1
|
Anderson AG, Moyers BA, Loupe JM, Rodriguez-Nunez I, Felker SA, Lawlor JMJ, Bunney WE, Bunney BG, Cartagena PM, Sequeira A, Watson SJ, Akil H, Mendenhall EM, Cooper GM, Myers RM. Allele-specific transcription factor binding across human brain regions offers mechanistic insight into eQTLs. Genome Res 2024; 34:1224-1234. [PMID: 39152038 PMCID: PMC11444172 DOI: 10.1101/gr.278601.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
Transcription factors (TFs) regulate gene expression by facilitating or disrupting the formation of transcription initiation machinery at particular genomic loci. Because TF occupancy is driven in part by recognition of DNA sequence, genetic variation can influence TF-DNA associations and gene regulation. To identify variants that impact TF binding in human brain tissues, we assessed allele-specific binding (ASB) at heterozygous variants for 94 TFs in nine brain regions from two donors. Leveraging graph genomes constructed from phased genomic sequence data, we compared ChIP-seq signals between alleles at heterozygous variants within each brain region and identified thousands of variants exhibiting ASB for at least one TF. ASB reproducibility was measured by comparisons between independent experiments both within and between donors. We found that rare alleles in the general population more frequently led to reduced TF binding, whereas common alleles had an equal likelihood of increasing or decreasing binding. Further, for ASB variants in predicted binding motifs, the favored allele tended to be the one with the stronger expected motif match, but this concordance was not observed within highly occupied sites. We also found that neuron-specific cis-regulatory elements (cCREs), in contrast with oligodendrocyte-specific cCREs, showed depletion of ASB variants. We identified 2670 ASB variants associated with evidence for allele-specific gene expression in the brain from GTEx data and observed increasing eQTL effect direction concordance as ASB significance increases. These results provide a valuable and unique resource for mechanistic analysis of cis-regulatory variation in human brain tissue.
Collapse
Affiliation(s)
- Ashlyn G Anderson
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
- University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Belle A Moyers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | - Jacob M Loupe
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | | | | | - James M J Lawlor
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | - William E Bunney
- Department of Psychiatry and Human Behavior, University of California, Irvine, California 92697, USA
| | - Blynn G Bunney
- Department of Psychiatry and Human Behavior, University of California, Irvine, California 92697, USA
| | - Preston M Cartagena
- Department of Psychiatry and Human Behavior, University of California, Irvine, California 92697, USA
| | - Adolfo Sequeira
- Department of Psychiatry and Human Behavior, University of California, Irvine, California 92697, USA
| | - Stanley J Watson
- The Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Huda Akil
- The Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Eric M Mendenhall
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | - Gregory M Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA;
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA;
| |
Collapse
|
2
|
Hao M, Qin Y, Li Y, Tang Y, Ma Z, Tan J, Jin L, Wang F, Gong X. Metabolome subtyping reveals multi-omics characteristics and biological heterogeneity in major psychiatric disorders. Psychiatry Res 2023; 330:115605. [PMID: 38006718 DOI: 10.1016/j.psychres.2023.115605] [Citation(s) in RCA: 1] [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: 08/26/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/27/2023]
Abstract
Growing evidence suggests that major psychiatric disorders (MPDs) share common etiologies and pathological processes. However, the diagnosis is currently based on descriptive symptoms, which ignores the underlying pathogenesis and hinders the development of clinical treatments. This highlights the urgency of characterizing molecular biomarkers and establishing objective diagnoses of MPDs. Here, we collected untargeted metabolomics, proteomics and DNA methylation data of 327 patients with MPDs, 131 individuals with genetic high risk and 146 healthy controls to explore the multi-omics characteristics of MPDs. First, differential metabolites (DMs) were identified and we classified MPD patients into 3 subtypes based on DMs. The subtypes showed distinct metabolomics, proteomics and DNA methylation signatures. Specifically, one subtype showed dysregulation of complement and coagulation proteins, while the DNA methylation showed abnormalities in chemical synapses and autophagy. Integrative analysis in metabolic pathways identified the important roles of the citrate cycle, sphingolipid metabolism and amino acid metabolism. Finally, we constructed prediction models based on the metabolites and proteomics that successfully captured the risks of MPD patients. Our study established molecular subtypes of MPDs and elucidated their biological heterogeneity through a multi-omics investigation. These results facilitate the understanding of pathological mechanisms and promote the diagnosis and prevention of MPDs.
Collapse
Affiliation(s)
- Meng Hao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China; Zhangjiang Fudan International Innovation Center, Fudan Zhangjiang Institute, Obstetrics and Gynecology Hospital, Human Phenome Institute, Fudan University, China
| | - Yue Qin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China; Zhangjiang Fudan International Innovation Center, Fudan Zhangjiang Institute, Obstetrics and Gynecology Hospital, Human Phenome Institute, Fudan University, China
| | - Yi Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China; Zhangjiang Fudan International Innovation Center, Fudan Zhangjiang Institute, Obstetrics and Gynecology Hospital, Human Phenome Institute, Fudan University, China; International Human Phenome Institutes, Shanghai, China
| | - Yanqing Tang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zehan Ma
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Jingze Tan
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China; Zhangjiang Fudan International Innovation Center, Fudan Zhangjiang Institute, Obstetrics and Gynecology Hospital, Human Phenome Institute, Fudan University, China; International Human Phenome Institutes, Shanghai, China
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China; Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China.
| | - Xiaohong Gong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Moyers BA, Loupe JM, Felker SA, Lawlor JM, Anderson AG, Rodriguez-Nunez I, Bunney WE, Bunney BG, Cartagena PM, Sequeira A, Watson SJ, Akil H, Mendenhall EM, Cooper GM, Myers RM. Allele biased transcription factor binding across human brain regions gives mechanistic insight into eQTLs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.06.561245. [PMID: 37873117 PMCID: PMC10592666 DOI: 10.1101/2023.10.06.561245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Transcription Factors (TFs) influence gene expression by facilitating or disrupting the formation of transcription initiation machinery at particular genomic loci. Because genomic localization of TFs is in part driven by TF recognition of DNA sequence, variation in TF binding sites can disrupt TF-DNA associations and affect gene regulation. To identify variants that impact TF binding in human brain tissues, we quantified allele bias for 93 TFs analyzed with ChIP-seq experiments of multiple structural brain regions from two donors. Using graph genomes constructed from phased genomic sequence data, we compared ChIP-seq signal between alleles at heterozygous variants within each tissue sample from each donor. Comparison of results from different brain regions within donors and the same regions between donors provided measures of allele bias reproducibility. We identified thousands of DNA variants that show reproducible bias in ChIP-seq for at least one TF. We found that alleles that are rarer in the general population were more likely than common alleles to exhibit large biases, and more frequently led to reduced TF binding. Combining ChIP-seq with RNA-seq, we identified TF-allele interaction biases with RNA bias in a phased allele linked to 6,709 eQTL variants identified in GTEx data, 3,309 of which were found in neural contexts. Our results provide insights into the effects of both common and rare variation on gene regulation in the brain. These findings can facilitate mechanistic understanding of cis-regulatory variation associated with biological traits, including disease.
Collapse
Affiliation(s)
| | - Jacob M. Loupe
- HudsonAlpha Institute for Biotechnology, Huntsville AL, USA
| | | | | | | | | | - William E. Bunney
- Department of Psychiatry and Human Behavior, University of California, Irvine CA, USA
| | - Blynn G. Bunney
- Department of Psychiatry and Human Behavior, University of California, Irvine CA, USA
| | - Preston M. Cartagena
- Department of Psychiatry and Human Behavior, University of California, Irvine CA, USA
| | - Adolfo Sequeira
- Department of Psychiatry and Human Behavior, University of California, Irvine CA, USA
| | - Stanley J. Watson
- The Michigan Neuroscience Institute, University of Michigan, Ann Arbor MI, USA
| | - Huda Akil
- The Michigan Neuroscience Institute, University of Michigan, Ann Arbor MI, USA
| | | | | | | |
Collapse
|
4
|
Palma-Lara I, García Alonso-Themann P, Pérez-Durán J, Godínez-Aguilar R, Bonilla-Delgado J, Gómez-Archila D, Espinosa-García AM, Nolasco-Quiroga M, Victoria-Acosta G, López-Ornelas A, Serrano-Bello JC, Olguín-García MG, Palacios-Reyes C. Potential Role of Protein Kinase FAM20C on the Brain in Raine Syndrome, an In Silico Analysis. Int J Mol Sci 2023; 24:ijms24108904. [PMID: 37240249 DOI: 10.3390/ijms24108904] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
FAM20C (family with sequence similarity 20, member C) is a serine/threonine-specific protein kinase that is ubiquitously expressed and mainly associated with biomineralization and phosphatemia regulation. It is mostly known due to pathogenic variants causing its deficiency, which results in Raine syndrome (RNS), a sclerosing bone dysplasia with hypophosphatemia. The phenotype is recognized by the skeletal features, which are related to hypophosphorylation of different FAM20C bone-target proteins. However, FAM20C has many targets, including brain proteins and the cerebrospinal fluid phosphoproteome. Individuals with RNS can have developmental delay, intellectual disability, seizures, and structural brain defects, but little is known about FAM20C brain-target-protein dysregulation or about a potential pathogenesis associated with neurologic features. In order to identify the potential FAM20C actions on the brain, an in silico analysis was conducted. Structural and functional defects reported in RNS were described; FAM20C targets and interactors were identified, including their brain expression. Gene ontology of molecular processes, function, and components was completed for these targets, as well as for potential involved signaling pathways and diseases. The BioGRID and Human Protein Atlas databases, the Gorilla tool, and the PANTHER and DisGeNET databases were used. Results show that genes with high expression in the brain are involved in cholesterol and lipoprotein processes, plus axo-dendritic transport and the neuron part. These results could highlight some proteins involved in the neurologic pathogenesis of RNS.
Collapse
Affiliation(s)
- Icela Palma-Lara
- Laboratorio de Morfología Celular y Molecular, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | | | - Javier Pérez-Durán
- Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de México 11000, Mexico
| | | | - José Bonilla-Delgado
- Unidad de Investigación, Hospital Regional de Ixtapaluca, Ixtapaluca 56530, Mexico
- Departamento de Biotecnología, Escuela de Ingeniería y Ciencias, Instituto Tecnológico de Monterrey, Toluca de Lerdo 50110, Mexico
| | - Damián Gómez-Archila
- Departamento de Oncología Quirúrgica, Hospital de Gineco-Obstetricia 3, Centro Médico Nacional "La Raza", Ciudad de México 02990, Mexico
| | | | - Manuel Nolasco-Quiroga
- Coordinación de Enseñanza e Investigación, Clínica Hospital Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Huauchinango 73177, Mexico
| | | | - Adolfo López-Ornelas
- División de Investigación, Hospital Juárez de México, Ciudad de México 11340, Mexico
| | - Juan Carlos Serrano-Bello
- Departamento de Patología Clínica y Experimental, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico
| | | | - Carmen Palacios-Reyes
- División de Investigación, Hospital Juárez de México, Ciudad de México 11340, Mexico
| |
Collapse
|
5
|
Arteriosclerosis Derived from Cutaneous Inflammation Is Ameliorated by the Deletion of IL-17A and IL-17F. Int J Mol Sci 2023; 24:ijms24065434. [PMID: 36982506 PMCID: PMC10049365 DOI: 10.3390/ijms24065434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/22/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
The skin is one of the major immune organs producing large amounts of proinflammatory and inflammatory cytokines in response to internal or exogenous stimuli, inducing systemic inflammation in various internal organs. In recent years, organ damage associated with inflammatory skin diseases such as psoriasis and atopic dermatitis has received increasing attention, and vascular disorder such as arteriosclerosis is one of the serious complications of chronic inflammatory skin diseases. However, the detailed mechanism of arteriosclerosis in dermatitis and the role of cytokines have not been clarified so far. In the current study, using a spontaneous dermatitis model, we investigated the pathophysiology of arteriosclerosis and the treatment option for inflammatory skin conditions. We employed spontaneous dermatitis model mice overexpressing human caspase-1 in the epidermal keratinocyte (Kcasp1Tg). The thoracic and abdominal aorta was investigated histologically. GeneChip and RT-PCR analysis were performed to measure the changes in mRNA levels in the aorta. To elucidate the direct effect on the artery by major inflammatory cytokines, endothelial cells, vascular smooth muscle cells, and fibroblast cells were co-cultured with several cytokines, and mRNA expression levels were measured. In order to observe the efficacy of IL-17A/F in arteriosclerosis, cross-mating with IL-17A, IL-17F, and IL-17A/F deficient mice was performed. Finally, we also measured snap tension in the abdominal aorta in WT, Kcasp1Tg, and IL17A/F-deficient mice. Kcasp1Tg showed a decrease in the diameter of the abdominal aorta compared to wild-type mice. mRNA levels for six genes including Apol11b, Camp, Chil3, S100a8, S100a9, and Spta1 were increased in the abdominal aorta of Kcasp1Tg. Some of the above mRNA levels were also increased in the co-culture with major inflammatory cytokines, IL-17A/F, IL-1β, and TNF-α. Dermatitis improved and mRNA levels were partially ameliorated in Kcasp1Tg with IL-17A/F deletion. Arterial fragility was also evidenced in the inflammatory model, but arterial flexibility was revealed in the IL-17A/F deletion model. Severe dermatitis is closely related to secondary arteriosclerosis caused by the persistent release of inflammatory cytokines. The results also proved that treatment against IL-17A and F may ameliorate arteriosclerosis.
Collapse
|
6
|
Singh M, Agarwal V, Jindal D, Pancham P, Agarwal S, Mani S, Tiwari RK, Das K, Alghamdi BS, Abujamel TS, Ashraf GM, Jha SK. Recent Updates on Corticosteroid-Induced Neuropsychiatric Disorders and Theranostic Advancements through Gene Editing Tools. Diagnostics (Basel) 2023; 13:diagnostics13030337. [PMID: 36766442 PMCID: PMC9914305 DOI: 10.3390/diagnostics13030337] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/28/2022] [Accepted: 10/16/2022] [Indexed: 01/19/2023] Open
Abstract
The vast use of corticosteroids (CCSs) globally has led to an increase in CCS-induced neuropsychiatric disorders (NPDs), a very common manifestation in patients after CCS consumption. These neuropsychiatric disorders range from depression, insomnia, and bipolar disorders to panic attacks, overt psychosis, and many other cognitive changes in such subjects. Though their therapeutic importance in treating and improving many clinical symptoms overrides the complications that arise after their consumption, still, there has been an alarming rise in NPD cases in recent years, and they are seen as the greatest public health challenge globally; therefore, these potential side effects cannot be ignored. It has also been observed that many of the neuronal functional activities are regulated and controlled by genomic variants with epigenetic factors (DNA methylation, non-coding RNA, and histone modeling, etc.), and any alterations in these regulatory mechanisms affect normal cerebral development and functioning. This study explores a general overview of emerging concerns of CCS-induced NPDs, the effective molecular biology approaches that can revitalize NPD therapy in an extremely specialized, reliable, and effective manner, and the possible gene-editing-based therapeutic strategies to either prevent or cure NPDs in the future.
Collapse
Affiliation(s)
- Manisha Singh
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201309, India
- Correspondence: (M.S.); (S.K.J.)
| | - Vinayak Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201309, India
| | - Divya Jindal
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201309, India
| | - Pranav Pancham
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201309, India
| | - Shriya Agarwal
- Department of Molecular Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Shalini Mani
- Department of Biotechnology, Jaypee Institute of Information Technology (JIIT), Noida 201309, India
| | - Raj Kumar Tiwari
- School of Health Sciences, Pharmaceutical Sciences, UPES, Dehradun 248007, India
| | - Koushik Das
- School of Health Sciences, Pharmaceutical Sciences, UPES, Dehradun 248007, India
| | - Badrah S. Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Tukri S. Abujamel
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ghulam Md. Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida 201310, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India
- Correspondence: (M.S.); (S.K.J.)
| |
Collapse
|
7
|
Cheng B, Yang X, Cheng S, Li C, Zhang H, Liu L, Meng P, Jia Y, Wen Y, Zhang F. A large-scale polygenic risk score analysis identified candidate proteins associated with anxiety, depression and neuroticism. Mol Brain 2022; 15:66. [PMID: 35870967 PMCID: PMC9308259 DOI: 10.1186/s13041-022-00954-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/09/2022] [Indexed: 11/10/2022] Open
Abstract
Psychiatric disorders and neuroticism are closely associated with central nervous system, whose proper functioning depends on efficient protein renewal. This study aims to systematically analyze the association between anxiety / depression / neuroticism and each of the 439 proteins. 47,536 pQTLs of 439 proteins in brain, plasma and cerebrospinal fluid (CSF) were collected from recent genome-wide association study. Polygenic risk scores (PRS) of the 439 proteins were then calculated using the UK Biobank cohort, including 120,729 subjects of neuroticism, 255,354 subjects of anxiety and 316,513 subjects of depression. Pearson correlation analyses were performed to evaluate the correlation between each protein and each of the mental traits by using calculated PRSs as the instrumental variables of protein. In general population, six correlations were identified in plasma and CSF such as plasma protease C1 inhibitor (C1-INH) with neuroticism score (r = - 0.011, P = 2.56 × 10- 9) in plasma, C1-INH with neuroticism score (r = -0.010, P = 3.09 × 10- 8) in CSF, and ERBB1 with self-reported depression (r = - 0.012, P = 4.65 × 10- 5) in CSF. C1-INH and ERBB1 may induce neuroticism and depression by affecting brain function and synaptic development. Gender subgroup analyses found that BST1 was correlated with neuroticism score in male CSF (r = - 0.011, P = 1.80 × 10- 5), while CNTN2 was correlated with depression score in female brain (r = - 0.013, P = 6.43 × 10- 4). BST1 and CNTN2 may be involved in nervous system metabolism and brain health. Six common candidate proteins were associated with all three traits (P < 0.05) and were confirmed in relevant proteomic studies, such as C1-INH in plasma, CNTN2 and MSP in the brain. Our results provide novel clues for revealing the roles of proteins in the development of anxiety, depression and neuroticism.
Collapse
Affiliation(s)
- Bolun Cheng
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 76 Yan Ta West Road, 710061, Xi'an, People's Republic of China.,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, People's Republic of China
| | - Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 76 Yan Ta West Road, 710061, Xi'an, People's Republic of China.,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, People's Republic of China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 76 Yan Ta West Road, 710061, Xi'an, People's Republic of China.,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, People's Republic of China
| | - Chun'e Li
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 76 Yan Ta West Road, 710061, Xi'an, People's Republic of China.,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, People's Republic of China
| | - Huijie Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 76 Yan Ta West Road, 710061, Xi'an, People's Republic of China.,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, People's Republic of China
| | - Li Liu
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 76 Yan Ta West Road, 710061, Xi'an, People's Republic of China.,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, People's Republic of China
| | - Peilin Meng
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 76 Yan Ta West Road, 710061, Xi'an, People's Republic of China.,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, People's Republic of China
| | - Yumeng Jia
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 76 Yan Ta West Road, 710061, Xi'an, People's Republic of China.,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, People's Republic of China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 76 Yan Ta West Road, 710061, Xi'an, People's Republic of China.,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, People's Republic of China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, 76 Yan Ta West Road, 710061, Xi'an, People's Republic of China. .,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, People's Republic of China.
| |
Collapse
|
8
|
Hok-A-Hin YS, Dijkstra AA, Rábano A, Hoozemans JJ, Castillo L, Seelaar H, van Swieten JC, Pijnenburg YAL, Teunissen CE, Del Campo M. Apolipoprotein L1 is increased in frontotemporal lobar degeneration post-mortem brain but not in ante-mortem cerebrospinal fluid. Neurobiol Dis 2022; 172:105813. [PMID: 35820647 DOI: 10.1016/j.nbd.2022.105813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022] Open
Abstract
AIMS Frontotemporal Dementia (FTD) is caused by frontal-temporal lobar degeneration (FTLD), characterized mainly by brain protein aggregates of tau (FTLD-Tau) or TDP-43 (FTLD-TDP). The clinicopathological heterogeneity makes ante-mortem diagnosis of these pathological subtypes challenging. Our proteomics study showed increased Apolipoprotein L1 (APOL1) levels in CSF from FTD patients, which was prominently expressed in FTLD-Tau. We aimed to understand APOL1 expression in FTLD post-mortem brain tissue and to validate its potential as a CSF biomarker for FTD and its pathological subtypes. METHODS APOL1 levels were analyzed in the frontal cortex of FTLD (including FTLD-Tau and FTLD-TDP) and non-demented controls by immunohistochemistry (FTLD total = 18 (12 FTLD-Tau and 6 FTLD-TDP); controls = 9), western blot (WB), and a novel prototype ELISA (FTLD total = 44 (21 FTLD-Tau and 23 FTLD-TDP); controls = 9). The association of APOL1 immunoreactivity with phosphorylated Tau (pTau) and TDP-43 (pTDP-43) immunoreactivity was assessed. CSF APOL1 was analyzed in confirmed FTD patients (n = 27, including 12 FTLD-Tau and 15 FTLD-TDP) and controls (n = 15) using the same ELISA. RESULTS APOL1 levels were significantly increased in FTLD post-mortem tissue compared to controls as measured by immunohistochemistry, WB, and ELISA. However, no differences between the pathological subtypes were observed. APOL1 immunoreactivity was present in neuronal and glial cells but did not co-localize with pTau or pTDP-43. CSF APOL1 levels were comparable between FTD patients and controls and between pathological subtypes. CONCLUSION APOL1 is upregulated in FTLD pathology irrespective of the subtypes, indicating a role of this novel protein in FTD pathophysiology. The APOL1 levels detected in brain tissue were not mirrored in the CSF, limiting its potential as a specific FTD biofluid-based biomarker using our current immunoassay.
Collapse
Affiliation(s)
- Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands.
| | - Anke A Dijkstra
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, the Netherlands
| | - Alberto Rábano
- CIEN Tissue Bank, Alzheimer's Centre Reina Sofía-CIEN Foundation, Madrid, Spain
| | - Jeroen J Hoozemans
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, the Netherlands
| | - Lucía Castillo
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands
| | - Harro Seelaar
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, the Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Centre Amsterdam and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands
| | - Marta Del Campo
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo- CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
9
|
Laudanski K. Persistence of Lipoproteins and Cholesterol Alterations after Sepsis: Implication for Atherosclerosis Progression. Int J Mol Sci 2021; 22:ijms221910517. [PMID: 34638860 PMCID: PMC8508791 DOI: 10.3390/ijms221910517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Sepsis is one of the most common critical care illnesses with increasing survivorship. The quality of life in sepsis survivors is adversely affected by several co-morbidities, including increased incidence of dementia, stroke, cardiac disease and at least temporary deterioration in cognitive dysfunction. One of the potential explanations for their progression is the persistence of lipid profile abnormalities induced during acute sepsis into recovery, resulting in acceleration of atherosclerosis. (2) Methods: This is a targeted review of the abnormalities in the long-term lipid profile abnormalities after sepsis; (3) Results: There is a well-established body of evidence demonstrating acute alteration in lipid profile (HDL-c ↓↓, LDL-C -c ↓↓). In contrast, a limited number of studies demonstrated depression of HDL-c levels with a concomitant increase in LDL-C -c in the wake of sepsis. VLDL-C -c and Lp(a) remained unaltered in few studies as well. Apolipoprotein A1 was altered in survivors suggesting abnormalities in lipoprotein metabolism concomitant to overall lipoprotein abnormalities. However, most of the studies were limited to a four-month follow-up and patient groups were relatively small. Only one study looked at the atherosclerosis progression in sepsis survivors using clinical correlates, demonstrating an acceleration of plaque formation in the aorta, and a large metanalysis suggested an increase in the risk of stroke or acute coronary event between 3% to 9% in sepsis survivors. (4) Conclusions: The limited evidence suggests an emergence and persistence of the proatherogenic lipid profile in sepsis survivors that potentially contributes, along with other factors, to the clinical sequel of atherosclerosis.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA 19104, USA; ; Tel.: +1-215-662-8200
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Leonard Davis Institute of Healthcare Economics, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Lindholm Carlström E, Niazi A, Etemadikhah M, Halvardson J, Enroth S, Stockmeier CA, Rajkowska G, Nilsson B, Feuk L. Transcriptome Analysis of Post-Mortem Brain Tissue Reveals Up-Regulation of the Complement Cascade in a Subgroup of Schizophrenia Patients. Genes (Basel) 2021; 12:1242. [PMID: 34440415 PMCID: PMC8393670 DOI: 10.3390/genes12081242] [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: 07/05/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 01/23/2023] Open
Abstract
Schizophrenia is a genetically complex neuropsychiatric disorder with largely unresolved mechanisms of pathology. Identification of genes and pathways associated with schizophrenia is important for understanding the development, progression and treatment of schizophrenia. In this study, pathways associated with schizophrenia were explored at the level of gene expression. The study included post-mortem brain tissue samples from 68 schizophrenia patients and 44 age and sex-matched control subjects. Whole transcriptome poly-A selected paired-end RNA sequencing was performed on tissue from the prefrontal cortex and orbitofrontal cortex. RNA expression differences were detected between case and control individuals, focusing both on single genes and pathways. The results were validated with RT-qPCR. Significant differential expression between patient and controls groups was found for 71 genes. Gene ontology analysis of differentially expressed genes revealed an up-regulation of multiple genes in immune response among the patients (corrected p-value = 0.004). Several genes in the category belong to the complement system, including C1R, C1S, C7, FCN3, SERPING1, C4A and CFI. The increased complement expression is primarily driven by a subgroup of patients with increased expression of immune/inflammatory response genes, pointing to important differences in disease etiology within the patient group. Weighted gene co-expression network analysis highlighted networks associated with both synaptic transmission and activation of the immune response. Our results demonstrate the importance of immune-related pathways in schizophrenia and provide evidence for elevated expression of the complement cascade as an important pathway in schizophrenia pathology.
Collapse
Affiliation(s)
- Eva Lindholm Carlström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden; (E.L.C.); (A.N.); (M.E.); (J.H.); (S.E.); (B.N.)
| | - Adnan Niazi
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden; (E.L.C.); (A.N.); (M.E.); (J.H.); (S.E.); (B.N.)
| | - Mitra Etemadikhah
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden; (E.L.C.); (A.N.); (M.E.); (J.H.); (S.E.); (B.N.)
| | - Jonatan Halvardson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden; (E.L.C.); (A.N.); (M.E.); (J.H.); (S.E.); (B.N.)
| | - Stefan Enroth
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden; (E.L.C.); (A.N.); (M.E.); (J.H.); (S.E.); (B.N.)
| | - Craig A. Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; (C.A.S.); (G.R.)
| | - Grazyna Rajkowska
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; (C.A.S.); (G.R.)
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden; (E.L.C.); (A.N.); (M.E.); (J.H.); (S.E.); (B.N.)
| | - Lars Feuk
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden; (E.L.C.); (A.N.); (M.E.); (J.H.); (S.E.); (B.N.)
| |
Collapse
|
11
|
Pays E. The function of apolipoproteins L (APOLs): relevance for kidney disease, neurotransmission disorders, cancer and viral infection. FEBS J 2021; 288:360-381. [PMID: 32530132 PMCID: PMC7891394 DOI: 10.1111/febs.15444] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/24/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022]
Abstract
The discovery that apolipoprotein L1 (APOL1) is the trypanolytic factor of human serum raised interest about the function of APOLs, especially following the unexpected finding that in addition to their protective action against sleeping sickness, APOL1 C-terminal variants also cause kidney disease. Based on the analysis of the structure and trypanolytic activity of APOL1, it was proposed that APOLs could function as ion channels of intracellular membranes and be involved in mechanisms triggering programmed cell death. In this review, the recent finding that APOL1 and APOL3 inversely control the synthesis of phosphatidylinositol-4-phosphate (PI(4)P) by the Golgi PI(4)-kinase IIIB (PI4KB) is commented. APOL3 promotes Ca2+ -dependent activation of PI4KB, but due to their increased interaction with APOL3, APOL1 C-terminal variants can inactivate APOL3, leading to reduction of Golgi PI(4)P synthesis. The impact of APOLs on several pathological processes that depend on Golgi PI(4)P levels is discussed. I propose that through their effect on PI4KB activity, APOLs control not only actomyosin activities related to vesicular trafficking, but also the generation and elongation of autophagosomes induced by inflammation.
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular ParasitologyIBMMUniversité Libre de BruxellesGosseliesBelgium
| |
Collapse
|
12
|
Nawaz R, Gul S, Amin R, Huma T, Al Mughairbi F. Overview of schizophrenia research and treatment in Pakistan. Heliyon 2020; 6:e05545. [PMID: 33294688 PMCID: PMC7695967 DOI: 10.1016/j.heliyon.2020.e05545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/12/2020] [Accepted: 11/16/2020] [Indexed: 11/26/2022] Open
Abstract
Mental health is the most neglected health sector in Pakistan, and the majority of citizens have limited or no access to primary and secondary psychiatric services. The incidence of schizophrenia (SCZ) has increased at an alarming rate in Pakistan, relative to that of other psychiatric disorders. While numerous studies have investigated SCZ, few have addressed the issue about the Pakistani population. In the present review, the researchers discuss current data integral to the prevalence, pathophysiology, and molecular genetics of SCZ; treatment approaches to the disease; and patient responses to drugs prescribed for SCZ in Pakistan. Most Pakistani patients exhibit poor responses to antipsychotic drugs. Based on our review, the researchers hypothesize that genetic dissimilarities between Pakistani and Western populations contribute to such poor responses. Consequently, an understanding of such genetic differences and the provision of personalized treatment may simultaneously aid in improving SCZ treatment in Pakistan.
Collapse
Affiliation(s)
- Rukhsana Nawaz
- Department of Clinical Psychology, College of Medicine & Health Sciences, UAE University 15551 Al Ain, United Arab Emirates
| | - Saima Gul
- Department of Rehabilitation Science, Faculty of Pharmacy & Allied Health Sciences, Shifa Tameer e Millat University, Islamabad, Pakistan
| | - Rafat Amin
- Department of Pathology, Institute of Biological, Biochemical and Pharmaceutical Sciences, Dow University of Health Sciences, Ojha Campus, Karachi, Pakistan
| | | | - Fadwa Al Mughairbi
- Department of Clinical Psychology, College of Medicine & Health Sciences, UAE University 15551 Al Ain, United Arab Emirates
| |
Collapse
|
13
|
Guo L, Lin W, Zhang Y, Li W, Wang J. BEST: a web server for brain expression Spatio-temporal pattern analysis. BMC Bioinformatics 2019; 20:632. [PMID: 31805847 PMCID: PMC6896511 DOI: 10.1186/s12859-019-3222-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022] Open
Abstract
Background Dysregulated gene expression patterns have been reported in several mental disorders. Limited by the difficulty of obtaining samples, psychiatric molecular mechanism research still relies heavily on clues from genetics studies. By using reference data from brain expression studies, multiple types of comprehensive gene expression pattern analysis have been performed on psychiatric genetic results. These systems-level spatial-temporal expression pattern analyses provided evidence on specific brain regions, developmental stages and molecular pathways that are possibly involved in psychiatric pathophysiology. At present, there is no online tool for such systematic analysis, which hinders the applications of analysis by non-informatics researchers such as experimental biologists and clinical molecular biologists. Results We developed the BEST web server to support Brain Expression Spatio-Temporal pattern analysis. There are three highlighted features of BEST: 1) visualization: it generates user-friendly visual results that are easy to interpret, including heatmaps, Venn diagrams, gene co-expression networks and cluster-based Manhattan gene plots; these results illustrate the complex spatio-temporal expression patterns, including expression quantification and correlation between genes; 2) integration: it provides comprehensive human brain spatio-temporal expression patterns by integrating data from currently available databases; 3) multi-dimensionality: it analyses input genes as both a whole set and several subsets (clusters) which are enriched according to co-expression patterns, and it also presents the correlation between genetic and expression data. Conclusions To the best of our knowledge, BEST is the first data tool to support comprehensive human brain spatial-temporal expression pattern analysis. It helps to bridge disease-related genetic studies and mechanism studies, provides clues for key gene and molecular system identification, and supports the analysis of disease sensitive brain region and age stages. BEST is freely available at http://best.psych.ac.cn.
Collapse
Affiliation(s)
- Liyuan Guo
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 16 Lincui Road, Chaoyang District, Beijing, 100101, China.,Department of Psycholog, University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Lin
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 16 Lincui Road, Chaoyang District, Beijing, 100101, China.,Department of Psycholog, University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yidan Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 16 Lincui Road, Chaoyang District, Beijing, 100101, China.,Department of Psycholog, University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenhan Li
- Oumeng V medical Laboratory, Hangzhou, 310013, Zhejiang, China
| | - Jing Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 16 Lincui Road, Chaoyang District, Beijing, 100101, China. .,Department of Psycholog, University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
14
|
Bar-Yosef T, Damri O, Agam G. Dual Role of Autophagy in Diseases of the Central Nervous System. Front Cell Neurosci 2019; 13:196. [PMID: 31191249 PMCID: PMC6548059 DOI: 10.3389/fncel.2019.00196] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/18/2019] [Indexed: 12/14/2022] Open
Abstract
Autophagy is a vital lysosomal degradation and recycling pathway in the eukaryotic cell, responsible for maintaining an intricate balance between cell survival and cell death, necessary for neuronal survival and function. This dual role played by autophagy raises the question whether this process is a protective or a destructive pathway, the contributor of neuronal cell death or a failed attempt to repair aberrant processes? Deregulated autophagy at different steps of the pathway, whether excessive or downregulated, has been proposed to be associated with neurodegenerative disorders such as Alzheimer's-, Huntington's-, and Parkinson's-disease, known for their intracellular accumulation of protein aggregates. Recent observations of impaired autophagy also appeared in psychiatric disorders such as schizophrenia and bipolar disorder suggesting an additional contribution to the pathophysiology of mental illness. Here we review the current understanding of autophagy's role in various neuropsychiatric disorders and, hitherto, the prevailing new potential autophagy-related therapeutic strategies for their treatment.
Collapse
Affiliation(s)
- Tamara Bar-Yosef
- Department of Clinical Biochemistry and Pharmacology and Psychiatry Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev and Mental Health Center, Beersheba, Israel
| | - Odeya Damri
- Department of Clinical Biochemistry and Pharmacology and Psychiatry Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev and Mental Health Center, Beersheba, Israel
| | - Galila Agam
- Department of Clinical Biochemistry and Pharmacology and Psychiatry Research Unit, Faculty of Health Sciences, Ben-Gurion University of the Negev and Mental Health Center, Beersheba, Israel
| |
Collapse
|
15
|
Mohammadi A, Rashidi E, Amooeian VG. Brain, blood, cerebrospinal fluid, and serum biomarkers in schizophrenia. Psychiatry Res 2018; 265:25-38. [PMID: 29680514 DOI: 10.1016/j.psychres.2018.04.036] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/20/2018] [Accepted: 04/11/2018] [Indexed: 12/29/2022]
Abstract
Over the last decade, finding a reliable biomarker for the early detection of schizophrenia (Scz) has been a topic of interest. The main goal of the current review is to provide a comprehensive view of the brain, blood, cerebrospinal fluid (CSF), and serum biomarkers of Scz disease. Imaging studies have demonstrated that the volumes of the corpus callosum, thalamus, hippocampal formation, subiculum, parahippocampal gyrus, superior temporal gyrus, prefrontal and orbitofrontal cortices, and amygdala-hippocampal complex were reduced in patients diagnosed with Scz. It has been revealed that the levels of interleukin 1β (IL-1β), IL-6, IL-8, and TNF-α were increased in patients with Scz. Decreased mRNA levels of brain-derived neurotrophic factor (BDNF), tropomyosin receptor kinase B (TrkB), neurotrophin-3 (NT-3), nerve growth factor (NGF), and vascular endothelial growth factor (VEGF) genes have also been reported in Scz patients. Genes with known strong relationships with this disease include BDNF, catechol-O-methyltransferase (COMT), regulator of G-protein signaling 4 (RGS4), dystrobrevin-binding protein 1 (DTNBP1), neuregulin 1 (NRG1), Reelin (RELN), Selenium-binding protein 1 (SELENBP1), glutamic acid decarboxylase 67 (GAD 67), and disrupted in schizophrenia 1 (DISC1). The levels of dopamine, tyrosine hydroxylase (TH), serotonin or 5-hydroxytryptamine (5-HT) receptor 1A and B (5-HTR1A and 5-HTR1B), and 5-HT1B were significantly increased in Scz patients, while the levels of gamma-aminobutyric acid (GABA), 5-HT transporter (5-HTT), and 5-HT receptor 2A (5-HTR2A) were decreased. The increased levels of SELENBP1 and Glycogen synthase kinase 3 subunit α (GSK3α) genes in contrast with reduced levels of B-cell translocation gene 1 (BTG1), human leukocyte antigen DRB1 (HLA-DRB1), heterogeneous nuclear ribonucleoprotein A3 (HNRPA3), and serine/arginine-rich splicing factor 1 (SFRS1) genes have also been reported. This review covers various dysregulation of neurotransmitters and also highlights the strengths and weaknesses of studies attempting to identify candidate biomarkers.
Collapse
Affiliation(s)
- Alireza Mohammadi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ehsan Rashidi
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Ghasem Amooeian
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Scarr E, Udawela M, Dean B. Changed frontal pole gene expression suggest altered interplay between neurotransmitter, developmental, and inflammatory pathways in schizophrenia. NPJ SCHIZOPHRENIA 2018; 4:4. [PMID: 29463818 PMCID: PMC5820249 DOI: 10.1038/s41537-018-0044-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/22/2022]
Abstract
Schizophrenia (Sz) probably occurs after genetically susceptible individuals encounter a deleterious environmental factor that triggers epigenetic mechanisms to change CNS gene expression. To determine if omnibus changes in CNS gene expression are present in Sz, we compared mRNA levels in the frontal pole (Brodmann’s area (BA) 10), the dorsolateral prefrontal cortex (BA 9) and cingulate cortex (BA 33) from 15 subjects with Sz and 15 controls using the Affymetrix™ Human Exon 1.0 ST Array. Differences in mRNA levels (±≥20%; p < 0.01) were identified (JMP Genomics 5.1) and used to predict pathways and gene x gene interactions that would be affected by the changes in gene expression using Ingenuity Pathway Analysis. There was significant variation in mRNA levels with diagnoses for 566 genes in BA 10, 65 genes in BA 9 and 40 genes in BA 33. In Sz, there was an over-representation of genes with changed expression involved in inflammation and development in BA 10, cell morphology in BA 9 and amino acid metabolism and small molecule biochemistry in BA 33. Using 94 genes with altered levels of expression in BA 10 from subjects with Sz, it was possible to construct an interactome of proven direct gene x gene interactions that was enriched for genes in inflammatory, developmental, oestrogen, serotonergic, cholinergic and NRG1 regulated pathways. Our data shows complex, regionally specific changes in cortical gene expression in Sz that are predicted to affect homeostasis between biochemical pathways already proposed to be important in the pathophysiology of the disorder. Anterior brain regions exhibit significant amounts of differentially-expressed genes which might cause dysfunction in schizophrenia. It’s thought that schizophrenia occurs when environmental factors trigger gene expression changes and downstream effects in the human brain, though this is not fully understood. An Australian research group led by Brian Dean, from the Florey Institute of Neuroscience and Mental Health, conducted a post-mortem human brain study in which they compared gene expression between 15 schizophrenia patients and 15 controls. They found 566 instances of altered gene expression in the most frontal part of the brain, Brodmann Area 10, and fewer changes in proximal regions. These are brain areas known to mediate schizophrenia-related traits and the changes in gene expression in these areas will affect a range of essential biological pathways. The group also found 97 differentially-expressed genes that have been shown to directly interact with each. This study paints a complex picture of the causes of schizophrenia but suggests modern technologies can help unravel these complexities.
Collapse
Affiliation(s)
- Elizabeth Scarr
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC 3052, Australia.,CRC for Mental Health, Carlton, VIC, 3053, Australia.,Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Madhara Udawela
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC 3052, Australia.,CRC for Mental Health, Carlton, VIC, 3053, Australia
| | - Brian Dean
- Molecular Psychiatry Laboratory, Florey Institute for Neuroscience and Mental Health, Parkville, VIC 3052, Australia. .,CRC for Mental Health, Carlton, VIC, 3053, Australia. .,Research Centre for Mental Health, the Faculty of Health, Arts and Design, Swinburne University, Hawthorne, VIC, 3122, Australia.
| |
Collapse
|
17
|
Wei Y, Collin G, Mandl RCW, Cahn W, Keunen K, Schmidt R, Kahn RS, van den Heuvel MP. Cortical magnetization transfer abnormalities and connectome dysconnectivity in schizophrenia. Schizophr Res 2018; 192:172-178. [PMID: 28601503 DOI: 10.1016/j.schres.2017.05.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/15/2017] [Accepted: 05/24/2017] [Indexed: 01/16/2023]
Abstract
Macroscale dysconnectivity in schizophrenia is associated with neuropathological abnormalities. The extent to which alterations in cortical myelination as revealed in vivo by magnetization transfer ratio (MTR) are related to macroscale dysconnectivity remains unknown. We acquired magnetization transfer imaging (MTI) data and diffusion weighted imaging (DWI) data from 78 schizophrenia patients and 93 healthy controls for MTR extraction and connectome reconstruction to examine the possible link between cortical myelination and macroscale dysconnectivity. Our findings showed significant cortical MTR disruptions in several prefrontal areas in schizophrenia patients, including bilateral rostral middle frontal areas, right pars orbitalis, and right frontal pole. Furthermore, cortical MTR alterations between patients and controls were significantly correlated with the level of regional disconnectivity. Together, our findings provide evidence that microstructural neuropathological abnormalities in schizophrenia are predominately present in prefrontal areas of the cortex and are associated with alterations in structural connectome architecture at the whole brain network level.
Collapse
Affiliation(s)
- Yongbin Wei
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Guusje Collin
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - René C W Mandl
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wiepke Cahn
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kristin Keunen
- Brain Center Rudolf Magnus, Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ruben Schmidt
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - René S Kahn
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martijn P van den Heuvel
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
18
|
Caramaschi D, Sharp GC, Nohr EA, Berryman K, Lewis SJ, Davey Smith G, Relton CL. Exploring a causal role of DNA methylation in the relationship between maternal vitamin B12 during pregnancy and child's IQ at age 8, cognitive performance and educational attainment: a two-step Mendelian randomization study. Hum Mol Genet 2018; 26:3001-3013. [PMID: 28453778 PMCID: PMC5703349 DOI: 10.1093/hmg/ddx164] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/24/2017] [Indexed: 12/15/2022] Open
Abstract
An adequate intake of vitamin B12 during pregnancy plays an important role in
offspring neurodevelopment, potentially via epigenetic processes. We used a two-step
Mendelian randomization approach to assess whether DNA methylation plays a mediating and
causal role in associations between maternal vitamin B12 status and offspring’s
cognition. Firstly, we estimated the causal effect of maternal vitamin B12
levels on cord blood DNA methylation using the maternal FUT2 genotypes
rs492602:A > G and rs1047781:A > T as proxies for circulating vitamin B12
levels in the Avon Longitudinal Study of Parents and Children (ALSPAC) and we tested the
observed associations in a replication cohort. Secondly, we estimated the causal effect of
DNA methylation on IQ using the offspring genotype at sites close to the methylated CpG
site as a proxy for DNA methylation in ALSPAC and in a replication sample. The first step
Mendelian randomization estimated that maternal vitamin B12 had a small causal
effect on DNA methylation in offspring at three CpG sites, which was replicated for one of
the sites. The second step Mendelian randomization found weak evidence of a causal effect
of DNA methylation at two of these sites on childhood performance IQ which was replicated
for one of the sites. The findings support a causal effect of maternal vitamin
B12 levels on cord blood DNA methylation, and a causal effect of vitamin
B12-responsive DNA methylation changes on children’s cognition. Some
limitations were identified and future studies using a similar approach should aim to
overcome such issues.
Collapse
Affiliation(s)
- Doretta Caramaschi
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine
| | - Gemma C Sharp
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine.,School of Oral and Dental Sciences, The Cleft Collective, University of Bristol, Bristol BS8 2BN, UK
| | - Ellen A Nohr
- Research Unit for Gynaecology and Obstetrics, Department of Clinical Research, University of Southern Denmark, Odense 5000C, Denmark
| | - Katie Berryman
- School of Social and Community Medicine, University of Bristol, Bristol BS8?2BN, UK
| | - Sarah J Lewis
- School of Social and Community Medicine, University of Bristol, Bristol BS8?2BN, UK
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine
| | - Caroline L Relton
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine
| |
Collapse
|
19
|
Peng T, Wang L, Li G. The analysis of APOL1 genetic variation and haplotype diversity provided by 1000 Genomes project. BMC Nephrol 2017; 18:267. [PMID: 28800731 PMCID: PMC5553676 DOI: 10.1186/s12882-017-0675-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 07/19/2017] [Indexed: 11/20/2022] Open
Abstract
Background The APOL1 gene variants has been shown to be associated with an increased risk of multiple kinds of diseases, particularly in African Americans, but not in Caucasians and Asians. In this study, we explored the single nucleotide polymorphism (SNP) and haplotype diversity of APOL1 gene in different races provided by 1000 Genomes project. Methods Variants of APOL1 gene in 1000 Genome Project were obtained and SNPs located in the regulatory region or coding region were selected for genetic variation analysis. Total 2504 individuals from 26 populations were classified as four groups that included Africa, Europe, Asia and Admixed populations. Tag SNPs were selected to evaluate the haplotype diversities in the four populations by HaploStats software. Results APOL1 gene was surrounded by some of the most polymorphic genes in the human genome, variation of APOL1 gene was common, with up to 613 SNP (1000 Genome Project reported) and 99 of them (16.2%) with MAF ≥ 1%. There were 79 SNPs in the URR and 92 SNPs in 3’UTR. Total 12 SNPs in URR and 24 SNPs in 3’UTR were considered as common variants with MAF ≥ 1%. It is worth noting that URR-1 was presents lower frequencies in European populations, while other three haplotypes taken an opposite pattern; 3’UTR presents several high-frequency variation sites in a short segment, and the differences of its haplotypes among different population were significant (P < 0.01), UTR-1 and UTR-5 presented much higher frequency in African population, while UTR-2, UTR-3 and UTR-4 were much lower. APOL1 coding region showed that two SNP of G1 with higher frequency are actually pull down the haplotype H-1 frequency when considering all populations pooled together, and the diversity among the four populations be widen by the G1 two mutation (P1 = 3.33E-4 vs P2 = 3.61E-30). Conclusions The distributions of APOL1 gene variants and haplotypes were significantly different among the different populations, in either regulatory or coding regions. It could provide clues for the future genetic study of APOL1 related diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12882-017-0675-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ting Peng
- Renal Division and Institute of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Duan, 1st Circle Road, Qingyang District, Chengdu, Sichuan, People's Republic of China, 610072
| | - Li Wang
- Renal Division and Institute of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Duan, 1st Circle Road, Qingyang District, Chengdu, Sichuan, People's Republic of China, 610072
| | - Guisen Li
- Renal Division and Institute of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Duan, 1st Circle Road, Qingyang District, Chengdu, Sichuan, People's Republic of China, 610072.
| |
Collapse
|
20
|
A co-evolutionary arms race: trypanosomes shaping the human genome, humans shaping the trypanosome genome. Parasitology 2017; 142 Suppl 1:S108-19. [PMID: 25656360 PMCID: PMC4413828 DOI: 10.1017/s0031182014000602] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Trypanosoma brucei is the causative agent of African sleeping sickness in humans and one of several pathogens that cause the related veterinary disease Nagana. A complex co-evolution has occurred between these parasites and primates that led to the emergence of trypanosome-specific defences and counter-measures. The first line of defence in humans and several other catarrhine primates is the trypanolytic protein apolipoprotein-L1 (APOL1) found within two serum protein complexes, trypanosome lytic factor 1 and 2 (TLF-1 and TLF-2). Two sub-species of T. brucei have evolved specific mechanisms to overcome this innate resistance, Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense. In T. b. rhodesiense, the presence of the serum resistance associated (SRA) gene, a truncated variable surface glycoprotein (VSG), is sufficient to confer resistance to lysis. The resistance mechanism of T. b. gambiense is more complex, involving multiple components: reduction in binding affinity of a receptor for TLF, increased cysteine protease activity and the presence of the truncated VSG, T. b. gambiense-specific glycoprotein (TgsGP). In a striking example of co-evolution, evidence is emerging that primates are responding to challenge by T. b. gambiense and T. b. rhodesiense, with several populations of humans and primates displaying resistance to infection by these two sub-species.
Collapse
|
21
|
Abstract
The etiology and pathophysiology of schizophrenia and related mental disorders such as bipolar disorder and major depression remain largely unclear. Recent advances in mRNA profiling techniques made it possible to perform genome-wide gene expression analysis in a hypothesis-free manner. It was thought that this large-scale data mining approach would reveal unknown molecular cascades involved in mental disorders. Contrary to this initial expectation, however, DNA microarray results in psychiatric fields have been notoriously discordant. Here the authors review the findings of DNA microarray analysis, focusing on systematic gene expression changes in schizophrenia, as well as alterations in the expression of specific genes, that have been reported and replicated. The authors also address the probable causes for the discordance among studies, possible ways to solve the problem, and their preferred approach for data interpretation.
Collapse
Affiliation(s)
- Kazuya Iwamoto
- Laboratory for Molecular Dynamics of Mental Disorders, Brain Science Institute, RIKEN, Saitama, Japan.
| | | |
Collapse
|
22
|
Dummer PD, Limou S, Rosenberg AZ, Heymann J, Nelson G, Winkler CA, Kopp JB. APOL1 Kidney Disease Risk Variants: An Evolving Landscape. Semin Nephrol 2016. [PMID: 26215860 DOI: 10.1016/j.semnephrol.2015.04.008] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Apolipoprotein L1 (APOL1) genetic variants account for much of the excess risk of chronic and end-stage kidney disease, which results in a significant global health disparity for persons of African ancestry. We estimate the lifetime risk of kidney disease in APOL1 dual-risk allele individuals to be at least 15%. Experimental evidence suggests a direct role of APOL1 in pore formation, cellular injury, and programmed cell death in renal injury. The APOL1 BH3 motif, often associated with cell death, is unlikely to play a role in APOL1-induced cytotoxicity because it is not conserved within the APOL family and is dispensable for cell death in vitro. We discuss two models for APOL1 trypanolytic activity: one involving lysosome permeabilization and another involving colloid-osmotic swelling of the cell body, as well as their relevance to human pathophysiology. Experimental evidence from human cell culture models suggests that both mechanisms may be operative. A systems biology approach whereby APOL1-associated perturbations in gene and protein expression in affected individuals are correlated with molecular pathways may be productive to elucidate APOL1 function in vivo.
Collapse
Affiliation(s)
- Patrick D Dummer
- Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Sophie Limou
- Molecular Epidemiology Genetics Section, Center for Cancer Research, National Cancer Institute, Frederick MD
| | - Avi Z Rosenberg
- Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD; Department of Pathology Johns Hopkins University, Baltimore, MD
| | - Jurgen Heymann
- Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - George Nelson
- Molecular Epidemiology Genetics Section, Center for Cancer Research, National Cancer Institute, Frederick MD
| | - Cheryl A Winkler
- Molecular Epidemiology Genetics Section, Center for Cancer Research, National Cancer Institute, Frederick MD
| | - Jeffrey B Kopp
- Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
23
|
Butler MG, McGuire AB, Masoud H, Manzardo AM. Currently recognized genes for schizophrenia: High-resolution chromosome ideogram representation. Am J Med Genet B Neuropsychiatr Genet 2016; 171B:181-202. [PMID: 26462458 PMCID: PMC6679920 DOI: 10.1002/ajmg.b.32391] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/02/2015] [Indexed: 11/09/2022]
Abstract
A large body of genetic data from schizophrenia-related research has identified an assortment of genes and disturbed pathways supporting involvement of complex genetic components for schizophrenia spectrum and other psychotic disorders. Advances in genetic technology and expanding studies with searchable genomic databases have led to multiple published reports, allowing us to compile a master list of known, clinically relevant, or susceptibility genes contributing to schizophrenia. We searched key words related to schizophrenia and genetics from peer-reviewed medical literature sources, authoritative public access psychiatric websites and genomic databases dedicated to gene discovery and characterization of schizophrenia. Our list of 560 genes were arranged in alphabetical order in tabular form with gene symbols placed on high-resolution human chromosome ideograms. Genome wide pathway analysis using GeneAnalytics was carried out on the resulting list of genes to assess the underlying genetic architecture for schizophrenia. Recognized genes of clinical relevance, susceptibility or causation impact a broad range of biological pathways and mechanisms including ion channels (e.g., CACNA1B, CACNA1C, CACNA1H), metabolism (e.g., CYP1A2, CYP2C19, CYP2D6), multiple targets of neurotransmitter pathways impacting dopamine, GABA, glutamate, and serotonin function, brain development (e.g., NRG1, RELN), signaling peptides (e.g., PIK3CA, PIK4CA) and immune function (e.g., HLA-DRB1, HLA-DQA1) and interleukins (e.g., IL1A, IL10, IL6). This summary will enable clinical and laboratory geneticists, genetic counselors, and other clinicians to access convenient pictorial images of the distribution and location of contributing genes to inform diagnosis and gene-based treatment as well as provide risk estimates for genetic counseling of families with affected relatives.
Collapse
Affiliation(s)
- Merlin G. Butler
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, Kansas,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas,Correspondence to: Merlin G. Butler, M.D., Ph.D., University of Kansas Medical Center, Department of Psychiatry and Behavioral Sciences, 3901 Rainbow Boulevard, MS 4015, Kansas City, KS 66160,
| | - Austen B. McGuire
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, Kansas
| | - Humaira Masoud
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, Kansas
| | - Ann M. Manzardo
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
24
|
Sparrow S, Manning JR, Cartier J, Anblagan D, Bastin ME, Piyasena C, Pataky R, Moore EJ, Semple SI, Wilkinson AG, Evans M, Drake AJ, Boardman JP. Epigenomic profiling of preterm infants reveals DNA methylation differences at sites associated with neural function. Transl Psychiatry 2016; 6:e716. [PMID: 26784970 PMCID: PMC5068883 DOI: 10.1038/tp.2015.210] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 12/13/2022] Open
Abstract
DNA methylation (DNAm) plays a determining role in neural cell fate and provides a molecular link between early-life stress and neuropsychiatric disease. Preterm birth is a profound environmental stressor that is closely associated with alterations in connectivity of neural systems and long-term neuropsychiatric impairment. The aims of this study were to examine the relationship between preterm birth and DNAm, and to investigate factors that contribute to variance in DNAm. DNA was collected from preterm infants (birth<33 weeks gestation) and healthy controls (birth>37 weeks), and a genome-wide analysis of DNAm was performed; diffusion magnetic resonance imaging (dMRI) data were acquired from the preterm group. The major fasciculi were segmented, and fractional anisotropy, mean diffusivity and tract shape were calculated. Principal components (PC) analysis was used to investigate the contribution of MRI features and clinical variables to variance in DNAm. Differential methylation was found within 25 gene bodies and 58 promoters of protein-coding genes in preterm infants compared with controls; 10 of these have neural functions. Differences detected in the array were validated with pyrosequencing. Ninety-five percent of the variance in DNAm in preterm infants was explained by 23 PCs; corticospinal tract shape associated with 6th PC, and gender and early nutritional exposure associated with the 7th PC. Preterm birth is associated with alterations in the methylome at sites that influence neural development and function. Differential methylation analysis has identified several promising candidate genes for understanding the genetic/epigenetic basis of preterm brain injury.
Collapse
Affiliation(s)
- S Sparrow
- MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - J R Manning
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - J Cartier
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - D Anblagan
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - M E Bastin
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - C Piyasena
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - R Pataky
- MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - E J Moore
- MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - S I Semple
- Clinical Research Imaging Centre, University of Edinburgh, Edinburgh, UK
| | | | - M Evans
- Department of Pathology, NHS Lothian, Edinburgh, UK
| | - A J Drake
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - J P Boardman
- MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK,MRC Centre for Reproductive Health, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Room W1.26, Edinburgh EH16 4TJ, UK. E-mail:
| |
Collapse
|
25
|
Song YR, Wu B, Yang YT, Chen J, Zhang LJ, Zhang ZW, Shi HY, Huang CL, Pan JX, Xie P. Specific alterations in plasma proteins during depressed, manic, and euthymic states of bipolar disorder. ACTA ACUST UNITED AC 2015; 48:973-82. [PMID: 26375446 PMCID: PMC4671523 DOI: 10.1590/1414-431x20154550] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 04/14/2015] [Indexed: 12/22/2022]
Abstract
Bipolar disorder (BD) is a common psychiatric mood disorder affecting more than 1-2%
of the general population of different European countries. Unfortunately, there is no
objective laboratory-based test to aid BD diagnosis or monitor its progression, and
little is known about the molecular basis of BD. Here, we performed a comparative
proteomic study to identify differentially expressed plasma proteins in various BD
mood states (depressed BD, manic BD, and euthymic BD) relative to healthy controls. A
total of 10 euthymic BD, 20 depressed BD, 15 manic BD, and 20 demographically matched
healthy control subjects were recruited. Seven high-abundance proteins were
immunodepleted in plasma samples from the 4 experimental groups, which were then
subjected to proteome-wide expression profiling by two-dimensional electrophoresis
and matrix-assisted laser desorption/ionization-time-of-flight/time-of-flight tandem
mass spectrometry. Proteomic results were validated by immunoblotting and
bioinformatically analyzed using MetaCore. From a total of 32 proteins identified
with 1.5-fold changes in expression compared with healthy controls, 16 proteins were
perturbed in BD independent of mood state, while 16 proteins were specifically
associated with particular BD mood states. Two mood-independent differential
proteins, apolipoprotein (Apo) A1 and Apo L1, suggest that BD pathophysiology may be
associated with early perturbations in lipid metabolism. Moreover, down-regulation of
one mood-dependent protein, carbonic anhydrase 1 (CA-1), suggests it may be involved
in the pathophysiology of depressive episodes in BD. Thus, BD pathophysiology may be
associated with early perturbations in lipid metabolism that are independent of mood
state, while CA-1 may be involved in the pathophysiology of depressive episodes.
Collapse
Affiliation(s)
- Y R Song
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - B Wu
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Y T Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - J Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - L J Zhang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Z W Zhang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - H Y Shi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - C L Huang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - J X Pan
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - P Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Kreit M, Vertommen D, Gillet L, Michiels T. The Interferon-Inducible Mouse Apolipoprotein L9 and Prohibitins Cooperate to Restrict Theiler's Virus Replication. PLoS One 2015. [PMID: 26196674 PMCID: PMC4510265 DOI: 10.1371/journal.pone.0133190] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Apolipoprotein L9b (Apol9b) is an interferon-stimulated gene (ISG) that has antiviral activity and is weakly expressed in primary mouse neurons as compared to other cell types. Here, we show that both Apol9 isoforms (Apol9b and Apol9a) inhibit replication of Theiler’s murine encephalomyelitis virus (TMEV) but not replication of vesicular stomatitis virus (VSV), Murid herpesvirus-4 (MuHV-4), or infection by a lentiviral vector. Apol9 genes are strongly expressed in mouse liver and, to a lesser extent, in pancreas, adipose tissue and intestine. Their expression is increased by type I interferon and viral infection. In contrast to genuine apolipoproteins that are involved in lipid transport, ApoL9 has an intracytoplasmic localization and does not seem to be secreted. The cytoplasmic localization of ApoL9 is in line with the observation that ApoL9 inhibits the replication step of TMEV infection. In contrast to human ApoL6, ApoL9 did not sensitize cells to apoptosis, in spite of the presence of a conserved putative BH3 domain, required for antiviral activity. ApoL9a and b isoforms interact with cellular prohibitin 1 (Phb1) and prohibitin 2 (Phb2) and this interaction might contribute to ApoL9 antiviral activity. Knocking down Phb2 slightly increased TMEV replication, irrespective of ApoL9 overexpression. The antiviral activity of prohibitins against TMEV contrasts with the pro-viral activity of prohibitins observed for VSV and reported previously for Dengue 2 (DENV-2), Chikungunya (CHIKV) and influenza H5N1 viruses. ApoL9 is thus an example of ISG displaying a narrow antiviral range, which likely acts in complex with prohibitins to restrict TMEV replication.
Collapse
Affiliation(s)
- Marguerite Kreit
- Université catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Didier Vertommen
- Université catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Laurent Gillet
- Université de Liège, FARAH Research Center and Faculté de Médecine Vétérinaire, Liège, Belgium
| | - Thomas Michiels
- Université catholique de Louvain, de Duve Institute, Brussels, Belgium
- * E-mail:
| |
Collapse
|
27
|
Long LE, Lind J, Webster M, Weickert CS. Developmental trajectory of the endocannabinoid system in human dorsolateral prefrontal cortex. BMC Neurosci 2012; 13:87. [PMID: 22827915 PMCID: PMC3464170 DOI: 10.1186/1471-2202-13-87] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Accepted: 06/28/2012] [Indexed: 12/11/2022] Open
Abstract
Background Endocannabinoids provide control over cortical neurotransmission. We investigated the developmental expression of key genes in the endocannabinoid system across human postnatal life and determined whether they correspond to the development of markers for inhibitory interneurons, which shape cortical development. We used microarray with qPCR validation and in situ hybridisation to quantify mRNA for the central endocannabinoid receptor CB1R, endocannabinoid synthetic enzymes (DAGLα for 2-arachidonylglycerol [2-AG] and NAPE-PLD for anandamide), and inactivating enzymes (MGL and ABHD6 for 2-AG and FAAH for anandamide) in human dorsolateral prefrontal cortex (39 days - 49 years). Results CB1R mRNA decreases until adulthood, particularly in layer II, after peaking between neonates and toddlers. DAGLα mRNA expression is lowest in early life and adulthood, peaking between school age and young adulthood. MGL expression declines after peaking in infancy, while ABHD6 increases from neonatal age. NAPE-PLD and FAAH expression increase steadily after infancy, peaking in adulthood. Conclusions Stronger endocannabinoid regulation of presynaptic neurotransmission in both supragranular and infragranular cortical layers as indexed through higher CB1R mRNA may occur within the first few years of human life. After adolescence, higher mRNA levels of the anandamide synthetic and inactivating enzymes NAPE-PLD and FAAH suggest that a late developmental switch may occur where anandamide is more strongly regulated after adolescence than earlier in life. Thus, expression of key genes in the endocannabinoid system changes with maturation of cortical function.
Collapse
Affiliation(s)
- Leonora E Long
- Schizophrenia Research Institute, Darlinghurst, NSW 2010, Australia
| | | | | | | |
Collapse
|
28
|
Wu JQ, Wang X, Beveridge NJ, Tooney PA, Scott RJ, Carr VJ, Cairns MJ. Transcriptome sequencing revealed significant alteration of cortical promoter usage and splicing in schizophrenia. PLoS One 2012; 7:e36351. [PMID: 22558445 PMCID: PMC3338678 DOI: 10.1371/journal.pone.0036351] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 04/03/2012] [Indexed: 12/21/2022] Open
Abstract
Background While hybridization based analysis of the cortical transcriptome has provided important insight into the neuropathology of schizophrenia, it represents a restricted view of disease-associated gene activity based on predetermined probes. By contrast, sequencing technology can provide un-biased analysis of transcription at nucleotide resolution. Here we use this approach to investigate schizophrenia-associated cortical gene expression. Methodology/Principal Findings The data was generated from 76 bp reads of RNA-Seq, aligned to the reference genome and assembled into transcripts for quantification of exons, splice variants and alternative promoters in postmortem superior temporal gyrus (STG/BA22) from 9 male subjects with schizophrenia and 9 matched non-psychiatric controls. Differentially expressed genes were then subjected to further sequence and functional group analysis. The output, amounting to more than 38 Gb of sequence, revealed significant alteration of gene expression including many previously shown to be associated with schizophrenia. Gene ontology enrichment analysis followed by functional map construction identified three functional clusters highly relevant to schizophrenia including neurotransmission related functions, synaptic vesicle trafficking, and neural development. Significantly, more than 2000 genes displayed schizophrenia-associated alternative promoter usage and more than 1000 genes showed differential splicing (FDR<0.05). Both types of transcriptional isoforms were exemplified by reads aligned to the neurodevelopmentally significant doublecortin-like kinase 1 (DCLK1) gene. Conclusions This study provided the first deep and un-biased analysis of schizophrenia-associated transcriptional diversity within the STG, and revealed variants with important implications for the complex pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Jing Qin Wu
- Schizophrenia Research Institute, Sydney, Australia
- School of Biomedical Sciences and Pharmacy, Faculty of Health, Priority Research Centre for Translational Neuroscience and Mental Health, The University of Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Xi Wang
- School of Biomedical Sciences and Pharmacy, Faculty of Health, Priority Research Centre for Translational Neuroscience and Mental Health, The University of Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Natalie J. Beveridge
- Schizophrenia Research Institute, Sydney, Australia
- School of Biomedical Sciences and Pharmacy, Faculty of Health, Priority Research Centre for Translational Neuroscience and Mental Health, The University of Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Paul A. Tooney
- Schizophrenia Research Institute, Sydney, Australia
- School of Biomedical Sciences and Pharmacy, Faculty of Health, Priority Research Centre for Translational Neuroscience and Mental Health, The University of Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Rodney J. Scott
- School of Biomedical Sciences and Pharmacy, Faculty of Health, Priority Research Centre for Translational Neuroscience and Mental Health, The University of Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Vaughan J. Carr
- Schizophrenia Research Institute, Sydney, Australia
- Research Unit for Schizophrenia Epidemiology, School of Psychiatry, University of New South Wales, St. Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | - Murray J. Cairns
- Schizophrenia Research Institute, Sydney, Australia
- School of Biomedical Sciences and Pharmacy, Faculty of Health, Priority Research Centre for Translational Neuroscience and Mental Health, The University of Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton, New South Wales, Australia
- * E-mail:
| |
Collapse
|
29
|
Hu CAA, Klopfer EI, Ray PE. Human apolipoprotein L1 (ApoL1) in cancer and chronic kidney disease. FEBS Lett 2012; 586:947-55. [PMID: 22569246 DOI: 10.1016/j.febslet.2012.03.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 02/28/2012] [Accepted: 03/01/2012] [Indexed: 02/09/2023]
Abstract
Human apolipoprotein L1 (ApoL1) possesses both extra- and intra-cellular functions crucial in host defense and cellular homeostatic mechanisms. Alterations in ApoL1 function due to genetic, environmental, and lifestyle factors have been associated with African sleeping sickness, atherosclerosis, lipid disorders, obesity, schizophrenia, cancer, and chronic kidney disease (CKD). Importantly, two alleles of APOL1 carrying three coding-sequence variants have been linked to CKD, particularly in Sub-Saharan Africans and African Americans. Intracellularly, elevated ApoL1 can induce autophagy and autophagy-associated cell death, which may be critical in the maintenance of cellular homeostasis in the kidney. Similarly, ApoL1 may protect kidney cells against renal cell carcinoma (RCC). We summarize the role of ApoL1 in RCC and CKD, highlighting the critical function of ApoL1 in autophagy.
Collapse
Affiliation(s)
- Chien-An A Hu
- Department of Biochemistry and Molecular Biology, University of New Mexico, Health Sciences Center, Albuquerque, NM 87131-0001, USA.
| | | | | |
Collapse
|
30
|
Rothmond DA, Weickert CS, Webster MJ. Developmental changes in human dopamine neurotransmission: cortical receptors and terminators. BMC Neurosci 2012; 13:18. [PMID: 22336227 PMCID: PMC3315415 DOI: 10.1186/1471-2202-13-18] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 02/15/2012] [Indexed: 11/10/2022] Open
Abstract
Background Dopamine is integral to cognition, learning and memory, and dysfunctions of the frontal cortical dopamine system have been implicated in several developmental neuropsychiatric disorders. The dorsolateral prefrontal cortex (DLPFC) is critical for working memory which does not fully mature until the third decade of life. Few studies have reported on the normal development of the dopamine system in human DLPFC during postnatal life. We assessed pre- and postsynaptic components of the dopamine system including tyrosine hydroxylase, the dopamine receptors (D1, D2 short and D2 long isoforms, D4, D5), catechol-O-methyltransferase, and monoamine oxidase (A and B) in the developing human DLPFC (6 weeks -50 years). Results Gene expression was first analysed by microarray and then by quantitative real-time PCR. Protein expression was analysed by western blot. Protein levels for tyrosine hydroxylase peaked during the first year of life (p < 0.001) then gradually declined to adulthood. Similarly, mRNA levels of dopamine receptors D2S (p < 0.001) and D2L (p = 0.003) isoforms, monoamine oxidase A (p < 0.001) and catechol-O-methyltransferase (p = 0.024) were significantly higher in neonates and infants as was catechol-O-methyltransferase protein (32 kDa, p = 0.027). In contrast, dopamine D1 receptor mRNA correlated positively with age (p = 0.002) and dopamine D1 receptor protein expression increased throughout development (p < 0.001) with adults having the highest D1 protein levels (p ≤ 0.01). Monoamine oxidase B mRNA and protein (p < 0.001) levels also increased significantly throughout development. Interestingly, dopamine D5 receptor mRNA levels negatively correlated with age (r = -0.31, p = 0.018) in an expression profile opposite to that of the dopamine D1 receptor. Conclusions We find distinct developmental changes in key components of the dopamine system in DLPFC over postnatal life. Those genes that are highly expressed during the first year of postnatal life may influence and orchestrate the early development of cortical neural circuitry while genes portraying a pattern of increasing expression with age may indicate a role in DLPFC maturation and attainment of adult levels of cognitive function.
Collapse
Affiliation(s)
- Debora A Rothmond
- Schizophrenia Research Institute, 405 Liverpool St, Darlinghurst NSW 2010 AU.
| | | | | |
Collapse
|
31
|
Expression profiling in neuropsychiatric disorders: emphasis on glutamate receptors in bipolar disorder. Pharmacol Biochem Behav 2011; 100:705-11. [PMID: 22005598 DOI: 10.1016/j.pbb.2011.09.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 09/20/2011] [Accepted: 09/30/2011] [Indexed: 02/08/2023]
Abstract
Functional genomics and proteomics approaches are being employed to evaluate gene and encoded protein expression changes with the tacit goal to find novel targets for drug discovery. Genome-wide association studies (GWAS) have attempted to identify valid candidate genes through single nucleotide polymorphism (SNP) analysis. Furthermore, microarray analysis of gene expression in brain regions and discrete cell populations has enabled the simultaneous quantitative assessment of relevant genes. The ability to associate gene expression changes with neuropsychiatric disorders, including bipolar disorder (BP), and their response to therapeutic drugs provides a novel means for pharmacotherapeutic interventions. This review summarizes gene and pathway targets that have been identified in GWAS studies and expression profiling of human postmortem brain in BP, with an emphasis on glutamate receptors (GluRs). Although functional genomic assessment of BP is in its infancy, results to date point towards a dysregulation of GluRs that bear some similarity to schizophrenia (SZ), although the pattern is complex, and likely to be more complementary than overlapping. The importance of single population expression profiling of specific neurons and intrinsic circuits is emphasized, as this approach provides informative gene expression profile data that may be underappreciated in regional studies with admixed neuronal and non-neuronal cell types.
Collapse
|
32
|
Mitchell AC, Mirnics K. Gene expression profiling of the brain: pondering facts and fiction. Neurobiol Dis 2011; 45:3-7. [PMID: 21689753 DOI: 10.1016/j.nbd.2011.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 05/31/2011] [Accepted: 06/06/2011] [Indexed: 10/18/2022] Open
Abstract
During the last decade brain transcriptome profiling by DNA microarrays has matured, developed sound experimental design standards, reporting practices, analytical procedures, and data sharing resources. It has become a powerful scientific tool in the exploratory research portfolio. Along this journey by trial and error, we encountered a number of intriguing questions and comments--pondering the value of hypothesis-driven research, appropriate sample size, the importance and interpretation of transcripts changes vis-à-vis protein changes, the role of statistical stringency, false discovery and magnitude of expression change, and many other interesting questions. Our field fully acknowledges and tries to address all of these challenges associated with high-throughput, data-driven transcriptomics. As a research field, we strongly advocate implementing the highest standards of our trade, and we deeply believe that transcriptome profiling studies will continue to be essential for deciphering the pathophysiological mechanisms leading to complex brain disorders.
Collapse
Affiliation(s)
- Amanda C Mitchell
- Department of Psychiatry, Vanderbilt University, Nashville, TN 37203, USA
| | | |
Collapse
|
33
|
Zhan L, Kerr JR, Lafuente MJ, Maclean A, Chibalina MV, Liu B, Burke B, Bevan S, Nasir J. Altered expression and coregulation of dopamine signalling genes in schizophrenia and bipolar disorder. Neuropathol Appl Neurobiol 2011; 37:206-19. [PMID: 20874815 DOI: 10.1111/j.1365-2990.2010.01128.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION signalling through dopamine receptors is of critical importance in the brain and is implicated in schizophrenia and bipolar disorder, but its underlying molecular mechanisms remain poorly understood. MATERIALS AND METHODS using a yeast two-hybrid approach, we previously identified 11 novel dopamine receptor-interacting proteins. Here we compare gene expression levels for 17 genes [including all 11 dopamine receptor interacting proteins, all 5 dopamine receptors (DRD1-DRD5) and DARPP-32] by real-time polymerase chain reaction, using prefrontal cortex post mortem brain samples from 33 schizophrenic, 32 bipolar disorder and 34 control subjects. RESULTS the expression of C14ORF28, GNB2L1, MLLT3, DRD2 and DARPP-32 genes was altered in schizophrenia and/or bipolar disorder samples relative to controls (P < 0.05). Hierarchical clustering analysis revealed the expression of these five genes (C14ORF28, GNB2L1, MLLT3, DARPP-32, DRD2) is closely correlated in patients. However, in controls, DRD2 expression in relation to the other genes appears to be very different, suggesting abnormal DRD2 activity is an important trigger in the pathophysiology of schizophrenia and bipolar disorder. CONCLUSIONS our data suggest: (i) C14ORF28, GNB2L1, MLLT3, DRD2 and DARPP-32 are important in the pathogenesis of schizophrenia and bipolar disorder; (ii) these two disorders share common disease-related mechanisms linked to dopamine signalling; (iii) the expression of these genes is closely correlated; and (iv) DRD2 provides the initial trigger in the pathogenesis of these disorders.
Collapse
Affiliation(s)
- L Zhan
- Clinical Developmental Sciences, St. George's University of London, Cranmer Terrace, London
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sequeira PA, Martin MV, Vawter MP. The first decade and beyond of transcriptional profiling in schizophrenia. Neurobiol Dis 2011; 45:23-36. [PMID: 21396449 DOI: 10.1016/j.nbd.2011.03.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 02/28/2011] [Accepted: 03/02/2011] [Indexed: 01/19/2023] Open
Abstract
Gene expression changes in brains of individuals with schizophrenia (SZ) have been hypothesized to reflect possible pathways related to pathophysiology and/or medication. Other factors having robust effects on gene expression profiling in brain and possibly influence the schizophrenia transcriptome such as age and pH are examined. Pathways of curated gene expression or gene correlation networks reported in SZ (white matter, apoptosis, neurogenesis, synaptic plasticity, glutamatergic and GABAergic neurotransmission, immune and stress-response, mitochondrial, and neurodevelopment) are not unique to SZ and have been associated with other psychiatric disorders. Suggestions going forward to improve the next decade of profiling: consider multiple brain regions that are carefully dissected, release large datasets from multiple brain regions in controls to better understand neurocircuitry, integrate genetics and gene expression, measure expression variants on genome wide level, peripheral biomarker studies, and analyze the transcriptome across a developmental series of brains. Gene expression, while an important feature of the genomic landscape, requires further systems biology to advance from control brains to a more precise definition of the schizophrenia interactome.
Collapse
Affiliation(s)
- P Adolfo Sequeira
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, Irvine, CA 92697-4260, USA
| | | | | |
Collapse
|
35
|
Abstract
Schizophrenia is one of the most severe psychiatric disorders affecting 1% of the world population. There is yet no empirical method to validate the diagnosis of the disease. The identification of an underlying molecular alteration could lead to an improved disease understanding and may yield an objective panel of biomarkers to aid in the diagnosis of this devastating disease. Presented is the largest reported liquid chromatography-mass spectrometry-based proteomic profiling study investigating serum samples taken from first-onset drug-naive patients compared with samples collected from healthy volunteers. The results of this large-scale study are presented along with enzyme-linked immunosorbent assay-based validation data.
Collapse
|
36
|
Jagannathan K, Calhoun VD, Gelernter J, Stevens MC, Liu J, Bolognani F, Windemuth A, Ruaño G, Assaf M, Pearlson GD. Genetic associations of brain structural networks in schizophrenia: a preliminary study. Biol Psychiatry 2010; 68:657-66. [PMID: 20691427 PMCID: PMC2990476 DOI: 10.1016/j.biopsych.2010.06.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Revised: 05/25/2010] [Accepted: 06/03/2010] [Indexed: 11/29/2022]
Abstract
BACKGROUND Schizophrenia is a complex genetic disorder, with multiple putative risk genes and many reports of reduced cortical gray matter. Identifying the genetic loci contributing to these structural alterations in schizophrenia (and likely also to normal structural gray matter patterns) could aid understanding of schizophrenia's pathophysiology. We used structural parameters as potential intermediate illness markers to investigate genomic factors derived from single nucleotide polymorphism (SNP) arrays. METHOD We used research quality structural magnetic resonance imaging (sMRI) scans from European American subjects including 33 healthy control subjects and 18 schizophrenia patients. All subjects were genotyped for 367 SNPs. Linked sMRI and genetic (SNP) components were extracted to reveal relationships between brain structure and SNPs, using parallel independent component analysis, a novel multivariate approach that operates effectively in small sample sizes. RESULTS We identified an sMRI component that significantly correlated with a genetic component (r = -.536, p < .00005); components also distinguished groups. In the sMRI component, schizophrenia gray matter deficits were in brain regions consistently implicated in previous reports, including frontal and temporal lobes and thalamus (p < .01). These deficits were related to SNPs from 16 genes, several previously associated with schizophrenia risk and/or involved in normal central nervous system development, including AKT, PI3K, SLC6A4, DRD2, CHRM2, and ADORA2A. CONCLUSIONS Despite the small sample size, this novel analysis method identified an sMRI component including brain areas previously reported to be abnormal in schizophrenia and an associated genetic component containing several putative schizophrenia risk genes. Thus, we identified multiple genes potentially underlying specific structural brain abnormalities in schizophrenia.
Collapse
Affiliation(s)
- Kanchana Jagannathan
- Olin Neuropsychiatry Research Center, Institute of Living/Hartford Hospital, Hartford, Connecticut 06106, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Carrera N, Arrojo M, Paz E, Ramos-Ríos R, Agra S, Páramo M, Brenlla J, Costas J. Testing the antagonistic pleiotropy model of schizophrenia susceptibility by analysis of DAOA, PPP1R1B, and APOL1 genes. Psychiatry Res 2010; 179:126-9. [PMID: 20483474 DOI: 10.1016/j.psychres.2009.05.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 04/16/2009] [Accepted: 05/26/2009] [Indexed: 10/19/2022]
Abstract
Schizophrenia is a common disease associated with reduced fertility. Therefore, the existence of common susceptibility alleles not removed by natural selection may be considered an evolutionary paradox. The antagonistic pleiotropy model, proposed to explain this paradox, states that an allele may be common because of its overall selective advantage, in spite of deleterious effects on specific traits. Recent work on DAOA, PPP1R1B, and APOL1 suggests that these genes present common alleles associated to increase risk of schizophrenia but conferring an overall selective advantage, related to better cognitive performance (DAOA and PPP1R1B) or protection against pathogens (APOL1). To test if these genes fit the antagonistic pleiotropy model, we searched for recent natural selection at these loci applying the long-range haplotype test on data from the HapMap Project; and performed case-control association analysis in a well-powered sample, including 301 schizophrenic patients and 604 controls from Spain. For DAOA and PPP1R1B, we genotyped the Single-nucleotide polymorphisms (SNPs) needed to replicate previous associations, while for APOL1, we genotyped 15 tagSNPs, and seven putative functional SNPs. We did not detect evidence of recent natural selection. Furthermore, we did not find significant associations. Thus, these genes do not fit the antagonistic pleiotropy model.
Collapse
Affiliation(s)
- Noa Carrera
- Fundación Pública Galega de Medicina Xenómica, Santiago de Compostela, Spain
| | | | | | | | | | | | | | | |
Collapse
|
38
|
McManus CA, Polden J, Cotter DR, Dunn MJ. Two-dimensional reference map for the basic proteome of the human dorsolateral prefrontal cortex (dlPFC) of the prefrontal lobe region of the brain. Proteomics 2010; 10:2551-5. [PMID: 20432482 DOI: 10.1002/pmic.200900705] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We describe a 2-DE proteomic reference map containing 227 basic proteins in the dorsolateral prefrontal cortex region of the human brain. Proteins were separated in the first dimension on pH 6-11 IPG strips using paper-bridge loading and on 12% SDS-PAGE in the second dimension. Proteins were subsequently identified by MS and spectra were analyzed using an in-house proteomics data analysis platform, Proline. The 2-DE reference map is available via the UCD 2-DE Proteome Database (http://proteomics-portal.ucd.ie:8082) and can also be accessed via the WORLD-2DPAGE Portal (http://www.expasy.ch/world-2dpage/). The associated protein identification data have been submitted to the PRIDE database (accession numbers 10018-10033). Separation of proteins in the basic region resolves more membrane associated proteins relevant to the synaptic pathology central to many neurological disorders. The 2-DE reference map will aid with further characterisation of neurological disorders such as bipolar and schizophrenia.
Collapse
Affiliation(s)
- Ciara A McManus
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland.
| | | | | | | |
Collapse
|
39
|
Elliott DA, Weickert CS, Garner B. Apolipoproteins in the brain: implications for neurological and psychiatric disorders. ACTA ACUST UNITED AC 2010; 51:555-573. [PMID: 21423873 DOI: 10.2217/clp.10.37] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The brain is the most lipid-rich organ in the body and, owing to the impermeable nature of the blood-brain barrier, lipid and lipoprotein metabolism within this organ is distinct from the rest of the body. Apolipoproteins play a well-established role in the transport and metabolism of lipids within the CNS; however, evidence is emerging that they also fulfill a number of functions that extend beyond lipid transport and are critical for healthy brain function. The importance of apolipoproteins in brain physiology is highlighted by genetic studies, where apolipoprotein gene polymorphisms have been identified as risk factors for several neurological diseases. Furthermore, the expression of brain apolipoproteins is significantly altered in several brain disorders. The purpose of this article is to provide an up-to-date assessment of the major apolipoproteins found in the brain (ApoE, ApoJ, ApoD and ApoA-I), covering their proposed roles and the factors influencing their level of expression. Particular emphasis is placed on associations with neurological and psychiatric disorders.
Collapse
Affiliation(s)
- David A Elliott
- Prince of Wales Medical Research Institute, Randwick, Sydney, NSW 2031, Australia
| | | | | |
Collapse
|
40
|
Prefrontal GABA(A) receptor alpha-subunit expression in normal postnatal human development and schizophrenia. J Psychiatr Res 2010; 44:673-81. [PMID: 20100621 DOI: 10.1016/j.jpsychires.2009.12.007] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2009] [Revised: 12/08/2009] [Accepted: 12/18/2009] [Indexed: 11/23/2022]
Abstract
Cortical GABA deficits that are consistently reported in schizophrenia may reflect an etiology of failed normal postnatal neurotransmitter maturation. Previous studies have found prefrontal cortical GABA(A) receptor alpha subunit alterations in schizophrenia, yet their relationship to normal developmental expression profiles in the human cortex has not been determined. The aim of this study was to quantify GABA(A) receptor alpha-subunit mRNA expression patterns in human dorsolateral prefrontal cortex (DLPFC) during normal postnatal development and in schizophrenia cases compared to controls. Transcript levels of GABA(A) receptor alpha subunits were measured using microarray and qPCR analysis of 60 normal individuals aged 6weeks to 49years and in 37 patients with schizophrenia/schizoaffective disorder and 37 matched controls. We detected robust opposing changes in cortical GABA(A) receptor alpha1 and alpha5 subunits during the first few years of postnatal development, with a 60% decrease in alpha5 mRNA expression and a doubling of alpha1 mRNA expression with increasing age. In our Australian schizophrenia cohort we detected decreased GAD67 mRNA expression (p=0.0012) and decreased alpha5 mRNA expression (p=0.038) in the DLPFC with no significant change of other alpha subunits. Our findings confirm that GABA deficits (reduced GAD67) are a consistent feature of schizophrenia postmortem brain studies. Our study does not confirm alterations in cortical alpha1 or alpha2 mRNA levels in the schizophrenic DLPFC, as seen in previous studies, but instead we report a novel down-regulation of alpha5 subunit mRNA suggesting that post-synaptic alterations of inhibitory receptors are an important feature of schizophrenia but may vary between cohorts.
Collapse
|
41
|
Venkatasubramanian G, Arasappa R, Rao NP, Behere RV, Kalmady S, Gangadhar BN. Inverse relationship between serum high density lipoprotein and negative syndrome in antipsychotic-naive schizophrenia. Clin Chem Lab Med 2010; 48:95-8. [PMID: 19929755 DOI: 10.1515/cclm.2010.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Recent literature suggests a role for apolipoprotein L (apoL) aberrations in the pathogenesis of schizophrenia. ApoL is almost exclusively associated with apolipoprotein A-I in high-density lipoproteins (HDLs). The objective of this study was to examine the correlation between symptom scores and serum HDL in antipsychotic-naive schizophrenia patients. METHODS In this cross-sectional study, 60 antipsychotic-naive schizophrenia patients were systematically examined for their symptom scores, with good inter-rater reliability. Concurrently, an overnight fasting serum lipid profile from these patients was assessed. RESULTS Serum HDL had a significant inverse correlation with a total negative syndrome score (rho=-0.43; p=0.001). CONCLUSIONS The study observation supports the potential role for HDL abnormalities in the genesis of negative symptoms in schizophrenia.
Collapse
Affiliation(s)
- Ganesan Venkatasubramanian
- The Metabolic Clinic in Psychiatry, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India.
| | | | | | | | | | | |
Collapse
|
42
|
Bousman CA, Chana G, Glatt SJ, Chandler SD, Lucero GR, Tatro E, May T, Lohr JB, Kremen WS, Tsuang MT, Everall IP. Preliminary evidence of ubiquitin proteasome system dysregulation in schizophrenia and bipolar disorder: convergent pathway analysis findings from two independent samples. Am J Med Genet B Neuropsychiatr Genet 2010; 153B:494-502. [PMID: 19582768 PMCID: PMC4165610 DOI: 10.1002/ajmg.b.31006] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Schizophrenia (SCZ) and bipolar disorder (BPD) are polygenic disorders with many genes contributing to their etiologies. The aim of this investigation was to search for dysregulated molecular and cellular pathways for these disorders as well as psychosis. We conducted a blood-based microarray investigation in two independent samples with SCZ and BPD from San Diego (SCZ = 13, BPD = 9, control = 8) and Taiwan (SCZ = 11, BPD = 14, control = 16). Diagnostic groups were compared to controls, and subjects with a history of psychosis [PSYCH(+): San Diego (n = 6), Taiwan (n = 14)] were compared to subjects without such history [PSYCH(-): San Diego (n = 11), Taiwan (n = 14)]. Analyses of covariance comparing mean expression levels on a gene-by-gene basis were conducted to generate the top 100 significantly dysregulated gene lists for both samples by each diagnostic group. Gene lists were imported into Ingenuity Pathway Analysis (IPA) software. Results showed the ubiquitin proteasome pathway (UPS) was listed in the top ten canonical pathways for BPD and psychosis diagnostic groups across both samples with a considerably low likelihood of a chance occurrence (P = 0.001). No overlap in dysregulated genes populating these pathways was observed between the two independent samples. Findings provide preliminary evidence of UPS dysregulation in BPD and psychosis as well as support further investigation of the UPS and other molecular and cellular pathways for potential biomarkers for SCZ, BPD, and/or psychosis.
Collapse
Affiliation(s)
- Chad A. Bousman
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Gursharan Chana
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Stephen J. Glatt
- Department of Psychiatry and Behavioral Sciences, Medical Genetics Research Center, SUNY Upstate Medical University, Syracuse, New York
| | - Sharon D. Chandler
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Ginger R. Lucero
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Erick Tatro
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Todd May
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - James B. Lohr
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - William S. Kremen
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, California,Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Ming T. Tsuang
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, California,Harvard Institute of Psychiatric Epidemiology and Genetics, Harvard Departments of Epidemiology and Psychiatry, Boston, Massachusetts,Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Ian P. Everall
- Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, La Jolla, California,Correspondence to: Ian P. Everall, M.D., Ph.D., Center for Behavioral Genomics, Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92039.
| |
Collapse
|
43
|
Gibbons AS, Thomas EA, Scarr E, Dean B. Low Density Lipoprotein Receptor-Related Protein and Apolipoprotein E Expression is Altered in Schizophrenia. Front Psychiatry 2010; 1:19. [PMID: 21423430 PMCID: PMC3059617 DOI: 10.3389/fpsyt.2010.00019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 06/11/2010] [Indexed: 11/17/2022] Open
Abstract
Our recent microarray study reported altered mRNA expression of several low density lipoprotein receptor-related proteins (LRP) associated with the first 4 years following diagnosis with schizophrenia. Whilst this finding is novel, apolipoprotein E (APOE), which mediates its activity through LRPs, has been reported by several studies to be altered in brains of subjects with schizophrenia. We used qPCR to measure the expression of LRP2, LRP4, LRP6, LRP8, LRP10 and LRP12 mRNA in Brodmann's area (BA) 46 of the dorsolateral prefrontal cortex in 15 subjects with short duration of illness schizophrenia (SDS) and 15 pair matched controls. We also used Western blotting to measure APOE protein expression in BA46 from these subjects. Amongst the LRPs examined, LRP10 expression was significantly increased (P = 0.03) and LRP12 was significantly decreased (P < 0.01) in SDS. APOE protein expression was also increased in SDS (P = 0.01). No other marker examined in this study was altered with diagnosis. Our data supports a role for distinct members of the LRP family in the pathology of schizophrenia and adds weight to the hypothesis that aberrant apolipoprotein signaling is involved in the early stages of schizophrenia.
Collapse
Affiliation(s)
- Andrew Stuart Gibbons
- Rebecca L Cooper Laboratories, Mental Health Research Institute of Victoria Parkville, VIC, Australia
| | | | | | | |
Collapse
|
44
|
Lichen planopilaris and pseudopelade of Brocq involve distinct disease associated gene expression patterns by microarray. J Dermatol Sci 2010; 57:27-36. [DOI: 10.1016/j.jdermsci.2009.10.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 10/19/2009] [Accepted: 10/22/2009] [Indexed: 12/26/2022]
|
45
|
Otowa T, Tanii H, Sugaya N, Yoshida E, Inoue K, Yasuda S, Shimada T, Kawamura Y, Tochigi M, Minato T, Umekage T, Miyagawa T, Nishida N, Tokunaga K, Okazaki Y, Kaiya H, Sasaki T. Replication of a genome-wide association study of panic disorder in a Japanese population. J Hum Genet 2009; 55:91-6. [PMID: 19960027 DOI: 10.1038/jhg.2009.127] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Panic disorder (PD) is an anxiety disorder characterized by recurrent and unexpected panic attacks, subsequent worry and phobic avoidance. Although a number of association and linkage studies have been conducted, no gene has been identified as a susceptibility locus. We previously conducted a genome-wide association analysis of PD in 200 Japanese patients and the same number of controls, using a 500 K single nucleotide polymorphisms (SNPs) chip. In this study, we report a replication analysis of PD using the DigTag2 assay. The second stage sample consisted of 558 Japanese patients and 566 controls. Thirty-two markers were tested in a replication sample. As a result, no significant association was found after correction for multiple testing. However, the difference was observed at the nominal allele P-value <0.05 for two SNPs (rs6733840 and rs132617). We also conducted haplotype analyses of SNPs in the APOL3 and CLU genes. Our results failed to show any significant association with PD in these genes. Further studies on these variants with a larger sample size may be worth testing to confirm the results.
Collapse
Affiliation(s)
- Takeshi Otowa
- Department of Neuropsychiatry, the University of Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gregg JR, Herring NR, Naydenov AV, Hanlin RP, Konradi C. Downregulation of oligodendrocyte transcripts is associated with impaired prefrontal cortex function in rats. Schizophr Res 2009; 113:277-87. [PMID: 19570651 PMCID: PMC2768476 DOI: 10.1016/j.schres.2009.05.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 05/19/2009] [Accepted: 05/22/2009] [Indexed: 11/18/2022]
Abstract
Abnormalities of brain white matter and oligodendroglia are among the most consistent findings in schizophrenia (Sz) research. Various gene expression microarray studies of post-mortem Sz brains showed a downregulation of myelin transcripts, while imaging and microscopy studies demonstrated decreases in prefrontal cortical (PFC) white matter volume and oligodendroglia density. Currently, the extent to which reduced oligodendrocyte markers contribute to pathophysiological domains of Sz is unknown. We exposed adolescent rats to cuprizone (CPZ), a copper chelator known to cause demyelination in mice, and examined expression of oligodendrocyte mRNA transcripts and PFC-mediated behavior. Rats on the CPZ diet showed decreased expression of mRNA transcripts encoding oligodendroglial proteins within the medial PFC, but not in the hippocampus or the striatum. These rats also displayed a specific deficit in the ability to shift between perceptual dimensions in the attentional set-shifting task, a PFC-mediated behavioral paradigm modeled after the Wisconsin Card Sorting Test (WCST). The inability to shift strategies corresponds to the deficits exhibited by Sz patients in the WCST. The results demonstrate that a reduction in oligodendrocyte markers is associated with impaired PFC-mediated behaviors. Thus, CPZ exposure of rats can serve as a model to examine the contribution of oligodendrocyte perturbation to cognitive deficits observed in Sz.
Collapse
Affiliation(s)
- Justin R. Gregg
- Departments of Pharmacology and Psychiatry, Vanderbilt University, Nashville, Tennessee, 37232
| | - Nicole R. Herring
- Departments of Pharmacology and Psychiatry, Vanderbilt University, Nashville, Tennessee, 37232
| | - Alipi V. Naydenov
- Departments of Pharmacology and Psychiatry, Vanderbilt University, Nashville, Tennessee, 37232
| | - Ryan P. Hanlin
- Departments of Pharmacology and Psychiatry, Vanderbilt University, Nashville, Tennessee, 37232
| | - Christine Konradi
- Departments of Pharmacology and Psychiatry, Vanderbilt University, Nashville, Tennessee, 37232
- Center for Molecular Neuroscience, Vanderbilt University, Nashville, Tennessee, 37232
- Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee, 37203
| |
Collapse
|
47
|
Kim WS, Wong J, Weickert CS, Webster MJ, Bahn S, Garner B. Apolipoprotein-D expression is increased during development and maturation of the human prefrontal cortex. J Neurochem 2009; 109:1053-66. [DOI: 10.1111/j.1471-4159.2009.06031.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
48
|
Smith EE, Malik HS. The apolipoprotein L family of programmed cell death and immunity genes rapidly evolved in primates at discrete sites of host-pathogen interactions. Genome Res 2009; 19:850-8. [PMID: 19299565 DOI: 10.1101/gr.085647.108] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Apolipoprotein L1 (APOL1) is a human protein that confers immunity to Trypanosoma brucei infections but can be countered by a trypanosome-encoded antagonist SRA. APOL1 belongs to a family of programmed cell death genes whose proteins can initiate host apoptosis or autophagic death. We report here that all six members of the APOL gene family (APOL1-6) present in humans have rapidly evolved in simian primates. APOL6, furthermore, shows evidence of an adaptive sweep during recent human evolution. In each APOL gene tested, we found rapidly evolving codons in or adjacent to the SRA-interacting protein domain (SID), which is the domain of APOL1 that interacts with SRA. In APOL6, we also found a rapidly changing 13-amino-acid cluster in the membrane-addressing domain (MAD), which putatively functions as a pH sensor and regulator of cell death. We predict that APOL genes are antagonized by pathogens by at least two distinct mechanisms: SID antagonists, which include SRA, that interact with the SID of various APOL proteins, and MAD antagonists that interact with the MAD hinge base of APOL6. These antagonists either block or prematurely cause APOL-mediated programmed cell death of host cells to benefit the infecting pathogen. These putative interactions must occur inside host cells, in contrast to secreted APOL1 that trafficks to the trypanosome lysosome. Hence, the dynamic APOL gene family appears to be an important link between programmed cell death of host cells and immunity to pathogens.
Collapse
Affiliation(s)
- Eric E Smith
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | |
Collapse
|
49
|
Ding L, Hegde AN. Expression of RGS4 splice variants in dorsolateral prefrontal cortex of schizophrenic and bipolar disorder patients. Biol Psychiatry 2009; 65:541-5. [PMID: 19041089 DOI: 10.1016/j.biopsych.2008.10.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Revised: 10/17/2008] [Accepted: 10/22/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Previous molecular and genetic studies have implicated RGS4 (regulator of G protein signaling 4) in schizophrenia (SCZ) and bipolar disorder (BPD), but the role of RGS4 in the pathology of the two disorders remains controversial. Recently we identified five different RGS4 splice variants in the human brain. In this study we tested whether expression of specific RGS4 splice variants is altered in the prefrontal cortex of schizophrenic and BPD subjects. METHODS Quantitative real-time polymerase chain reaction was used to detect overall RGS4 expression and the messenger RNA levels of the four RGS4 splice variants in the prefrontal cortex of schizophrenic (n = 27), BPD (n = 27), and normal (n = 27) subjects. RESULTS Compared with the normal group, the expression of a specific splice variant RGS4-3 was decreased in the dorsolateral prefrontal cortex of the SCZ group, whereas overall RGS4 expression and expression of other RGS4 isoforms did not differ significantly between the control and SCZ groups. The messenger RNA levels of RGS4 isoforms did not change between the control group and the BPD group. CONCLUSIONS Our results suggest the possibility that alterations in the expression of RGS4-3 contribute to the development of SCZ.
Collapse
Affiliation(s)
- Lan Ding
- Department of Neurobiology and Anatomy, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, North Carolina 27157, USA
| | | |
Collapse
|
50
|
Independent protein-profiling studies show a decrease in apolipoprotein A1 levels in schizophrenia CSF, brain and peripheral tissues. Mol Psychiatry 2008; 13:1118-28. [PMID: 17938634 DOI: 10.1038/sj.mp.4002108] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Although some insights into the etiology of schizophrenia have been gained, an understanding of the illness at the molecular level remains elusive. Recent advances in proteomic profiling offer great promise for the discovery of markers underlying pathophysiology of diseases. In the present study, we employed two high-throughput proteomic techniques together with traditional methods to investigate cerebrospinal fluid (CSF), brain and peripheral tissues (liver, red blood cells and serum) of schizophrenia patients in an attempt to identify peripheral/surrogate disease markers. The cohorts used to investigate each tissue were largely independent, although some CSF and serum samples were collected from the same patient. To address the major confounding factor of antipsychotic drug treatment, we also included a large cohort of first-onset drug-naive patients. Apolipoprotein A1 (apoA1) showed a significant decrease in expression in schizophrenia patients compared to controls in all five tissues examined. Specifically, using SELDI-TOF mass spectrometry, apoA1 was found decreased in CSF from schizophrenia patients (-35%, P=0.00001) and, using 2D-DIGE, apoA1 was also found downregulated in liver (-30%, P=0.02) and RBCs (-60%, P=0.003). Furthermore, we found a significant reduction of apoA1 in sera of first-onset drug-naive schizophrenia patients using enzyme-linked immunosorbent assay (-18%, P=0.00008) and in two investigations of post-mortem brain tissue using western blot analysis (-35%, P=0.05; -51%, P=0.05). These results show that apoA1 is consistently downregulated in the central nervous system as well as peripheral tissues of schizophrenia patients and may be linked to the underlying disease mechanism.
Collapse
|