.2024.01.217] [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: 11/16/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 01/29/2024] Open
Abstract
The choroid plexus (ChP) in the brain ventricles has a major influence on brain homeostasis. In this study, we aimed to determine whether the circadian clock located in ChP is affected by chronodisruption caused by misalignment with the external light/dark cycle and/or inflammation. Adult mPer2Luc mice were maintained in the LD12:12 cycle or exposed to one of two models of chronic chronodisruption - constant light for 22-25 weeks (cLL) or 6-hour phase advances of the LD12:12 cycle repeated weekly for 12 weeks (cLD-shifts). Locomotor activity was monitored before the 4th ventricle ChP and suprachiasmatic nuclei (SCN) explants were recorded in real time for PER2-driven population and single-cell bioluminescence rhythms. In addition, plasma immune marker concentrations and gene expression in ChP, prefrontal cortex, hippocampus and cerebellum were analyzed. cLL dampened the SCN clock but did not shorten the inactivity interval (sleep). cLD-shifts had no effect on the SCN clock, but transiently affected sleep duration and fragmentation. Both chronodisruption protocols dampened the ChP clock. Although immune markers were elevated in plasma and hippocampus, levels in ChP were unaffected, and unlike the liver clock, the ChP clock was resistant to lipopolysaccharide treatment. Importantly, both chronodisruption protocols reduced glucocorticoid signaling in ChP. The data demonstrate the high resistance of the ChP clock to inflammation, highlighting its role in protecting the brain from neuroinflammation, and on the other hand its high sensitivity to chronodisruption. Our results provide a novel link between human lifestyle-induced chronodisruption and the impairment of ChP-dependent brain homeostasis.
Collapse
Affiliation(s)
- Milica Drapšin
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tereza Dočkal
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Pavel Houdek
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Sládek
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Kateryna Semenovykh
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alena Sumová
- Laboratory of Biological Rhythms, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
8
|
Murck H, Fava M, Cusin C, Fatt CC, Trivedi M. Brain ventricle and choroid plexus morphology as predictor of treatment response in major depression: Findings from the EMBARC study. Brain Behav Immun Health 2024; 35:100717. [PMID: 38186634 PMCID: PMC10767278 DOI: 10.1016/j.bbih.2023.100717] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/09/2024] Open
Abstract
Recent observations suggest a role of the volume of the cerebral ventricle volume, corpus callosum (CC) segment volume, in particular that of the central-anterior part, and choroid plexus (CP) volume for treatment resistance of major depressive disorder (MDD). An increased CP volume has been associated with increased inflammatory activity and changes in the structure of the ventricles and corpus callosum. We attempt to replicate and confirm that these imaging markers are associated with clinical outcome in subjects from the EMBARC study, as implied by a recent pilot study. The EMBARC study is a placebo controlled randomized study comparing sertraline vs. placebo in patients with MDD to identify biological markers of therapy resistance. Association of baseline volumes of the lateral ventricles (LVV), choroid plexus volume (CPV) and volume of segments of the CC with treatment response after 4 weeks treatment was evaluated. 171 subjects (61 male, 110 female) completed the 4 week assessments; gender and age were taken into account for this analyses. As previously reported, no treatment effect of sertraline vs. placebo was observed, therefore the study characterized prognostic markers of response in the pooled population. Change in depression severity was identified by the ratio of the Hamilton-Depression rating scale 17 (HAMD-17) at week 4 divided by the HAMD-17 at baseline (HAMD-17 ratio). Volumes of the lateral ventricles and choroid plexi were positively correlated with the HAMD-17 ratio, indication worse outcome with larger ventricles and choroid plexus volumes, whereas the volume of the central-anterior corpus callosum was negatively correlated with the HAMD-17 ratio. Responders (n = 54) had significantly smaller volumes of the lateral ventricles and CP compared to non-responders (n = 117), whereas the volume of mid-anterior CC was significantly larger compared to non-responders (n = 117), confirming our previous findings. In an exploratory way associations between enlarged LVV and CPV and signs of lipid dysregulation were observed. In conclusion, we confirmed that volumes of lateral ventricles, choroid plexi and the mid-anterior corpus callosum are associated with clinical improvement of depression and may be indicators of metabolic/inflammatory activity.
Collapse
Affiliation(s)
- Harald Murck
- Dept. of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Maurizio Fava
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Cusin
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cherise Chin Fatt
- The University of Texas Southwestern Medical Center, Department of Psychiatry, Center for Depression Research and Clinical Care, Dallas, USA
| | - Madhukar Trivedi
- The University of Texas Southwestern Medical Center, Department of Psychiatry, Center for Depression Research and Clinical Care, Dallas, USA
| |
Collapse
|
9
|
Pańczyszyn-Trzewik P, Czechowska E, Stachowicz K, Sowa-Kućma M. The Importance of α-Klotho in Depression and Cognitive Impairment and Its Connection to Glutamate Neurotransmission-An Up-to-Date Review. Int J Mol Sci 2023; 24:15268. [PMID: 37894946 PMCID: PMC10607524 DOI: 10.3390/ijms242015268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/11/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023] Open
Abstract
Depression is a serious neuropsychiatric disease affecting an increasing number of people worldwide. Cognitive deficits (including inattention, poor memory, and decision-making difficulties) are common in the clinical picture of depression. Cognitive impairment has been hypothesized to be one of the most important components of major depressive disorder (MDD; referred to as clinical depression), although typical cognitive symptoms are less frequent in people with depression than in people with schizophrenia or bipolar disorder (BD; sometimes referred to as manic-depressive disorder). The importance of α-Klotho in the aging process has been well-documented. Growing evidence points to the role of α-Klotho in regulating other biological functions, including responses to oxidative stress and the modulation of synaptic plasticity. It has been proven that a Klotho deficit may contribute to the development of various nervous system pathologies, such as behavioral disorders or neurodegeneration. Given the growing evidence of the role of α-Klotho in depression and cognitive impairment, it is assumed that this protein may be a molecular link between them. Here, we provide a research review of the role of α-Klotho in depression and cognitive impairment. Furthermore, we propose potential mechanisms (related to oxidative stress and glutamatergic transmission) that may be important in α-Klotho-mediated regulation of mental and cognitive function.
Collapse
Affiliation(s)
- Patrycja Pańczyszyn-Trzewik
- Department of Human Physiology, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland; (P.P.-T.); (E.C.)
| | - Ewelina Czechowska
- Department of Human Physiology, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland; (P.P.-T.); (E.C.)
| | - Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland;
| | - Magdalena Sowa-Kućma
- Department of Human Physiology, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland; (P.P.-T.); (E.C.)
- Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, Warzywna Street 1A, 35-595 Rzeszow, Poland
| |
Collapse
|
10
|
Philip VM, He H, Saul MC, Dickson PE, Bubier JA, Chesler EJ. Gene expression genetics of the striatum of Diversity Outbred mice. Sci Data 2023; 10:522. [PMID: 37543624 PMCID: PMC10404230 DOI: 10.1038/s41597-023-02426-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/28/2023] [Indexed: 08/07/2023] Open
Abstract
Brain transcriptional variation is a heritable trait that mediates complex behaviors, including addiction. Expression quantitative trait locus (eQTL) mapping reveals genomic regions harboring genetic variants that influence transcript abundance. In this study, we profiled transcript abundance in the striatum of 386 Diversity Outbred (J:DO) mice of both sexes using RNA-Seq. All mice were characterized using a behavioral battery of widely-used exploratory and risk-taking assays prior to transcriptional profiling. We performed eQTL mapping, incorporated the results into a browser-based eQTL viewer, and deposited co-expression network members in GeneWeaver. The eQTL viewer allows researchers to query specific genes to obtain allelic effect plots, analyze SNP associations, assess gene expression correlations, and apply mediation analysis to evaluate whether the regulatory variant is acting through the expression of another gene. GeneWeaver allows multi-species comparison of gene sets using statistical and combinatorial tools. This data resource allows users to find genetic variants that regulate differentially expressed transcripts and place them in the context of other studies of striatal gene expression and function in addiction-related behavior.
Collapse
Affiliation(s)
- Vivek M Philip
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, 04605, USA
| | - Hao He
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Michael C Saul
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, 04605, USA
| | - Price E Dickson
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine Marshall University, Huntington, WV, 25703, USA
| | - Jason A Bubier
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, 04605, USA
| | - Elissa J Chesler
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, 04605, USA.
| |
Collapse
|
11
|
Philip VM, He H, Saul MC, Dickson PE, Bubier JA, Chesler EJ. Gene expression genetics of the striatum of Diversity Outbred mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.11.540390. [PMID: 37214980 PMCID: PMC10197688 DOI: 10.1101/2023.05.11.540390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Brain transcriptional variation is a heritable trait that mediates complex behaviors, including addiction. Expression quantitative trait locus (eQTL) mapping reveals genomic regions harboring genetic variants that influence transcript abundance. In this study, we profiled transcript abundance in the striatum of 386 Diversity Outbred (J:DO) mice of both sexes using RNA-Seq. All mice were characterized using a behavioral battery of widely-used exploratory and risk-taking assays prior to transcriptional profiling. We performed eQTL mapping, incorporated the results into a browser-based eQTL viewer, and deposited co-expression network members in GeneWeaver. The eQTL viewer allows researchers to query specific genes to obtain allelic effect plots, analyze SNP associations, assess gene expression correlations, and apply mediation analysis to evaluate whether the regulatory variant is acting through the expression of another gene. GeneWeaver allows multi-species comparison of gene sets using statistical and combinatorial tools. This data resource allows users to find genetic variants that regulate differentially expressed transcripts and place them in the context of other studies of striatal gene expression and function in addiction-related behavior.
Collapse
Affiliation(s)
- Vivek M. Philip
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04605
| | - Hao He
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | - Michael C. Saul
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04605
| | - Price E. Dickson
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine Marshall University, 1700 3rd Ave. Huntington, WV 25703
| | - Jason A. Bubier
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04605
| | | |
Collapse
|
12
|
Zhang J, Zhang R, Peng Y, Aa J, Wang G. AI Machine Learning Technique Characterizes Potential Markers of Depression in Two Animal Models of Depression. Brain Sci 2023; 13:brainsci13050763. [PMID: 37239235 DOI: 10.3390/brainsci13050763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/26/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023] Open
Abstract
(1) Background: there is an urgent clinical need for rapid and effective antidepressants. (2) Methods: We employed proteomics to profile proteins in two animal models (n = 48) of Chronic Unpredictable Stress and Chronic Social Defeat Stress. Additionally, partial least squares projection to latent structure discriminant analysis and machine learning were used to distinguish the models and the healthy control, extract and select protein features and build biomarker panels for the identification of different mouse models of depression. (3) Results: The two depression models were significantly different from the healthy control, and there were common changes in proteins in the depression-related brain regions of the two models; i.e., SRCN1 was down-regulated in the dorsal raphe nucleus in both models of depression. Additionally, SYIM was up-regulated in the medial prefrontal cortex in the two depression models. Bioinformatics analysis suggested that perturbed proteins are involved in energy metabolism, nerve projection, etc. Further examination confirmed that the trends of feature proteins were consistent with mRNA expression levels. (4) Conclusions: To the best of our knowledge, this is the first study to probe new targets of depression in multiple brain regions of two typical models of depression, which could be targets worthy of study.
Collapse
Affiliation(s)
- Jing Zhang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Ran Zhang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Ying Peng
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Jiye Aa
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| | - Guangji Wang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, Nanjing 210009, China
| |
Collapse
|
13
|
Senay O, Seethaler M, Makris N, Yeterian E, Rushmore J, Cho KIK, Rizzoni E, Heller C, Pasternak O, Szczepankiewicz F, Westin C, Losak J, Ustohal L, Tomandl J, Vojtisek L, Kudlicka P, Kikinis Z, Holt D, Lewandowski KE, Lizano P, Keshavan MS, Öngür D, Kasparek T, Breier A, Shenton ME, Seitz‐Holland J, Kubicki M. A preliminary choroid plexus volumetric study in individuals with psychosis. Hum Brain Mapp 2023; 44:2465-2478. [PMID: 36744628 PMCID: PMC10028672 DOI: 10.1002/hbm.26224] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 12/13/2022] [Accepted: 01/21/2023] [Indexed: 02/07/2023] Open
Abstract
The choroid plexus (ChP) is part of the blood-cerebrospinal fluid barrier, regulating brain homeostasis and the brain's response to peripheral events. Its upregulation and enlargement are considered essential in psychosis. However, the timing of the ChP enlargement has not been established. This study introduces a novel magnetic resonance imaging-based segmentation method to examine ChP volumes in two cohorts of individuals with psychosis. The first sample consists of 41 individuals with early course psychosis (mean duration of illness = 1.78 years) and 30 healthy individuals. The second sample consists of 30 individuals with chronic psychosis (mean duration of illness = 7.96 years) and 34 healthy individuals. We utilized manual segmentation to measure ChP volumes. We applied ANCOVAs to compare normalized ChP volumes between groups and partial correlations to investigate the relationship between ChP, LV volumes, and clinical characteristics. Our segmentation demonstrated good reliability (.87). We further showed a significant ChP volume increase in early psychosis (left: p < .00010, right: p < .00010) and a significant positive correlation between higher ChP and higher LV volumes in chronic psychosis (left: r = .54, p = .0030, right: r = .68; p < .0010). Our study suggests that ChP enlargement may be a marker of acute response around disease onset. It might also play a modulatory role in the chronic enlargement of lateral ventricles, often reported in psychosis. Future longitudinal studies should investigate the dynamics of ChP enlargement as a promising marker for novel therapeutic strategies.
Collapse
Affiliation(s)
- Olcay Senay
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of PsychiatryIstanbul Faculty of Medicine, Istanbul UniversityIstanbulTurkey
| | - Magdalena Seethaler
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of Psychiatry and Psychotherapy, Campus Charité MittePsychiatric University Hospital Charité at St. Hedwig Hospital, Charité‐Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
| | - Nikos Makris
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of Anatomy and NeurobiologyBoston University School of MedicineBostonMassachusettsUSA
- Center for Morphometric Analysis, Department of PsychiatryMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Edward Yeterian
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Center for Morphometric Analysis, Department of PsychiatryMassachusetts General HospitalCharlestownMassachusettsUSA
- Department of PsychologyColby CollegeWatervilleMaineUSA
| | - Jarrett Rushmore
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of Anatomy and NeurobiologyBoston University School of MedicineBostonMassachusettsUSA
- Center for Morphometric Analysis, Department of PsychiatryMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Kang Ik K. Cho
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Elizabeth Rizzoni
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Carina Heller
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of Clinical PsychologyFriedrich‐Schiller‐University JenaJenaGermany
| | - Ofer Pasternak
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Filip Szczepankiewicz
- Department of RadiologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Carl‐Frederik Westin
- Department of RadiologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Jan Losak
- Central European Institute of Technology (CEITEC)Masaryk University, Neuroscience Centre, Brno, Czech Republic; Departments of Psychiatry and Biochemistry, Faculty of Medicine, Masaryk University and University Hospital BrnoBrnoCzech Republic
| | - Libor Ustohal
- Central European Institute of Technology (CEITEC)Masaryk University, Neuroscience Centre, Brno, Czech Republic; Departments of Psychiatry and Biochemistry, Faculty of Medicine, Masaryk University and University Hospital BrnoBrnoCzech Republic
| | - Josef Tomandl
- Central European Institute of Technology (CEITEC)Masaryk University, Neuroscience Centre, Brno, Czech Republic; Departments of Psychiatry and Biochemistry, Faculty of Medicine, Masaryk University and University Hospital BrnoBrnoCzech Republic
| | - Lubomir Vojtisek
- Central European Institute of Technology (CEITEC)Masaryk University, Neuroscience Centre, Brno, Czech Republic; Departments of Psychiatry and Biochemistry, Faculty of Medicine, Masaryk University and University Hospital BrnoBrnoCzech Republic
| | - Peter Kudlicka
- Central European Institute of Technology (CEITEC)Masaryk University, Neuroscience Centre, Brno, Czech Republic; Departments of Psychiatry and Biochemistry, Faculty of Medicine, Masaryk University and University Hospital BrnoBrnoCzech Republic
| | - Zora Kikinis
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Daphne Holt
- Department of PsychiatryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Paulo Lizano
- Department of Psychiatry, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Matcheri S. Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Dost Öngür
- Department of Psychiatry, McLean HospitalHarvard Medical SchoolBelmontMassachusettsUSA
| | - Tomas Kasparek
- Department of Psychiatry, Faculty of MedicineMasaryk University and University Hospital BrnoBrnoCzech Republic
| | - Alan Breier
- Department of PsychiatryIndiana University School of MedicineIndianapolisIndianaUSA
| | - Martha E. Shenton
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of RadiologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Johanna Seitz‐Holland
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of PsychiatryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Marek Kubicki
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of RadiologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of PsychiatryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
14
|
Murck H, Fava M, Cusin C, Chin Fatt C, Trivedi M. Brain Ventricle and Choroid Plexus Morphology as Predictor of Treatment Response: Findings from the EMBARC Study. RESEARCH SQUARE 2023:rs.3.rs-2618151. [PMID: 36909585 PMCID: PMC10002825 DOI: 10.21203/rs.3.rs-2618151/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Recent observations suggest a role of the choroid plexus (CP) and cerebral ventricle volume (CV), to identify treatment resistance of major depressive disorder (MDD). We tested the hypothesis that these markers are associated with clinical improvement in subjects from the EMBARC study, as implied by a recent pilot study. The EMBARC study characterized biological markers in a randomized placebo-controlled trial of sertraline vs. placebo in patients with MDD. Association of baseline volumes of CV, CP and of the corpus callosum (CC) with treatment response after 4 weeks treatment were evaluated. 171 subjects (61 male, 110 female) completed the 4 week assessments; gender, site and age were taken into account for this analyses. As previously reported, no treatment effect of sertraline was observed, but prognostic markers for clinical improvement were identified. Responders (n = 54) had significantly smaller volumes of the CP and lateral ventricles, whereas the volume of mid-anterior and mid-posterior CC was significantly larger compared to non-responders (n = 117). A positive correlation between CV volume and CP volume was observed, whereas a negative correlation between CV volume and both central-anterior and central-posterior parts of the CC emerged. In an exploratory way correlations between enlarged VV and CP volume on the one hand and signs of metabolic syndrome, in particular triglyceride plasma concentrations, were observed. A primary abnormality of CP function in MDD may be associated with increased ventricles, compression of white matter volume, which may affect treatment response speed or outcome. Metabolic markers may mediate this relationship.
Collapse
Affiliation(s)
- Harald Murck
- Dept. of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Maurizio Fava
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Cusin
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cherise Chin Fatt
- The University of Texas Southwestern Medical Center, Department of Psychiatry, Center for Depression Research and Clinical Care, Department of Psychiatry, Dallas, USA
| | - Madhukar Trivedi
- The University of Texas Southwestern Medical Center, Department of Psychiatry, Center for Depression Research and Clinical Care, Department of Psychiatry, Dallas, USA
| |
Collapse
|
15
|
Murck H, Lehr L, Jezova D. A viewpoint on aldosterone and BMI related brain morphology in relation to treatment outcome in patients with major depression. J Neuroendocrinol 2023; 35:e13219. [PMID: 36539978 DOI: 10.1111/jne.13219] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/06/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
Abstract
An abundance of knowledge has been collected describing the involvement of neuroendocrine parameters in major depression. The hypothalamic-pituitary-adrenocortical (HPA) axis regulating cortisol release has been extensively studied; however, attempts to target the HPA axis pharmacologically to treat major depression have failed. This review focuses on the importance of the adrenocortical stress hormone aldosterone, which is released by adrenocorticotropic hormone and angiotensin, and the mineralocorticoid receptor (MR) in depression. Depressed patients, in particular those with atypical depression, have signs of central hyperactivation of the aldosterone sensitive MR, potentially as a consequence of a reactive aldosterone release induced by low blood pressure and as a result of low sensitivity of peripheral MR. This is reflected in reduced heart rate variability, increased salt appetite and sleep changes in this group of patients. In addition, enlarged brain ventricles, compressed corpus callosum and changes of the choroid plexus are associated with increased aldosterone (in relation to cortisol). Furthermore, subjects with these features often show obesity. These characteristics are related to a worse antidepressant treatment outcome. Alterations in choroid plexus function as a consequence of increased aldosterone levels, autonomic dysregulation, metabolic changes and/or inflammation may be involved. The characterization of this regulatory system is in its early days but may identify new targets for therapeutic interventions.
Collapse
Affiliation(s)
- Harald Murck
- Philipps-University Marburg, Marburg, Germany
- Murck-Neuroscience LLC Westfield, Westfield, NJ, USA
| | - Lisa Lehr
- Department of Nephrology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Daniela Jezova
- Slovak Academy of Sciences, Biomedical Research Center, Institute of Experimental Endocrinology, Bratislava, Slovakia
| |
Collapse
|
16
|
Klotho Regulated by Estrogen Plays a Key Role in Sex Differences in Stress Resilience in Rats. Int J Mol Sci 2023; 24:ijms24021206. [PMID: 36674721 PMCID: PMC9862442 DOI: 10.3390/ijms24021206] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
Klotho (KL) is a glycosyl hydrolase and aging-suppressor gene. Stress is a risk factor for depression and anxiety, which are highly comorbid with each other. The aim of this study is to determine whether KL is regulated by estrogen and plays an important role in sex differences in stress resilience. Our results showed that KL is regulated by estrogen in rat hippocampal neurons in vivo and in vitro and is essential for the estrogen-mediated increase in the number of presynaptic vesicular glutamate transporter 1 (Vglut1)-positive clusters on the dendrites of hippocampal neurons. The role of KL in sex differences in stress response was examined in rats using 3-week chronic unpredictable mild stress (CUMS). CUMS produced a deficit in spatial learning and memory, anhedonic-like behaviors, and anxiety-like behaviors in male but not female rats, which was accompanied by a reduction in KL protein levels in the hippocampus of male but not female rats. This demonstrated the resilience of female rats to CUMS. Interestingly, the knockdown of KL protein levels in the rat hippocampus of both sexes caused a decrease in stress resilience in both sexes, especially in female rats. These results suggest that the regulation of KL by estrogen plays an important role in estrogen-mediated synapse formation and that KL plays a critical role in the sex differences in cognitive deficit, anhedonic-like behaviors, and anxiety-like behaviors induced by chronic stress in rats, highlighting an important role of KL in sex differences in stress resilience.
Collapse
|
17
|
Stankiewicz AM, Jaszczyk A, Goscik J, Juszczak GR. Stress and the brain transcriptome: Identifying commonalities and clusters in standardized data from published experiments. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110558. [PMID: 35405299 DOI: 10.1016/j.pnpbp.2022.110558] [Citation(s) in RCA: 3] [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: 07/06/2021] [Revised: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 12/28/2022]
Abstract
Interpretation of transcriptomic experiments is hindered by many problems including false positives/negatives inherent to big-data methods and changes in gene nomenclature. To find the most consistent effect of stress on brain transcriptome, we retrieved data from 79 studies applying animal models and 3 human studies investigating post-traumatic stress disorder (PTSD). The analyzed data were obtained either with microarrays or RNA sequencing applied to samples collected from more than 1887 laboratory animals and from 121 human subjects. Based on the initial database containing a quarter million differential expression effect sizes representing transcripts in three species, we identified the most frequently reported genes in 223 stress-control comparisons. Additionally, the analysis considers sex, individual vulnerability and contribution of glucocorticoids. We also found an overlap between gene expression in PTSD patients and animals which indicates relevance of laboratory models for human stress response. Our analysis points to genes that, as far as we know, were not specifically tested for their role in stress response (Pllp, Arrdc2, Midn, Mfsd2a, Ccn1, Htra1, Csrnp1, Tenm4, Tnfrsf25, Sema3b, Fmo2, Adamts4, Gjb1, Errfi1, Fgf18, Galnt6, Slc25a42, Ifi30, Slc4a1, Cemip, Klf10, Tom1, Dcdc2c, Fancd2, Luzp2, Trpm1, Abcc12, Osbpl1a, Ptp4a2). Provided transcriptomic resource will be useful for guiding the new research.
Collapse
Affiliation(s)
- Adrian M Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Poland
| | - Aneta Jaszczyk
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Poland
| | - Joanna Goscik
- Faculty of Computer Science, Bialystok University of Technology, Bialystok, Poland
| | - Grzegorz R Juszczak
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzebiec, Poland.
| |
Collapse
|
18
|
Bitanihirwe BKY, Lizano P, Woo TUW. Deconstructing the functional neuroanatomy of the choroid plexus: an ontogenetic perspective for studying neurodevelopmental and neuropsychiatric disorders. Mol Psychiatry 2022; 27:3573-3582. [PMID: 35618887 PMCID: PMC9133821 DOI: 10.1038/s41380-022-01623-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 02/08/2023]
Abstract
The choroid plexus (CP) is a delicate and highly vascularized structure in the brain comprised of a dense network of fenestrated capillary loops that help in the synthesis, secretion and circulation of cerebrospinal fluid (CSF). This unique neuroanatomical structure is comprised of arachnoid villi stemming from frond-like surface projections-that protrude into the lumen of the four cerebral ventricles-providing a key source of nutrients to the brain parenchyma in addition to serving as a 'sink' for central nervous system metabolic waste. In fact, the functions of the CP are often described as being analogous to those of the liver and kidney. Beyond forming a barrier/interface between the blood and CSF compartments, the CP has been identified as a modulator of leukocyte trafficking, inflammation, cognition, circadian rhythm and the gut brain-axis. In recent years, advances in molecular biology techniques and neuroimaging along with the use of sophisticated animal models have played an integral role in shaping our understanding of how the CP-CSF system changes in relation to the maturation of neural circuits during critical periods of brain development. In this article we provide an ontogenetic perspective of the CP and review the experimental evidence implicating this structure in the pathophysiology of neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Byron K Y Bitanihirwe
- Humanitarian and Conflict Response Institute, University of Manchester, Manchester, UK.
| | - Paulo Lizano
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Translational Neuroscience Division, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tsung-Ung W Woo
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Program in Molecular Neuropathology, McLean Hospital, Belmont, MA, USA
| |
Collapse
|
19
|
Andreassen SN, Toft-Bertelsen TL, Wardman JH, Villadsen R, MacAulay N. Transcriptional profiling of transport mechanisms and regulatory pathways in rat choroid plexus. Fluids Barriers CNS 2022; 19:44. [PMID: 35659263 PMCID: PMC9166438 DOI: 10.1186/s12987-022-00335-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dysregulation of brain fluid homeostasis associates with brain pathologies in which fluid accumulation leads to elevated intracranial pressure. Surgical intervention remains standard care, since specific and efficient pharmacological treatment options are limited for pathologies with disturbed brain fluid homeostasis. Such lack of therapeutic targets originates, in part, from the incomplete map of the molecular mechanisms underlying cerebrospinal fluid (CSF) secretion by the choroid plexus. METHODS The transcriptomic profile of rat choroid plexus was generated by RNA Sequencing (RNAseq) of whole tissue and epithelial cells captured by fluorescence-activated cell sorting (FACS), and compared to proximal tubules. The bioinformatic analysis comprised mapping to reference genome followed by filtering for type, location, and association with alias and protein function. The transporters and associated regulatory modules were arranged in discovery tables according to their transcriptional abundance and tied together in association network analysis. RESULTS The transcriptomic profile of choroid plexus displays high similarity between sex and species (human, rat, and mouse) and lesser similarity to another high-capacity fluid-transporting epithelium, the proximal tubules. The discovery tables provide lists of transport mechanisms that could participate in CSF secretion and suggest regulatory candidates. CONCLUSIONS With quantification of the transport protein transcript abundance in choroid plexus and their potentially linked regulatory modules, we envision a molecular tool to devise rational hypotheses regarding future delineation of choroidal transport proteins involved in CSF secretion and their regulation. Our vision is to obtain future pharmaceutical targets towards modulation of CSF production in pathologies involving disturbed brain water dynamics.
Collapse
Affiliation(s)
- Søren N Andreassen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Trine L Toft-Bertelsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Jonathan H Wardman
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - René Villadsen
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|
20
|
Li HZ, Liu KG, Zeng NX, Wu XF, Lu WJ, Xu HF, Yan C, Wu LL. Luteolin Enhances Choroid Plexus 5-MTHF Brain Transport to Promote Hippocampal Neurogenesis in LOD Rats. Front Pharmacol 2022; 13:826568. [PMID: 35401160 PMCID: PMC8993213 DOI: 10.3389/fphar.2022.826568] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/22/2022] [Indexed: 12/19/2022] Open
Abstract
Folates, provided by food, are commonly used antidepressant synergists in late-onset depression (LOD). However, increased intake of folic acid in the elderly population might lead to the accumulation of unmetabolized folic acid in the systemic circulation, leading to enhanced deterioration of the central nervous system function. In addition, folates cannot access the brain directly because of the blood-brain barrier. Choroid plexus (CP) 5-methyltetrahydrofolate (5-MTHF) brain transport plays a critical role in regulating the cerebrospinal fluid (CSF) 5-MTHF content. Luteolin is a natural flavonoid that has antidepressant effects and is involved in the anti-folate resistance pathway. It remains unclear whether the antidepressant effects of luteolin are associated with the CP 5-MTHF brain transport. In this study, 20-21-month-old Wistar rats were exposed to the chronic unpredictable mild stress (CUMS) protocol for 6 consecutive weeks to explore the long-term effects of luteolin on behavior, 5-MTHF levels, hippocampal neurogenesis, and folate brain transport of the CP. In vitro primary hippocampal neural stem cells (NSCs) cultured in media containing 10% CSF from each group of rats and choroid plexus epithelial cells (CPECs) cultured in media containing 20 μM luteolin were treated with 100 μM corticosterone and 40 mg/ml D-galactose. We found that aged rats exposed to CUMS showed a significantly reduced sucrose preference, decreased locomotion activity in the open field test and accuracy of the Morris water maze test, increased immobility time in the forced swimming test, accelerated dysfunctional neurogenesis and neuronal loss in the dentate gyrus of LOD rats, as well as decreased CSF and hippocampus 5-MTHF levels, and zona occludens protein 1 (ZO-1), proton-coupled folate transporter (PCFT), and reduced folate carrier (RFC) protein levels. In vitro assays showed media containing 10% aged CSF or LOD+ Luteolin-CSF significantly increased the viability of CORT + D-gal-injured NSCs and alleviated dysfunctional neurogenesis and neuronal loss compared with the CORT + D-gal medium. However, media containing 10% LOD-CSF had no such effect. In the meantime, induction of CORT + D-gal significantly decreased the ZO-1, PCFT, RFC, and folate receptor alpha (FR-α) protein levels and transepithelial electrical resistance in rat CPECs. As expected, luteolin treatment was effective in improving these abnormal changes. These findings suggested that luteolin could ameliorate CUMS-induced LOD-like behaviors by enhancing the folate brain transport.
Collapse
Affiliation(s)
- Hui-Zhen Li
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kai-Ge Liu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ning-Xi Zeng
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiao-Feng Wu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wen-Jun Lu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Han-Fang Xu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Can Yan
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li-Li Wu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
21
|
Hopfenmüller VL, Perner B, Reuter H, Bates TJD, Große A, Englert C. The Wilms Tumor Gene wt1a Contributes to Blood-Cerebrospinal Fluid Barrier Function in Zebrafish. Front Cell Dev Biol 2022; 9:809962. [PMID: 35087838 PMCID: PMC8786916 DOI: 10.3389/fcell.2021.809962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/07/2021] [Indexed: 11/30/2022] Open
Abstract
The Wilms tumor suppressor gene Wt1 encodes a zinc finger transcription factor, which is highly conserved among vertebrates. It is a key regulator of urogenital development and homeostasis but also plays a role in other organs including the spleen and the heart. More recently additional functions for Wt1 in the mammalian central nervous system have been described. In contrast to mammals, bony fish possess two paralogous Wt1 genes, namely wt1a and wt1b. By performing detailed in situ hybridization analyses during zebrafish development, we discovered new expression domains for wt1a in the dorsal hindbrain, the caudal medulla and the spinal cord. Marker analysis identified wt1a expressing cells of the dorsal hindbrain as ependymal cells of the choroid plexus in the myelencephalic ventricle. The choroid plexus acts as a blood-cerebrospinal fluid barrier and thus is crucial for brain homeostasis. By employing wt1a mutant larvae and a dye accumulation assay with fluorescent tracers we demonstrate that Wt1a is required for proper choroid plexus formation and function. Thus, Wt1a contributes to the barrier properties of the choroid plexus in zebrafish, revealing an unexpected role for Wt1 in the zebrafish brain.
Collapse
Affiliation(s)
| | - Birgit Perner
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Hanna Reuter
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Thomas J D Bates
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Andreas Große
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Christoph Englert
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,Institute of Biochemistry and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
22
|
Gómez-Arnaiz S, Tate RJ, Grant MH. Cobalt Neurotoxicity: Transcriptional Effect of Elevated Cobalt Blood Levels in the Rodent Brain. TOXICS 2022; 10:toxics10020059. [PMID: 35202246 PMCID: PMC8878729 DOI: 10.3390/toxics10020059] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/11/2022]
Abstract
Metal-on-metal (MoM) hip implants made of cobalt chromium (CoCr) alloy have shown early failure compared with other bearing materials. A consequence of the abnormal wear produced by these prostheses is elevated levels of cobalt in the blood of patients, which can lead to systemic conditions involving cardiac and neurological symptoms. In order to better understand the implications for patients with these implants, we carried out metal content and RNA-Seq analysis of excised tissue from rats treated intraperitonially for 28 days with low concentrations of cobalt. Cobalt blood levels in dosed rats were found to be similar to those seen in some patients with MoM implants (range: 4–38 μg/L Co in blood). Significant accumulation of cobalt was measured in a range of tissues including kidney, liver, and heart, but also in brain tissue. RNA-Seq analysis of neural tissue revealed that exposure to cobalt induces a transcriptional response in the prefrontal cortex (pref. cortex), cerebellum, and hippocampus. Many of the most up- and downregulated genes appear to correspond to choroid plexus transcripts. These results indicate that the choroid plexus could be the brain tissue most affected by cobalt. More specifically, the differentially expressed genes show a disruption of steroidogenesis and lipid metabolism. Several other transcripts also demonstrate that cobalt induces an immune response. In summary, cobalt exposure induces alterations in the brain transcriptome, more specifically, the choroid plexus, which is in direct contact with neurotoxicants at the blood–cerebrospinal fluid barrier.
Collapse
Affiliation(s)
- Sara Gómez-Arnaiz
- Wolfson Centre, Biomedical Engineering Department, University of Strathclyde, Glasgow G4 0NW, UK;
| | - Rothwelle J. Tate
- Strathclyde Institute for Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK;
| | - Mary Helen Grant
- Wolfson Centre, Biomedical Engineering Department, University of Strathclyde, Glasgow G4 0NW, UK;
- Correspondence:
| |
Collapse
|
23
|
Liu LL, Du D, Zheng W, Zhang Y. Age-dependent decline of copper clearance at the blood-cerebrospinal fluid barrier. Neurotoxicology 2022; 88:44-56. [PMID: 34718061 PMCID: PMC8748412 DOI: 10.1016/j.neuro.2021.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 10/16/2021] [Accepted: 10/26/2021] [Indexed: 02/08/2023]
Abstract
The homeostasis of copper (Cu) in the central nervous system is regulated by the blood-brain barrier and blood-cerebrospinal (CSF) barrier (BCB) in the choroid plexus. While proteins responsible for Cu uptake, release, storage and intracellular trafficking exist in the choroid plexus, the influence of age on Cu clearance from the CSF via the choroid plexus and how Cu transporting proteins contribute to the process are unelucidated. This study was designed to test the hypothesis that the aging process diminishes Cu clearance from the CSF of rats by disrupting Cu transporting proteins in the choroid plexus. Data from ventriculo-cisternal perfusion experiments demonstrated greater 64Cu radioactivity in the CSF effluents of older rats (18 months) compared to younger (1 month) and adult (2 months) rats, suggesting much slower removal of Cu by the choroid plexus in old animals. Studies utilizing qPCR and immunofluorescence revealed an age-specific expression pattern of Cu transporting proteins in the choroid plexus. Moreover, proteomic analyses unraveled age-specific proteomes in the choroid plexus with distinct pathway differences, particularly associated with extracellular matrix and neurodevelopment between young and old animals. Taken together, these findings support an age-dependent deterioration in CSF Cu clearance, which appears to be associated with altered subcellular distribution of Cu transporting proteins and proteomes in the choroid plexus.
Collapse
Affiliation(s)
- Luke L. Liu
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - David Du
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA,To whom correspondences shall be sent : Wei Zheng, Ph.D. (contact corresponding author), School of Health Sciences, Purdue University, 550 Stadium Mall Drive, HAMP-1273, West Lafayette, IN 47907, Phone: (765) 496-6447, , Yanshu Zhang, Ph.D., School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China,
| | - Yanshu Zhang
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA,School of Public Health, North China University of Science and Technology, Tangshan, China,To whom correspondences shall be sent : Wei Zheng, Ph.D. (contact corresponding author), School of Health Sciences, Purdue University, 550 Stadium Mall Drive, HAMP-1273, West Lafayette, IN 47907, Phone: (765) 496-6447, , Yanshu Zhang, Ph.D., School of Public Health, North China University of Science and Technology, Tangshan, Hebei, China,
| |
Collapse
|
24
|
The association of matrix metalloproteinase 9 (MMP9) with hippocampal volume in schizophrenia: a preliminary MRI study. Neuropsychopharmacology 2022; 47:524-530. [PMID: 33833403 PMCID: PMC8674225 DOI: 10.1038/s41386-021-00997-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/13/2021] [Accepted: 03/05/2021] [Indexed: 02/02/2023]
Abstract
Matrix metalloproteinases 9 (MMP9) are enzymes involved in regulating neuroplasticity in the hippocampus. This, combined with evidence for disrupted hippocampal structure and function in schizophrenia, has prompted our current investigation into the relationship between MMP9 and hippocampal volumes in schizophrenia. 34 healthy individuals (mean age = 32.50, male = 21, female = 13) and 30 subjects with schizophrenia (mean age = 33.07, male = 19, female = 11) underwent a blood draw and T1-weighted magnetic resonance imaging. The hippocampus was automatically segmented utilizing FreeSurfer. MMP9 plasma levels were measured with ELISA. ANCOVAs were conducted to compare MMP9 plasma levels (corrected for age and sex) and hippocampal volumes between groups (corrected for age, sex, total intracranial volume). Spearman correlations were utilized to investigate the relationship between symptoms, medication, duration of illness, number of episodes, and MMP9 plasma levels in patients. Last, we explored the correlation between MMP9 levels and hippocampal volumes in patients and healthy individuals separately. Patients displayed higher MMP9 plasma levels than healthy individuals (F(1, 60) = 21.19, p < 0.0001). MMP9 levels correlated with negative symptoms in patients (R = 0.39, p = 0.035), but not with medication, duration of illness, or the number of episodes. Further, patients had smaller left (F(1,59) = 9.12, p = 0.0040) and right (F(1,59) = 6.49, p = 0.013) hippocampal volumes. Finally, left (R = -0.39, p = 0.034) and right (R = -0.37, p = 0.046) hippocampal volumes correlated negatively with MMP9 plasma levels in patients. We observe higher MMP9 plasma levels in SCZ, associated with lower hippocampal volumes, suggesting involvement of MMP9 in the pathology of SCZ. Future studies are needed to investigate how MMP9 influences the pathology of SCZ over the lifespan, whether the observed associations are specific for schizophrenia, and if a therapeutic modulation of MMP9 promotes neuroprotective effects in SCZ.
Collapse
|
25
|
Wang YJ, Downey MA, Choi S, Shoup TM, Elmaleh DR. Cromolyn platform suppresses fibrosis and inflammation, promotes microglial phagocytosis and neurite outgrowth. Sci Rep 2021; 11:22161. [PMID: 34772945 PMCID: PMC8589953 DOI: 10.1038/s41598-021-00465-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/07/2021] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases are characterized by chronic neuroinflammation and may perpetuate ongoing fibrotic reactions within the central nervous system. Unfortunately, there is no therapeutic available that treats neurodegenerative inflammation and its sequelae. Here we utilize cromolyn, a mast cell inhibitor with anti-inflammatory capabilities, and its fluorinated analogue F-cromolyn to study fibrosis-related protein regulation and secretion downstream of neuroinflammation and their ability to promote microglial phagocytosis and neurite outgrowth. In this report, RNA-seq analysis shows that administration of the pro-inflammatory cytokine TNF-α to HMC3 human microglia results in a robust upregulation of fibrosis-associated genes. Subsequent treatment with cromolyn and F-cromolyn resulted in reduced secretion of collagen XVIII, fibronectin, and tenascin-c. Additionally, we show that cromolyn and F-cromolyn reduce pro-inflammatory proteins PLP1, PELP1, HSP90, IL-2, GRO-α, Eotaxin, and VEGF-Α, while promoting secretion of anti-inflammatory IL-4 in HMC3 microglia. Furthermore, cromolyn and F-cromolyn augment neurite outgrowth in PC12 neuronal cells in concert with nerve growth factor. Treatment also differentially altered secretion of neurogenesis-related proteins TTL, PROX1, Rab35, and CSDE1 in HMC3 microglia. Finally, iPSC-derived human microglia more readily phagocytose Aβ42 with cromolyn and F-cromolyn relative to controls. We propose the cromolyn platform targets multiple proteins upstream of PI3K/Akt/mTOR, NF-κB, and GSK-3β signaling pathways to affect cytokine, chemokine, and fibrosis-related protein expression.
Collapse
Affiliation(s)
| | | | - Sungwoon Choi
- Department of New Drug Discovery, Chungnam National University, Daejeon, South Korea
| | - Timothy M Shoup
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129-2060, USA
| | - David R Elmaleh
- AZTherapies, Inc., Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129-2060, USA.
| |
Collapse
|
26
|
Herzog DP, Perumal N, Manicam C, Treccani G, Nadig J, Rossmanith M, Engelmann J, Jene T, Hasch A, van der Kooij MA, Lieb K, Gassen NC, Grus FH, Müller MB. Longitudinal CSF proteome profiling in mice to uncover the acute and sustained mechanisms of action of rapid acting antidepressant (2R,6R)-hydroxynorketamine (HNK). Neurobiol Stress 2021; 15:100404. [PMID: 34632008 PMCID: PMC8488754 DOI: 10.1016/j.ynstr.2021.100404] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/16/2021] [Accepted: 09/26/2021] [Indexed: 02/04/2023] Open
Abstract
Delayed onset of antidepressant action is a shortcoming in depression treatment. Ketamine and its metabolite (2R,6R)-hydroxynorketamine (HNK) have emerged as promising rapid-acting antidepressants. However, their mechanism of action remains unknown. In this study, we first described the anxious and depression-prone inbred mouse strain, DBA/2J, as an animal model to assess the antidepressant-like effects of ketamine and HNK in vivo. To decode the molecular mechanisms mediating HNK's rapid antidepressant effects, a longitudinal cerebrospinal fluid (CSF) proteome profiling of its acute and sustained effects was conducted using an unbiased, hypothesis-free mass spectrometry-based proteomics approach. A total of 387 proteins were identified, with a major implication of significantly differentially expressed proteins in the glucocorticoid receptor (GR) signaling pathway, providing evidence for a link between HNK and regulation of the stress hormone system. Mechanistically, we identified HNK to repress GR-mediated transcription and reduce hormonal sensitivity of GR in vitro. In addition, mammalian target of rapamycin (mTOR) and brain-derived neurotrophic factor (BDNF) were predicted to be important upstream regulators of HNK treatment. Our results contribute to precise understanding of the temporal dynamics and molecular targets underlying HNK's rapid antidepressant-like effects, which can be used as a benchmark for improved treatment strategies for depression in future.
Collapse
Affiliation(s)
- David P Herzog
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Natarajan Perumal
- Experimental and Translational Ophthalmology, Department of Ophthalmology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Caroline Manicam
- Experimental and Translational Ophthalmology, Department of Ophthalmology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Giulia Treccani
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany.,Institute for Microscopic Anatomy and Neurobiology, Johannes Gutenberg University Medical Center, Mainz, Germany.,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Jens Nadig
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Milena Rossmanith
- Experimental and Translational Ophthalmology, Department of Ophthalmology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Jan Engelmann
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Tanja Jene
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Annika Hasch
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Michael A van der Kooij
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany.,Leibniz Institute for Resilience Research, Mainz, Germany
| | - Klaus Lieb
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany.,Leibniz Institute for Resilience Research, Mainz, Germany
| | - Nils C Gassen
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, University Medical Center Bonn, Bonn, Germany
| | - Franz H Grus
- Experimental and Translational Ophthalmology, Department of Ophthalmology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Marianne B Müller
- Department of Psychiatry and Psychotherapy and Focus Program Translational Neurosciences, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany.,Leibniz Institute for Resilience Research, Mainz, Germany
| |
Collapse
|
27
|
Plectin in the Central Nervous System and a Putative Role in Brain Astrocytes. Cells 2021; 10:cells10092353. [PMID: 34572001 PMCID: PMC8464768 DOI: 10.3390/cells10092353] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Plectin, a high-molecular-mass cytolinker, is abundantly expressed in the central nervous system (CNS). Currently, a limited amount of data about plectin in the CNS prevents us from seeing the complete picture of how plectin affects the functioning of the CNS as a whole. Yet, by analogy to its role in other tissues, it is anticipated that, in the CNS, plectin also functions as the key cytoskeleton interlinking molecule. Thus, it is likely involved in signalling processes, thereby affecting numerous fundamental functions in the brain and spinal cord. Versatile direct and indirect interactions of plectin with cytoskeletal filaments and enzymes in the cells of the CNS in normal physiological and in pathologic conditions remain to be fully addressed. Several pathologies of the CNS related to plectin have been discovered in patients with plectinopathies. However, in view of plectin as an integrator of a cohesive mesh of cellular proteins, it is important that the role of plectin is also considered in other CNS pathologies. This review summarizes the current knowledge of plectin in the CNS, focusing on plectin isoforms that have been detected in the CNS, along with its expression profile and distribution alongside diverse cytoskeleton filaments in CNS cell types. Considering that the bidirectional communication between neurons and glial cells, especially astrocytes, is crucial for proper functioning of the CNS, we place particular emphasis on the known roles of plectin in neurons, and we propose possible roles of plectin in astrocytes.
Collapse
|
28
|
Ivan DC, Walthert S, Berve K, Steudler J, Locatelli G. Dwellers and Trespassers: Mononuclear Phagocytes at the Borders of the Central Nervous System. Front Immunol 2021; 11:609921. [PMID: 33746939 PMCID: PMC7973121 DOI: 10.3389/fimmu.2020.609921] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/29/2020] [Indexed: 01/02/2023] Open
Abstract
The central nervous system (CNS) parenchyma is enclosed and protected by a multilayered system of cellular and acellular barriers, functionally separating glia and neurons from peripheral circulation and blood-borne immune cells. Populating these borders as dynamic observers, CNS-resident macrophages contribute to organ homeostasis. Upon autoimmune, traumatic or neurodegenerative inflammation, these phagocytes start playing additional roles as immune regulators contributing to disease evolution. At the same time, pathological CNS conditions drive the migration and recruitment of blood-borne monocyte-derived cells across distinct local gateways. This invasion process drastically increases border complexity and can lead to parenchymal infiltration of blood-borne phagocytes playing a direct role both in damage and in tissue repair. While recent studies and technical advancements have highlighted the extreme heterogeneity of these resident and CNS-invading cells, both the compartment-specific mechanism of invasion and the functional specification of intruding and resident cells remain unclear. This review illustrates the complexity of mononuclear phagocytes at CNS interfaces, indicating how further studies of CNS border dynamics are crucially needed to shed light on local and systemic regulation of CNS functions and dysfunctions.
Collapse
|
29
|
Abstract
Group B Streptococcus (GBS) remains the leading cause of neonatal meningitis, a disease associated with high rates of adverse neurological sequelae. The in vivo relationship between GBS and brain tissues remains poorly characterized, partly because past studies had focused on microbial rather than host processes. Additionally, the field has not capitalized on systems-level technologies to probe the host-pathogen relationship. Here, we use multiplexed quantitative proteomics to investigate the effect of GBS infection in the murine brain at various levels of tissue complexity, beginning with the whole organ and moving to brain vascular substructures. Infected whole brains showed classical signatures associated with the acute-phase response. In isolated brain microvessels, classical blood-brain barrier proteins were unaltered, but interferon signaling and leukocyte recruitment proteins were upregulated. The choroid plexus showed increases in peripheral immune cell proteins. Proteins that increased in abundance in the vasculature during GBS invasion were associated with major histocompatibility complex (MHC) class I antigen processing and endoplasmic reticulum dysfunction, a finding which correlated with altered host protein glycosylation profiles. Globally, there was low concordance between the infection proteome of whole brains and isolated vascular tissues. This report underscores the utility of unbiased, systems-scale analyses of functional tissue substructures for understanding disease.IMPORTANCE Group B Streptococcus (GBS) meningitis remains a major cause of poor health outcomes very early in life. Both the host-pathogen relationship leading to disease and the massive host response to infection contributing to these poor outcomes are orchestrated at the tissue and cell type levels. GBS meningitis is thought to result when bacteria present in the blood circumvent the selectively permeable vascular barriers that feed the brain. Additionally, tissue damage subsequent to bacterial invasion is mediated by inflammation and by immune cells from the periphery crossing the blood-brain barrier. Indeed, the vasculature plays a central role in disease processes occurring during GBS infection of the brain. Here, we employed quantitative proteomic analysis of brain vascular substructures during invasive GBS disease. We used the generated data to map molecular alterations associated with tissue perturbation, finding widespread intracellular dysfunction and punctuating the importance of investigations relegated to tissue type over the whole organ.
Collapse
|
30
|
Pellegrini L, Bonfio C, Chadwick J, Begum F, Skehel M, Lancaster MA. Human CNS barrier-forming organoids with cerebrospinal fluid production. Science 2020; 369:eaaz5626. [PMID: 32527923 PMCID: PMC7116154 DOI: 10.1126/science.aaz5626] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 05/22/2020] [Indexed: 12/13/2022]
Abstract
Cerebrospinal fluid (CSF) is a vital liquid, providing nutrients and signaling molecules and clearing out toxic by-products from the brain. The CSF is produced by the choroid plexus (ChP), a protective epithelial barrier that also prevents free entry of toxic molecules or drugs from the blood. Here, we establish human ChP organoids with a selective barrier and CSF-like fluid secretion in self-contained compartments. We show that this in vitro barrier exhibits the same selectivity to small molecules as the ChP in vivo and that ChP-CSF organoids can predict central nervous system (CNS) permeability of new compounds. The transcriptomic and proteomic signatures of ChP-CSF organoids reveal a high degree of similarity to the ChP in vivo. Finally, the intersection of single-cell transcriptomics and proteomic analysis uncovers key human CSF components produced by previously unidentified specialized epithelial subtypes.
Collapse
Affiliation(s)
- Laura Pellegrini
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Claudia Bonfio
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Jessica Chadwick
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Farida Begum
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Mark Skehel
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| |
Collapse
|
31
|
Seiler A, Sood AK, Jenewein J, Fagundes CP. Can stress promote the pathophysiology of brain metastases? A critical review of biobehavioral mechanisms. Brain Behav Immun 2020; 87:860-880. [PMID: 31881262 DOI: 10.1016/j.bbi.2019.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 12/15/2019] [Accepted: 12/20/2019] [Indexed: 01/20/2023] Open
Abstract
Chronic stress can promote tumor growth and progression through immunosuppressive effects and bi-directional interactions between tumor cells and their microenvironment. β-Adrenergic receptor signaling plays a critical role in mediating stress-related effects on tumor progression. Stress-related mechanisms that modulate the dissemination of tumor cells to the brain have received scant attention. Brain metastases are highly resistant to chemotherapy and contribute considerably to morbidity and mortality in various cancers, occurring in up to 20% of patients in some cancer types. Understanding the mechanisms promoting brain metastasis could help to identify interventions that improve disease outcomes. In this review, we discuss biobehavioral, sympathetic, neuroendocrine, and immunological mechanisms by which chronic stress can impact tumor progression and metastatic dissemination to the brain. The critical role of the inflammatory tumor microenvironment in tumor progression and metastatic dissemination to the brain, and its association with stress pathways are delineated. We also discuss translational implications for biobehavioral and pharmacological interventions.
Collapse
Affiliation(s)
- Annina Seiler
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland.
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Josef Jenewein
- Clinic Zugersee, Center for Psychiatry and Psychotherapy, Oberwil-Zug, Switzerland
| | - Christopher P Fagundes
- Department of Psychology, Rice University, Houston, TX, United States; Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
32
|
Solár P, Zamani A, Kubíčková L, Dubový P, Joukal M. Choroid plexus and the blood-cerebrospinal fluid barrier in disease. Fluids Barriers CNS 2020; 17:35. [PMID: 32375819 PMCID: PMC7201396 DOI: 10.1186/s12987-020-00196-2] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/22/2020] [Indexed: 01/08/2023] Open
Abstract
The choroid plexus (CP) forming the blood-cerebrospinal fluid (B-CSF) barrier is among the least studied structures of the central nervous system (CNS) despite its clinical importance. The CP is an epithelio-endothelial convolute comprising a highly vascularized stroma with fenestrated capillaries and a continuous lining of epithelial cells joined by apical tight junctions (TJs) that are crucial in forming the B-CSF barrier. Integrity of the CP is critical for maintaining brain homeostasis and B-CSF barrier permeability. Recent experimental and clinical research has uncovered the significance of the CP in the pathophysiology of various diseases affecting the CNS. The CP is involved in penetration of various pathogens into the CNS, as well as the development of neurodegenerative (e.g., Alzheimer´s disease) and autoimmune diseases (e.g., multiple sclerosis). Moreover, the CP was shown to be important for restoring brain homeostasis following stroke and trauma. In addition, new diagnostic methods and treatment of CP papilloma and carcinoma have recently been developed. This review describes and summarizes the current state of knowledge with regard to the roles of the CP and B-CSF barrier in the pathophysiology of various types of CNS diseases and sets up the foundation for further avenues of research.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, CZ-625 00, Brno, Czech Republic
- Department of Neurosurgery, Faculty of Medicine, Masaryk University and St. Anne´s University Hospital Brno, Pekařská 53, CZ-656 91, Brno, Czech Republic
| | - Alemeh Zamani
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, CZ-625 00, Brno, Czech Republic
| | - Lucie Kubíčková
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, CZ-625 00, Brno, Czech Republic
| | - Petr Dubový
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, CZ-625 00, Brno, Czech Republic
| | - Marek Joukal
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, CZ-625 00, Brno, Czech Republic.
| |
Collapse
|
33
|
Bitanihirwe BKY, Woo TUW. A conceptualized model linking matrix metalloproteinase-9 to schizophrenia pathogenesis. Schizophr Res 2020; 218:28-35. [PMID: 32001079 DOI: 10.1016/j.schres.2019.12.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinase 9 (MMP-9) is an extracellularly operating zinc-dependent endopeptidase that is commonly expressed in the brain, other tissues. It is synthesized in a latent zymogen form known as pro-MMP-9 that is subsequently converted to the active MMP-9 enzyme following cleavage of the pro-domain. Within the central nervous system, MMP-9 is localized and released from neurons, astrocytes and microglia where its expression levels are modulated by cytokines and growth factors during both normal and pathological conditions as well as by reactive oxygen species generated during oxidative stress. MMP-9 is involved in a number of key neurodevelopmental processes that are thought to be affected in schizophrenia, including maturation of the inhibitory neurons that contain the calcium-binding protein parvalbumin, developmental formation of the specialized extracellular matrix structure perineuronal net, synaptic pruning, and myelination. In this context, the present article provides a narrative synthesis of the existing evidence linking MMP-9 dysregulation to schizophrenia pathogenesis. We start by providing an overview of MMP-9 involvement in brain development and physiology. We then discuss the potential mechanisms through which MMP-9 dysregulation may affect neural circuitry maturation as well as how these anomalies may contribute to the disease process of schizophrenia. We conclude by articulating a comprehensive, cogent, and experimentally testable hypothesis linking MMP-9 to the developmental pathophysiologic cascade that triggers the onset and sustains the chronicity of the illness.
Collapse
Affiliation(s)
| | - Tsung-Ung W Woo
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Program in Cellular Neuropathology, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Cicvaric A, Sachernegg HM, Stojanovic T, Symmank D, Smani T, Moeslinger T, Uhrin P, Monje FJ. Podoplanin Gene Disruption in Mice Promotes in vivo Neural Progenitor Cells Proliferation, Selectively Impairs Dentate Gyrus Synaptic Depression and Induces Anxiety-Like Behaviors. Front Cell Neurosci 2020; 13:561. [PMID: 32009902 PMCID: PMC6974453 DOI: 10.3389/fncel.2019.00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Podoplanin (Pdpn), a brain-tumor-related glycoprotein identified in humans and animals, is endogenously expressed in several organs critical for life support such as kidney, lung, heart and brain. In the brain, Pdpn has been identified in proliferative nestin-positive adult neural progenitor cells and in neurons of the neurogenic hippocampal dentate gyrus (DG), a structure associated to anxiety, critical for learning and memory functions and severely damaged in people with Alzheimer's Disease (AD). The in vivo role of Pdpn in adult neurogenesis and anxiety-like behavior remained however unexplored. Using mice with disrupted Pdpn gene as a model organism and applying combined behavioral, molecular biological and electrophysiological assays, we here show that the absence of Pdpn selectively impairs long-term synaptic depression in the neurogenic DG without affecting the CA3-Schaffer's collateral-CA1 synapses. Pdpn deletion also enhanced the proliferative capacity of DG neural progenitor cells and diminished survival of differentiated neuronal cells in vitro. In addition, mice with podoplanin gene disruption showed increased anxiety-like behaviors in experimentally validated behavioral tests as compared to wild type littermate controls. Together, these findings broaden our knowledge on the molecular mechanisms influencing hippocampal synaptic plasticity and neurogenesis in vivo and reveal Pdpn as a novel molecular target for future studies addressing general anxiety disorder and synaptic depression-related memory dysfunctions.
Collapse
Affiliation(s)
- Ana Cicvaric
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Hannah M. Sachernegg
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Tamara Stojanovic
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Dörte Symmank
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Seville (IBiS)/University of Seville/CIBERCV, Seville, Spain
| | - Thomas Moeslinger
- Center for Physiology and Pharmacology, Institute for Physiology, Medical University of Vienna, Vienna, Austria
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Guha D, Lorenz DR, Misra V, Chettimada S, Morgello S, Gabuzda D. Proteomic analysis of cerebrospinal fluid extracellular vesicles reveals synaptic injury, inflammation, and stress response markers in HIV patients with cognitive impairment. J Neuroinflammation 2019; 16:254. [PMID: 31805958 PMCID: PMC6896665 DOI: 10.1186/s12974-019-1617-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/10/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are nano-sized particles present in most body fluids including cerebrospinal fluid (CSF). Little is known about CSF EV proteins in HIV+ individuals. Here, we characterize the CSF EV proteome in HIV+ subjects and its relationship to neuroinflammation, stress responses, and HIV-associated neurocognitive disorders (HAND). METHODS CSF EVs isolated from 20 HIV+ subjects with (n = 10) or without (n = 10) cognitive impairment were characterized by electron microscopy, nanoparticle tracking analysis, immunoblotting, and untargeted LC/MS/MS mass spectrometry. Functional annotation was performed by gene ontology (GO) mapping and expression annotation using Biobase Transfac and PANTHER software. Cultured astrocytic U87 cells were treated with hydrogen peroxide for 4 h to induce oxidative stress and EVs isolated by ultracentrifugation. Selected markers of astrocytes (GFAP, GLUL), inflammation (CRP), and stress responses (PRDX2, PARK7, HSP70) were evaluated in EVs released by U87 cells following induction of oxidative stress and in CSF EVs from HIV+ patients by immunoblotting. RESULTS Mass spectrometry identified 2727 and 1626 proteins in EV fractions and EV-depleted CSF samples, respectively. CSF EV fractions were enriched with exosomal markers including Alix, syntenin, tetraspanins, and heat-shock proteins and a subset of neuronal, astrocyte, oligodendrocyte, and choroid plexus markers, in comparison to EV-depleted CSF. Proteins related to synapses, immune/inflammatory responses, stress responses, metabolic processes, mitochondrial functions, and blood-brain barrier were also identified in CSF EV fractions by GO mapping. HAND subjects had higher abundance of CSF EVs and proteins mapping to GO terms for synapses, glial cells, inflammation, and stress responses compared to those without HAND. GFAP, GLUL, CRP, PRDX2, PARK7, and HSP70 were confirmed by immunoblotting of CSF EVs from subjects with HAND and were also detected in EVs released by U87 cells under oxidative stress. CONCLUSIONS These findings suggest that CSF EVs derived from neurons, glial cells, and choroid plexus carry synaptic, immune/inflammation-related, and stress response proteins in HIV+ individuals with cognitive impairment, representing a valuable source for biomarker discovery.
Collapse
Affiliation(s)
- Debjani Guha
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, CLS 1010, 450 Brookline Ave, Boston, MA, 02215, USA
| | - David R Lorenz
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, CLS 1010, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Vikas Misra
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, CLS 1010, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Sukrutha Chettimada
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, CLS 1010, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Susan Morgello
- Departments of Neurology, Neuroscience and Pathology, Mount Sinai Medical Center, New York, NY, USA
| | - Dana Gabuzda
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, CLS 1010, 450 Brookline Ave, Boston, MA, 02215, USA. .,Department of Neurology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Abstract
Global depletion of klotho accelerates aging, whereas klotho overexpression counteracts aging-related impairments. Why klotho is expressed at much higher levels in the choroid plexus than in other brain regions is unknown. We demonstrate in mice that aging is associated with klotho depletion in the choroid plexus. Reducing klotho selectively within the choroid plexus triggered inflammation within this structure and enhanced activation of innate immune cells within an adjacent brain region following a peripheral immune challenge. In cell culture, we identified a signaling pathway by which klotho suppresses activation of macrophages. Our findings shed light on klotho functions in the choroid plexus and provide a plausible mechanism by which klotho depletion from this structure promotes brain inflammation during the aging process. Located within the brain’s ventricles, the choroid plexus produces cerebrospinal fluid and forms an important barrier between the central nervous system and the blood. For unknown reasons, the choroid plexus produces high levels of the protein klotho. Here, we show that these levels naturally decline with aging. Depleting klotho selectively from the choroid plexus via targeted viral vector-induced knockout in Klothoflox/flox mice increased the expression of multiple proinflammatory factors and triggered macrophage infiltration of this structure in young mice, simulating changes in unmanipulated old mice. Wild-type mice infected with the same Cre recombinase-expressing virus did not show such alterations. Experimental depletion of klotho from the choroid plexus enhanced microglial activation in the hippocampus after peripheral injection of mice with lipopolysaccharide. In primary cultures, klotho suppressed thioredoxin-interacting protein-dependent activation of the NLRP3 inflammasome in macrophages by enhancing fibroblast growth factor 23 signaling. We conclude that klotho functions as a gatekeeper at the interface between the brain and immune system in the choroid plexus. Klotho depletion in aging or disease may weaken this barrier and promote immune-mediated neuropathogenesis.
Collapse
|
37
|
Rostami S, Emami-Aleagha MS, Ghasemi-Kasman M, Allameh A. Cross-talks between the kidneys and the central nervous system in multiple sclerosis. CASPIAN JOURNAL OF INTERNAL MEDICINE 2018; 9:206-210. [PMID: 30197763 PMCID: PMC6121345 DOI: 10.22088/cjim.9.3.206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease, which is considered as a common autoimmune disorder in young adults. A growing number of evidences indicated that the impairment in non-neural tissues plays a significant role in pathology of MS disease. There are bidirectional relationship, metabolic activities and functional similarity between central nervous system (CNS) and kidneys which suggest that kidney tissue may exert remarkable effects on some aspects of MS disorder and CNS impairment in these patients compels the kidney to respond to central inflammation. Recently, it has been well documented that hormonal secretion possesses the important role on CNS abnormalities. In this regard, due to the functional similarity and significant hormonal and non-hormonal relationship between CNS and kidneys, we hypothesized that kidneys exert significant effect on initiation, progression or amelioration of MS disease which might be regarded as potential therapeutic approach in the treatment of MS patients in the future.
Collapse
Affiliation(s)
- Sahar Rostami
- Department of Clinical Biochemistry, Faculty of Medicine, Tarbiat Modarres University, Tehran, Iran
| | | | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medicine, Tarbiat Modarres University, Tehran, Iran
| |
Collapse
|
38
|
Abstract
Brain expression of klotho was first described with the initial discovery of the klotho gene. The prominent age-regulating effects of klotho are attributed to regulation of ion homeostasis through klotho function in the kidney. However, recent advances identified brain functions and cell populations, including adult hippocampal neural progenitors, which require klotho. As well, both human correlational studies and mouse models of disease show that klotho is protective against multiple neurological and psychological disorders. This review focuses on current knowledge as to how the klotho protein effects the brain.
Collapse
Affiliation(s)
- Hai T Vo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ann M Laszczyk
- Department of Cell and Developmental Biology, University of Michigan, Zina Pitcher Pl, Ann Arbor, MI, USA
| | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
39
|
Hoyer C, Sartorius A, Aksay SS, Bumb JM, Janke C, Thiel M, Haffner D, Leifheit-Nestler M, Kranaster L. Electroconvulsive therapy enhances the anti-ageing hormone Klotho in the cerebrospinal fluid of geriatric patients with major depression. Eur Neuropsychopharmacol 2018; 28:428-435. [PMID: 29274997 DOI: 10.1016/j.euroneuro.2017.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/25/2017] [Accepted: 12/06/2017] [Indexed: 02/06/2023]
Abstract
Klotho is a humoral factor with pleiotropic effects. Most notably, Klotho deficiency is associated with a phenotype comprising organ manifestations accompanying aging including atherosclerosis and cognitive impairment. Research on the role of Klotho in affective disorder is scarce, which is surprising in light of the fact that depression is associated with accelerated cellular aging as well as aging-related phenotypes and comorbidity observed in Klotho deficiency. On these grounds we investigated Klotho levels in the cerebrospinal fluid (CSF) and serum of eight geriatric patients undergoing electroconvulsive therapy (ECT) for severe depression. We hypothesize that ECT as a highly effective antidepressant treatment leads enhances Klotho levels. We found a significant difference between pre- and post-ECT CSF Klotho (792.5pg/ml vs. 991.3pg/ml, p=0.0020), but no difference in serum Klotho (602.5 vs. 594.3, p=0.32). Moreover, CSF Klotho increase positively correlated with the number of single ECT sessions that were performed in each patient (F1, 6)=7.84, p=0.031). Conjointly, the results of our exploratory study with a small sample size suggest a central nervous system-specific impact of ECT on Klotho, which may in turn partake in mediating the antidepressant effect of ECT. We suggest the modulation of neuroinflammatory processes, which have been ascribed pathophysiological relevance within the conceptual framework of the neuroinflammation hypothesis of depression, through ECT as a potential mechanism by which Klotho is enhanced in response to treatment. Further preclinical and clinical investigation should aim for a precise identification of the role of Klotho in depressive disorder.
Collapse
Affiliation(s)
- Carolin Hoyer
- Department of Neurology, University Medical Centre Mannheim, Mannheim, Germany
| | - Alexander Sartorius
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Suna Su Aksay
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Jan Malte Bumb
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Christoph Janke
- Department of Anesthesiology and Critical Care Medicine, University Medical Centre Mannheim, Mannheim, Germany
| | - Manfred Thiel
- Department of Anesthesiology and Critical Care Medicine, University Medical Centre Mannheim, Mannheim, Germany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Maren Leifheit-Nestler
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Laura Kranaster
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany.
| |
Collapse
|
40
|
Cruz Y, García EE, Gálvez JV, Arias-Santiago SV, Carvajal HG, Silva-García R, Bonilla-Jaime H, Rojas-Castañeda J, Ibarra A. Release of interleukin-10 and neurotrophic factors in the choroid plexus: possible inductors of neurogenesis following copolymer-1 immunization after cerebral ischemia. Neural Regen Res 2018; 13:1743-1752. [PMID: 30136689 PMCID: PMC6128049 DOI: 10.4103/1673-5374.238615] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Copolymer-1 (Cop-1) is a peptide with immunomodulatory properties, approved by the Food and Drug Administration of United States in the treatment of multiple sclerosis. Cop-1 has been shown to exert neuroprotective effects and induce neurogenesis in cerebral ischemia models. Nevertheless, the mechanism involved in the neurogenic action of this compound remains unknown. The choroid plexus (CP) is a network of cells that constitute the interphase between the immune and central nervous systems, with the ability to mediate neurogenesis through the release of cytokines and growth factors. Therefore, the CP could play a role in Cop-1-induced neurogenesis. In order to determine the participation of the CP in the induction of neurogenesis after Cop-1 immunization, we evaluated the gene expression of various growth factors (brain-derived neurotrophic factor, insulin-like growth factor 1, neurotrophin-3) and cytokines (tumor necrosis factor alpha, interferon-gamma, interleukin-4 (IL-4), IL-10 and IL-17), in the CP at 14 days after ischemia. Furthermore, we analyzed the correlation between the expression of these genes and neurogenesis. Our results showed that Cop-1 was capable of stimulating an upregulation in the expression of the genes encoding for brain-derived neurotrophic factor, insulin-like growth factor 1, neurotrophin-3 and IL-10 in the CP, which correlated with an increase in neurogenesis in the subventricular and subgranular zone. As well, we observed a downregulation of IL-17 gene expression. This study demonstrates the effect of Cop-1 on the expression of growth factors and IL-10 in the CP, in the same way, presents a possible mechanism involved in the neurogenic effect of Cop-1.
Collapse
Affiliation(s)
- Yolanda Cruz
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México; Lab. De Biología de la reproducción, UAMI. Ciudad de México; Doctorado en Ciencias Biológicas, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa. Ciudad de México, México
| | - Edna E García
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México, México
| | - Jessica V Gálvez
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México, México
| | - Stella V Arias-Santiago
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México, México
| | - Horacio G Carvajal
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México, México
| | | | | | - Julio Rojas-Castañeda
- Subdirección de Medicina Experimental, Instituto Nacional de Pediatría. Ciudad de México, México
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Norte, Huixquilucan, Estado de México, México
| |
Collapse
|
41
|
Olauson H, Mencke R, Hillebrands JL, Larsson TE. Tissue expression and source of circulating αKlotho. Bone 2017; 100:19-35. [PMID: 28323144 DOI: 10.1016/j.bone.2017.03.043] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/16/2022]
Abstract
αKlotho (Klotho), a type I transmembrane protein and a coreceptor for Fibroblast Growth Factor-23, was initially thought to be expressed only in a limited number of tissues, most importantly the kidney, parathyroid gland and choroid plexus. Emerging data may suggest a more ubiquitous Klotho expression pattern which has prompted reevaluation of the restricted Klotho paradigm. Herein we systematically review the evidence for Klotho expression in various tissues and cell types in humans and other mammals, and discuss potential reasons behind existing conflicting data. Based on current literature and tissue expression atlases, we propose a classification of tissues into high, intermediate and low/absent Klotho expression. The functional relevance of Klotho in organs with low expression levels remain uncertain and there is currently limited data on a role for membrane-bound Klotho outside the kidney. Finally, we review the evidence for the tissue source of soluble Klotho, and conclude that the kidney is likely to be the principal source of circulating Klotho in physiology.
Collapse
Affiliation(s)
- Hannes Olauson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Rik Mencke
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tobias E Larsson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
42
|
The relevance of α-KLOTHO to the central nervous system: Some key questions. Ageing Res Rev 2017; 36:137-148. [PMID: 28323064 DOI: 10.1016/j.arr.2017.03.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/10/2017] [Accepted: 03/16/2017] [Indexed: 12/20/2022]
Abstract
α-Klotho is well described as an anti-aging protein, with critical roles in kidney function as a transmembrane co-receptor for FGF23, and as a soluble factor in serum. α-Klotho is also expressed in the choroid plexus, where it is released into the cerebrospinal fluid. Nonetheless, α-Klotho is also expressed in the brain parenchyma. Accumulating evidence indicates that this pool of α-Klotho, which we define as brain α-Klotho, may play important roles as a neuroprotective factor and in promoting myelination, thereby supporting healthy brain aging. Here we summarize what is known about brain α-Klotho before focusing on the outstanding scientific questions related to its function. We believe there is a need for in vitro studies designed to distinguish between brain α-Klotho and other pools of α-Klotho, and for a greater understanding of the basic function of soluble α-Klotho. The mechanism by which the human KL-VS variant affects cognition also requires further elucidation. To help address these questions we suggest some experimental approaches that other laboratories might consider. In short, we hope to stimulate fresh ideas and encourage new research approaches that will allow the importance of α-Klotho for the aging brain to become clear.
Collapse
|
43
|
Morris ME, Rodriguez-Cruz V, Felmlee MA. SLC and ABC Transporters: Expression, Localization, and Species Differences at the Blood-Brain and the Blood-Cerebrospinal Fluid Barriers. AAPS JOURNAL 2017; 19:1317-1331. [PMID: 28664465 DOI: 10.1208/s12248-017-0110-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/05/2017] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) separate the brain and cerebrospinal fluid (CSF) from the systemic circulation and represent a barrier to the uptake of both endogenous compounds and xenobiotics into the brain. For compounds whose passive diffusion is limited due to their ionization or hydrophilicity, membrane transporters can facilitate their uptake across the BBB or BCSFB. Members of the solute carrier (SLC) and ATP-binding case (ABC) families are present on these barriers. Differences exist in the localization and expression of transport proteins between the BBB and BCSFB, resulting in functional differences in transport properties. This review focuses on the expression, membrane localization, and different isoforms present at each barrier. Diseases that affect the central nervous system including brain tumors, HIV, Alzheimer's disease, Parkinson's disease, and stroke affect the integrity and expression of transporters at the BBB and BCSFB and will be briefly reviewed.
Collapse
Affiliation(s)
- Marilyn E Morris
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA.
| | - Vivian Rodriguez-Cruz
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA
| | - Melanie A Felmlee
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 3601 Pacific Ave, Stockton, California, 95211, USA
| |
Collapse
|
44
|
Kim S, Hwang Y, Lee D, Webster MJ. Transcriptome sequencing of the choroid plexus in schizophrenia. Transl Psychiatry 2016; 6:e964. [PMID: 27898074 PMCID: PMC5290353 DOI: 10.1038/tp.2016.229] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 09/19/2015] [Accepted: 09/28/2016] [Indexed: 01/11/2023] Open
Abstract
The choroid plexus (CP) has a key role in maintaining brain homeostasis by producing cerebrospinal fluid (CSF), by mediating transport of nutrients and removing metabolic products from the central nervous system and by responding to peripheral inflammatory signals. Although abnormal markers of immune response and inflammation are apparent in individuals with schizophrenia, the CP of these individuals has not been characterized. We therefore sequenced mRNA from the CP from two independent collections of individuals with schizophrenia and unaffected controls. Genes related to immune function and inflammation were upregulated in both collections. In addition, a co-expression module related to immune/inflammation response that was generated by combining mRNA-Seq data from both collections was significantly associated with disease status. The immune/inflammation-related co-expression module was positively correlated with levels of C-reactive protein (CRP), cortisol and several immune modulator proteins in the serum of the same individuals and was also positively correlated with CRP, cortisol and pro-inflammatory cytokines in the frontal cortex of the same individuals. In addition, we found a substantial number of nodes (genes) that were common to our schizophrenia-associated immune/inflammation module from the pooled data and a module we generated from lippopolysaccharides-treated mouse model data. These results suggest that the CP of individuals with schizophrenia are responding to signals from the periphery by upregulating immune/inflammation-related genes to protect the brain and maintain the homeostasis but nevertheless fails to completely prevent immune/inflammation related changes in the brain.
Collapse
Affiliation(s)
- S Kim
- Stanley Brain Research Laboratory, Stanley Medical Research Institute, Rockville, MD, USA
| | - Y Hwang
- Department of Bio and Brain Engineering, KAIST, Daejeon, Korea
| | - D Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, Korea,Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Korea E-mail:
| | - M J Webster
- Stanley Brain Research Laboratory, Stanley Medical Research Institute, Rockville, MD, USA,Stanley Brain Research Laboratory, Stanley Medical Research Institute, 9800 Medical Center Drive, Suite C-050, Rockville, MD 20850, USA. E-mail:
| |
Collapse
|
45
|
Ellidag HY, Yilmaz N, Kurtulus F, Aydin O, Eren E, Inci A, Dolu S, Ince FDA, Giray Ö, Yaman A. The Three Sisters of Fate in Multiple Sclerosis: Klotho (Clotho), Fibroblast Growth Factor-23 (Lachesis), and Vitamin D (Atropos). Ann Neurosci 2016; 23:155-161. [PMID: 27721584 DOI: 10.1159/000449181] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 05/19/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The klotho (Klt)-fibroblast growth factor-23 (FGF-23)-vitamin D axis is the main component of calcium (Ca) and phosphorus (P) metabolisms; on the contrary, it is also secreted from the choroid plexus (CP). PURPOSE This study is aimed at evaluating serum soluble Klt (sKlt), FGF-23, and 25-(OH)-vitamin D levels in multiple sclerosis (MS) patients. METHODS Thirty-two relapsing-remitting MS patients (11 males and 21 females; mean age 38.3 years) and 31 age-sex matched healthy controls (12 males and 19 females; median age 38.5 years) were included in this study. All patients were diagnosed with MS according to the criteria of McDonald. RESULTS Serum sKlt, FGF-23, and P levels were significantly higher in MS patients compared to the control group (p < 0.01, p < 0.01, and p = 0.02, respectively). Serum 25-(OH)-vitamin D and Ca levels were significantly lower in MS patients (p < 0.01 and p = 0.04, respectively). CONCLUSION Klt, which is secreted from CP, could be a response to the inflammatory condition in MS. Elevated FGF-23 levels suppress 1α-hydroxylase and upregulates 24α-hydroxylase, which results in a decrease in 1,25-(OH)2D3 levels. Thus, the neuroprotective and immunomodulatory effects of vitamin D might not be seen in MS patients.
Collapse
Affiliation(s)
- Hamit Yasar Ellidag
- Central Laboratories, Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| | - Necat Yilmaz
- Central Laboratories, Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| | - Fatma Kurtulus
- Neurology Clinic, Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| | - Ozgur Aydin
- Biochemistry Laboratory, Maternity and Children's Hospital, Batman, Turkey
| | - Esin Eren
- Central Laboratories, Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| | - Ayca Inci
- Nephrology Clinic, Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| | - Suleyman Dolu
- Nephrology Clinic, Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| | - Fatma Demet Arslan Ince
- Department of Biochemistry, Tepecik Education and Research Hospital of Ministry of Health, Izmir, Turkey
| | - Özlem Giray
- Central Laboratories, Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| | - Aylin Yaman
- Neurology Clinic, Antalya Education and Research Hospital of Ministry of Health, Antalya, Turkey
| |
Collapse
|
46
|
Klotho Gene and Selective Serotonin Reuptake Inhibitors: Response to Treatment in Late-Life Major Depressive Disorder. Mol Neurobiol 2016; 54:1340-1351. [PMID: 26843110 DOI: 10.1007/s12035-016-9711-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 01/11/2016] [Indexed: 02/07/2023]
Abstract
Klotho protein, encoded by the Klotho gene (KL) at locus 13q12, is an antiaging hormone-like protein playing a pivotal role in cell metabolism homeostasis and associated to longevity and age-related diseases. In particular, altered cell metabolism in central nervous system may influence the behavior of serotoninergic neurons. The role of KL in the response to treatment with selective serotonin reuptake inhibitors (SSRIs) in late-life depressive syndromes and late-life major depressive disorder (MDD) is unclear. We genotyped three single-nucleotide polymorphisms (SNPs) of KL in 329 older patients with diagnosis of late-life MDD, treated with SSRIs and evaluated with the Hamilton Rating Scale for Depression 21-items (HRSD-21) at baseline and after 6 months. A reduction ≥50 and <10 % in HDRS-21 score was considered as response or nonresponse to therapy, respectively, and the values of reduction between 10 and 49 % as poor responders. After 6 months of SSRI treatment, 176 patients responded, 54 patients did not respond and 99 patients showed a poor response. Ordinal logistic models showed a significant association between mutation of SNP rs1207568 and responders and, similarly, for each unitary risk allele increase overlapping results were found. Conversely, a significantly higher frequency of the minor genotype of SNP rs9536314 was found in nonresponders. Considering the pre-post differences of HRSD-21 scores as a continue variable, we confirmed a significant improvement of depressive symptoms after treatment in patients carrying at least one minor allele at rs1207568 and a worse response in patients homozygous for the minor allele at rs9536314. Our results were the first that suggested a possible role of KL in the complex pathway of SSRI response in late-life MDD.
Collapse
|
47
|
The Effect of Acute and Chronic Social Stress on the Hippocampal Transcriptome in Mice. PLoS One 2015; 10:e0142195. [PMID: 26556046 PMCID: PMC4640871 DOI: 10.1371/journal.pone.0142195] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 10/19/2015] [Indexed: 12/19/2022] Open
Abstract
Psychogenic stress contributes to the formation of brain pathology. Using gene expression microarrays, we analyzed the hippocampal transcriptome of mice subjected to acute and chronic social stress of different duration. The longest period of social stress altered the expression of the highest number of genes and most of the stress-induced changes in transcription were reversible after 5 days of rest. Chronic stress affected genes involved in the functioning of the vascular system (Alas2, Hbb-b1, Hba-a2, Hba-a1), injury response (Vwf, Mgp, Cfh, Fbln5, Col3a1, Ctgf) and inflammation (S100a8, S100a9, Ctla2a, Ctla2b, Lcn2, Lrg1, Rsad2, Isg20). The results suggest that stress may affect brain functions through the stress-induced dysfunction of the vascular system. An important issue raised in our work is also the risk of the contamination of brain tissue samples with choroid plexus. Such contamination would result in a consistent up- or down-regulation of genes, such as Ttr, Igf2, Igfbp2, Prlr, Enpp2, Sostdc1, 1500015O10RIK (Ecrg4), Kl, Clic6, Kcne2, F5, Slc4a5, and Aqp1. Our study suggests that some of the previously reported, supposedly specific changes in hippocampal gene expression, may be a result of the inclusion of choroid plexus in the hippocampal samples.
Collapse
|
48
|
Indomethacin induced gene regulation in the rat hippocampus. Mol Brain 2015; 8:59. [PMID: 26438564 PMCID: PMC4595115 DOI: 10.1186/s13041-015-0150-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/30/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Non-steroidal anti-inflammatory drugs such as indomethacin are widely used to treat inflammatory diseases and manage pain, fever and inflammation in several conditions, including neuropsychiatric disorders. Although they predominantly function by inhibiting cyclooxygenase (COX) activity, important COX-independent actions also occur. These actions could be responsible for the adverse side effects associated with chronic and/or high dose usage of this popular drug class. RESULTS We examined gene regulation in the hippocampus after peripheral administration of indomethacin by employing a microarray approach. Secondary confirmation and the brain expression pattern of regulated genes was examined by in situ hybridization and immunohistochemistry. Transglutaminase 2, serum glucocorticoid inducible kinase, Inhibitor of NF-kappa B and vascular endothelial growth factor were among genes that were prominently upregulated, while G-protein coupled receptor 56 and neuropeptide Y were among genes that were downregulated by indomethacin. Co-localization studies using blood vessel markers revealed that transglutaminase 2 was induced specifically in brain vasculature. CONCLUSIONS The data demonstrate that COX-inhibitors can differentially regulate gene transcription in multiple, functionally distinctly cell types in the brain. The results provide additional insight into the molecular actions of COX-inhibitors and indicate that their effects on vasculature could influence cerebral blood flow mechanisms.
Collapse
|
49
|
Lavagnino L, Amianto F, Mwangi B, D'Agata F, Spalatro A, Zunta-Soares GB, Abbate Daga G, Mortara P, Fassino S, Soares JC. Identifying neuroanatomical signatures of anorexia nervosa: a multivariate machine learning approach. Psychol Med 2015; 45:2805-2812. [PMID: 25990697 DOI: 10.1017/s0033291715000768] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND There are currently no neuroanatomical biomarkers of anorexia nervosa (AN) available to make clinical inferences at an individual subject level. We present results of a multivariate machine learning (ML) approach utilizing structural neuroanatomical scan data to differentiate AN patients from matched healthy controls at an individual subject level. METHOD Structural neuroimaging scans were acquired from 15 female patients with AN (age = 20, s.d. = 4 years) and 15 demographically matched female controls (age = 22, s.d. = 3 years). Neuroanatomical volumes were extracted using the FreeSurfer software and input into the Least Absolute Shrinkage and Selection Operator (LASSO) multivariate ML algorithm. LASSO was 'trained' to identify 'novel' individual subjects as either AN patients or healthy controls. Furthermore, the model estimated the probability that an individual subject belonged to the AN group based on an individual scan. RESULTS The model correctly predicted 25 out of 30 subjects, translating into 83.3% accuracy (sensitivity 86.7%, specificity 80.0%) (p < 0.001; χ 2 test). Six neuroanatomical regions (cerebellum white matter, choroid plexus, putamen, accumbens, the diencephalon and the third ventricle) were found to be relevant in distinguishing individual AN patients from healthy controls. The predicted probabilities showed a linear relationship with drive for thinness clinical scores (r = 0.52, p < 0.005) and with body mass index (BMI) (r = -0.45, p = 0.01). CONCLUSIONS The model achieved a good predictive accuracy and drive for thinness showed a strong neuroanatomical signature. These results indicate that neuroimaging scans coupled with ML techniques have the potential to provide information at an individual subject level that might be relevant to clinical outcomes.
Collapse
Affiliation(s)
- L Lavagnino
- UT Center of Excellence on Mood Disorders,Department of Psychiatry and Behavioral Sciences,UT Houston Medical School,Houston,TX,USA
| | - F Amianto
- Department of Neuroscience,AOU San Giovanni Battista,Turin,Italy
| | - B Mwangi
- UT Center of Excellence on Mood Disorders,Department of Psychiatry and Behavioral Sciences,UT Houston Medical School,Houston,TX,USA
| | - F D'Agata
- Department of Neuroscience,AOU San Giovanni Battista,Turin,Italy
| | - A Spalatro
- Department of Neuroscience,AOU San Giovanni Battista,Turin,Italy
| | - G B Zunta-Soares
- UT Center of Excellence on Mood Disorders,Department of Psychiatry and Behavioral Sciences,UT Houston Medical School,Houston,TX,USA
| | - G Abbate Daga
- Department of Neuroscience,AOU San Giovanni Battista,Turin,Italy
| | - P Mortara
- Department of Neuroscience,AOU San Giovanni Battista,Turin,Italy
| | - S Fassino
- Department of Neuroscience,AOU San Giovanni Battista,Turin,Italy
| | - J C Soares
- UT Center of Excellence on Mood Disorders,Department of Psychiatry and Behavioral Sciences,UT Houston Medical School,Houston,TX,USA
| |
Collapse
|
50
|
Haley SA, O'Hara BA, Nelson CDS, Brittingham FLP, Henriksen KJ, Stopa EG, Atwood WJ. Human polyomavirus receptor distribution in brain parenchyma contrasts with receptor distribution in kidney and choroid plexus. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2246-58. [PMID: 26056932 DOI: 10.1016/j.ajpath.2015.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/21/2015] [Accepted: 04/14/2015] [Indexed: 11/25/2022]
Abstract
The human polyomavirus, JCPyV, is the causative agent of progressive multifocal leukoencephalopathy, a rare demyelinating disease that occurs in the setting of prolonged immunosuppression. After initial asymptomatic infection, the virus establishes lifelong persistence in the kidney and possibly other extraneural sites. In rare instances, the virus traffics to the central nervous system, where oligodendrocytes, astrocytes, and glial precursors are susceptible to lytic infection, resulting in progressive multifocal leukoencephalopathy. The mechanisms by which the virus traffics to the central nervous system from peripheral sites remain unknown. Lactoseries tetrasaccharide c (LSTc), a pentasaccharide containing a terminal α2,6-linked sialic acid, is the major attachment receptor for polyomavirus. In addition to LSTc, type 2 serotonin receptors are required for facilitating virus entry into susceptible cells. We studied the distribution of virus receptors in kidney and brain using lectins, antibodies, and labeled virus. The distribution of LSTc, serotonin receptors, and virus binding sites overlapped in kidney and in the choroid plexus. In brain parenchyma, serotonin receptors were expressed on oligodendrocytes and astrocytes, but these cells were negative for LSTc and did not bind virus. LSTc was instead found on microglia and vascular endothelium, to which virus bound abundantly. Receptor distribution was not changed in the brains of patients with progressive multifocal leukoencephalopathy. Virus infection of oligodendrocytes and astrocytes during disease progression is LSTc independent.
Collapse
Affiliation(s)
- Sheila A Haley
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Bethany A O'Hara
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Christian D S Nelson
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Frances L P Brittingham
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Kammi J Henriksen
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Edward G Stopa
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Walter J Atwood
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island.
| |
Collapse
|