1
|
Liu A, Zhao Y, Shen H, Ding Z, Deng HW. ResSAT: Enhancing Spatial Transcriptomics Prediction from H&E- Stained Histology Images with Interactive Spot Transformer. RESEARCH SQUARE 2024:rs.3.rs-4707959. [PMID: 39149477 PMCID: PMC11326376 DOI: 10.21203/rs.3.rs-4707959/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Spatial transcriptomics (ST) revolutionizes RNA quantification with high spatial resolution. Hematoxylin and eosin (H&E) images, the gold standard in medical diagnosis, offer insights into tissue structure, correlating with gene expression patterns. Current methods for predicting spatial gene expression from H&E images often overlook spatial relationships. We introduce ResSAT (Residual networks - Self-Attention Transformer), a framework generating spatially resolved gene expression profiles from H&E images by capturing tissue structures and using a self-attention transformer to enhance prediction.Benchmarking on 10× Visium datasets, ResSAT significantly outperformed existing methods, promising reduced ST profiling costs and rapid acquisition of numerous profiles.
Collapse
|
2
|
Mey M, Bhatta S, Suresh S, Labrador LM, Piontkivska H, Casadesus G. Therapeutic benefits of central LH receptor agonism in the APP/PS1 AD model involve trophic and immune regulation and are reproductive status dependent. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167165. [PMID: 38653355 DOI: 10.1016/j.bbadis.2024.167165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
The mechanisms that underly reproductive hormone effects on cognition, neuronal plasticity, and AD risk, particularly in relation to gonadotropin LH receptor (LHCGR) signaling, remain poorly understood. To address this gap in knowledge and clarify the impact of circulating steroid hormones on the therapeutic effects of CNS LHCGR activation, we delivered the LHCGR agonist human chorionic gonadotropin (hCG) intracerebroventricularly (ICV) and evaluated functional, structural, plasticity-related signaling cascades, Aβ pathology, and transcriptome differences in reproductively intact and ovariectomized (OVX) APP/PS1 AD female mice. Here we demonstrate that CNS hCG delivery restored function to wild-type levels only in OVX APP/PS1 mice. Spine density was increased in all hCG treated groups independently of reproductive status. Notably, increases in BDNF signaling and cognition, were selectively upregulated only in the OVX hCG-treated group. RNA sequencing analyses identified a significant increase in peripheral myeloid and pro-inflammatory genes within the hippocampi of the OVX group that were completely reversed by hCG treatment, identifying a potential mechanism underlying the selective therapeutic effect of LHCGR activation. Interestingly, in intact mice, hCG administration mimicked the effects of gonadectomy. Together, our findings indicate that CNS LHCGR agonism in the post-menopausal context is beneficial through trophic and immune mechanisms. Our findings also underscore the presence of a steroid-LHCGR mechanistic interaction that is unexplored yet potentially meaningful to fully understand "post-menopausal" brain function and CNS hormone treatment response.
Collapse
Affiliation(s)
- Megan Mey
- Kent State University, Kent, OH 44240, United States of America
| | - Sabina Bhatta
- Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Sneha Suresh
- University of Florida, Gainesville, FL 32606, United States of America
| | | | | | - Gemma Casadesus
- University of Florida, Gainesville, FL 32606, United States of America.
| |
Collapse
|
3
|
Sunila BG, Dhanushkumar T, Dasegowda KR, Vasudevan K, Rambabu M. Unraveling the molecular landscape of Ataxia Telangiectasia: Insights into Neuroinflammation, immune dysfunction, and potential therapeutic target. Neurosci Lett 2024; 828:137764. [PMID: 38582325 DOI: 10.1016/j.neulet.2024.137764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/23/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND Ataxia Telangiectasia (AT) is a genetic disorder characterized by compromised DNA repair, cerebellar degeneration, and immune dysfunction. Understanding the molecular mechanisms driving AT pathology is crucial for developing targeted therapies. METHODS In this study, we conducted a comprehensive analysis to elucidate the molecular mechanisms underlying AT pathology. Using publicly available RNA-seq datasets comparing control and AT samples, we employed in silico transcriptomics to identify potential genes and pathways. We performed differential gene expression analysis with DESeq2 to reveal dysregulated genes associated with AT. Additionally, we constructed a Protein-Protein Interaction (PPI) network to explore the interactions between proteins implicated in AT. RESULTS The network analysis identified hub genes, including TYROBP and PCP2, crucial in immune regulation and cerebellar function, respectively. Furthermore, pathway enrichment analysis unveiled dysregulated pathways linked to AT pathology, providing insights into disease progression. CONCLUSION Our integrated approach offers a holistic understanding of the complex molecular landscape of AT and identifies potential targets for therapeutic intervention. By combining transcriptomic analysis with network-based methods, we provide valuable insights into the underlying mechanisms of AT pathogenesis.
Collapse
Affiliation(s)
- B G Sunila
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - T Dhanushkumar
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - K R Dasegowda
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Karthick Vasudevan
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Majji Rambabu
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India.
| |
Collapse
|
4
|
Cooper CP, Cheng L, Bhatti J, Melendez ER, Huell D, Banuelos C, Perez E, Long JM, Rapp PR. Cerebellum Purkinje cell vulnerability in aged rats with memory impairment. J Comp Neurol 2024; 532:e25610. [PMID: 38605461 PMCID: PMC11027960 DOI: 10.1002/cne.25610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/22/2024] [Accepted: 03/24/2024] [Indexed: 04/13/2024]
Abstract
The cerebellum is involved in higher order cognitive function and is susceptible to age-related atrophy. However, limited evidence has directly examined the cerebellum's role in cognitive aging. To interrogate potential substrates of the relationship between cerebellar structure and memory in aging, here we target the Purkinje cells (PCs). The sole output neurons of the cerebellum, PC loss and/or degeneration underlie a variety of behavioral abnormalities. Using a rat model of normal cognitive aging, we immunostained sections through the cerebellum for the PC-specific protein, calbindin-D28k. Although morphometric quantification revealed no significant difference in total PC number as a function of age or cognitive status, regional cell number was a more robust correlate of memory performance in the young cerebellum than in aged animals. Parallel biochemical analysis of PC-specific protein levels in whole cerebellum additionally revealed that calbindin-D28k and Purkinje cell protein-2 (pcp-2) levels were lower selectively in aged rats with spatial memory impairment compared to both young animals and aged rats with intact memory. These results suggest that cognitive aging is associated with cerebellum vulnerability, potentially reflecting disruption of the cerebellum-medial temporal lobe network.
Collapse
Affiliation(s)
- C’iana P. Cooper
- Neurocognitive Aging Section, Laboratory of Behavioral
Neuroscience, National Institute on Aging, Baltimore, Maryland
| | - Liam Cheng
- Neurocognitive Aging Section, Laboratory of Behavioral
Neuroscience, National Institute on Aging, Baltimore, Maryland
| | - Jafar Bhatti
- Neurocognitive Aging Section, Laboratory of Behavioral
Neuroscience, National Institute on Aging, Baltimore, Maryland
| | - Edward R. Melendez
- Neurocognitive Aging Section, Laboratory of Behavioral
Neuroscience, National Institute on Aging, Baltimore, Maryland
| | - Derek Huell
- Neurocognitive Aging Section, Laboratory of Behavioral
Neuroscience, National Institute on Aging, Baltimore, Maryland
| | - Cristina Banuelos
- Neurocognitive Aging Section, Laboratory of Behavioral
Neuroscience, National Institute on Aging, Baltimore, Maryland
| | - Evelyn Perez
- Neurocognitive Aging Section, Laboratory of Behavioral
Neuroscience, National Institute on Aging, Baltimore, Maryland
| | - Jeffrey M. Long
- Neurocognitive Aging Section, Laboratory of Behavioral
Neuroscience, National Institute on Aging, Baltimore, Maryland
| | - Peter R. Rapp
- Neurocognitive Aging Section, Laboratory of Behavioral
Neuroscience, National Institute on Aging, Baltimore, Maryland
| |
Collapse
|
5
|
Zhang C, Gao J, Chen HY, Kong L, Cao G, Guo X, Liu W, Ren B, Wei DQ. STGIC: A graph and image convolution-based method for spatial transcriptomic clustering. PLoS Comput Biol 2024; 20:e1011935. [PMID: 38416785 PMCID: PMC10927115 DOI: 10.1371/journal.pcbi.1011935] [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: 10/31/2023] [Revised: 03/11/2024] [Accepted: 02/20/2024] [Indexed: 03/01/2024] Open
Abstract
Spatial transcriptomic (ST) clustering employs spatial and transcription information to group spots spatially coherent and transcriptionally similar together into the same spatial domain. Graph convolution network (GCN) and graph attention network (GAT), fed with spatial coordinates derived adjacency and transcription profile derived feature matrix are often used to solve the problem. Our proposed method STGIC (spatial transcriptomic clustering with graph and image convolution) is designed for techniques with regular lattices on chips. It utilizes an adaptive graph convolution (AGC) to get high quality pseudo-labels and then resorts to dilated convolution framework (DCF) for virtual image converted from gene expression information and spatial coordinates of spots. The dilation rates and kernel sizes are set appropriately and updating of weight values in the kernels is made to be subject to the spatial distance from the position of corresponding elements to kernel centers so that feature extraction of each spot is better guided by spatial distance to neighbor spots. Self-supervision realized by Kullback-Leibler (KL) divergence, spatial continuity loss and cross entropy calculated among spots with high confidence pseudo-labels make up the training objective of DCF. STGIC attains state-of-the-art (SOTA) clustering performance on the benchmark dataset of 10x Visium human dorsolateral prefrontal cortex (DLPFC). Besides, it's capable of depicting fine structures of other tissues from other species as well as guiding the identification of marker genes. Also, STGIC is expandable to Stereo-seq data with high spatial resolution.
Collapse
Affiliation(s)
- Chen Zhang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Junhui Gao
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hong-Yu Chen
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Lingxin Kong
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Guangshuo Cao
- State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang
| | - Xiangyu Guo
- Smart-Health Initiative, King Abdullah University of Science and Technology, Jeddah, Saudi Arabia
| | - Wei Liu
- Marine Science and Technology College, Zhejiang Ocean University, Zhoushan, China
| | - Bin Ren
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Dong-Qing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
6
|
Mey M, Bhatta S, Suresh S, Montero Labrador L, Piontkivska H, Casadesus G. The LH receptor regulates hippocampal spatial memory and restores dendritic spine density in ovariectomized APP/PS1 AD mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573087. [PMID: 38187770 PMCID: PMC10769359 DOI: 10.1101/2023.12.22.573087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Activation of the luteinizing hormone receptor (LHCGR) rescues spatial memory function and spine density losses associated with gonadectomy and high circulating gonadotropin levels in females. However, whether this extends to the AD brain or the mechanisms that underlie these benefits remain unknown. To address this question, we delivered the LHCGR agonist human chorionic gonadotropin (hCG) intracerebroventricularly (ICV), under reproductively intact and ovariectomized conditions to mimic the post-menopausal state in the APP/PS1mouse brain. Cognitive function was tested using the Morris water maze task, and hippocampal dendritic spine density, Aβ pathology, and signaling changes associated with these endpoints were determined to address mechanisms. Here we show that central LHCGR activation restored function in ovariectomized APP/PS1 female mice to wild-type levels without altering Aβ pathology. LHCGR activation increased hippocampal dendritic spine density regardless of reproductive status, and this was mediated by BDNF-dependent and independent signaling. We also show that ovariectomy in the APP/PS1 brain elicits an increase in peripherally derived pro-inflammatory genes which are inhibited by LHCGR activation. This may mediate reproductive status specific effects of LHCGR agonism on cognitive function and BDNF expression. Together, this work highlights the relevance of the LHCGR on cognition and its therapeutic potential in the "menopausal" AD brain.
Collapse
|
7
|
Rodriguez-Duboc A, Basille-Dugay M, Debonne A, Rivière MA, Vaudry D, Burel D. Apnea of prematurity induces short and long-term development-related transcriptional changes in the murine cerebellum. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100113. [PMID: 38020806 PMCID: PMC10663136 DOI: 10.1016/j.crneur.2023.100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Apnea of prematurity (AOP) affects more than 50% of preterm infants and leads to perinatal intermittent hypoxia (IH) which is a major cause of morbimortality worldwide. At birth, the human cerebellar cortex is still immature, making it vulnerable to perinatal events. Additionally, studies have shown a correlation between cerebellar functions and the deficits observed in children who have experienced AOP. Yet, the cerebellar alterations underpinning this link remain poorly understood. To gain insight into the involvement of the cerebellum in perinatal hypoxia-related consequences, we developed a mouse model of AOP. Our previous research has revealed that IH induces oxidative stress in the developing cerebellum, as evidenced by the over-expression of genes involved in reactive oxygen species production and the under-expression of genes encoding antioxidant enzymes. These changes suggest a failure of the defense system against oxidative stress and could be responsible for neuronal death in the cerebellum. Building upon these findings, we conducted a transcriptomic study of the genes involved in the processes that occur during cerebellar development. Using real-time PCR, we analyzed the expression of these genes at different developmental stages and in various cell types. This enabled us to pinpoint a timeframe of vulnerability at P8, which represents the age with the highest number of downregulated genes in the cerebellum. Furthermore, we discovered that our IH protocol affects several molecular pathways, including proliferation, migration, and differentiation. This indicates that IH can impact the development of different cell types, potentially contributing to the histological and behavioral deficits observed in this model. Overall, our data strongly suggest that the cerebellum is highly sensitive to IH, and provide valuable insights into the cellular and molecular mechanisms underlying AOP. In the long term, these findings may contribute to the identification of novel therapeutic targets for improving the clinical management of this prevalent pathology.
Collapse
Affiliation(s)
- A. Rodriguez-Duboc
- Univ Rouen Normandie, Inserm, U1245, Normandie Univ, F-76000, Rouen, France
| | - M. Basille-Dugay
- Univ Rouen Normandie, Inserm, U1239, Normandie Univ, F-76000, Rouen, France
| | - A. Debonne
- Univ Rouen Normandie, Inserm, U1245, Normandie Univ, F-76000, Rouen, France
- Univ Rouen Normandie, INSERM, CNRS, HeRacLeS US 51 UAR 2026, PRIMACEN, Normandie Univ, F-76000, Rouen, France
| | - M.-A. Rivière
- Univ Rouen Normandie, UR 4108, LITIS Lab, INSA Rouen, NormaSTIC, CNRS 3638, Normandie Univ, F-76000, Rouen, France
| | - D. Vaudry
- Univ Rouen Normandie, Inserm, U1245, Normandie Univ, F-76000, Rouen, France
- Univ Rouen Normandie, INSERM, CNRS, HeRacLeS US 51 UAR 2026, PRIMACEN, Normandie Univ, F-76000, Rouen, France
| | - D. Burel
- Univ Rouen Normandie, Inserm, U1245, Normandie Univ, F-76000, Rouen, France
- Univ Rouen Normandie, INSERM, CNRS, HeRacLeS US 51 UAR 2026, PRIMACEN, Normandie Univ, F-76000, Rouen, France
| |
Collapse
|
8
|
Whittaker DE, Oleari R, Gregory LC, Le Quesne-Stabej P, Williams HJ, Torpiano JG, Formosa N, Cachia MJ, Field D, Lettieri A, Ocaka LA, Paganoni AJ, Rajabali SH, Riegman KL, De Martini LB, Chaya T, Robinson IC, Furukawa T, Cariboni A, Basson MA, Dattani MT. A recessive PRDM13 mutation results in congenital hypogonadotropic hypogonadism and cerebellar hypoplasia. J Clin Invest 2021; 131:e141587. [PMID: 34730112 PMCID: PMC8670848 DOI: 10.1172/jci141587] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
The positive regulatory (PR) domain containing 13 (PRDM13) putative chromatin modifier and transcriptional regulator functions downstream of the transcription factor PTF1A, which controls GABAergic fate in the spinal cord and neurogenesis in the hypothalamus. Here, we report a recessive syndrome associated with PRDM13 mutation. Patients exhibited intellectual disability, ataxia with cerebellar hypoplasia, scoliosis, and delayed puberty with congenital hypogonadotropic hypogonadism (CHH). Expression studies revealed Prdm13/PRDM13 transcripts in the developing hypothalamus and cerebellum in mouse and human. An analysis of hypothalamus and cerebellum development in mice homozygous for a Prdm13 mutant allele revealed a significant reduction in the number of Kisspeptin (Kiss1) neurons in the hypothalamus and PAX2+ progenitors emerging from the cerebellar ventricular zone. The latter was accompanied by ectopic expression of the glutamatergic lineage marker TLX3. Prdm13-deficient mice displayed cerebellar hypoplasia and normal gonadal structure, but delayed pubertal onset. Together, these findings identify PRDM13 as a critical regulator of GABAergic cell fate in the cerebellum and of hypothalamic kisspeptin neuron development, providing a mechanistic explanation for the cooccurrence of CHH and cerebellar hypoplasia in this syndrome. To our knowledge, this is the first evidence linking disrupted PRDM13-mediated regulation of Kiss1 neurons to CHH in humans.
Collapse
Affiliation(s)
- Danielle E. Whittaker
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, United Kingdom
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Louise C. Gregory
- Section of Molecular Basis of Rare Disease, Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Polona Le Quesne-Stabej
- Section of Molecular Basis of Rare Disease, Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Hywel J. Williams
- Section of Molecular Basis of Rare Disease, Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - GOSgene
- Section of Molecular Basis of Rare Disease, Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- GOSgene is detailed in Supplemental Acknowledgments
| | - John G. Torpiano
- Department of Paediatrics and
- Adult Endocrinology Service, Mater Dei Hospital, Msida, Malta
| | | | - Mario J. Cachia
- Adult Endocrinology Service, Mater Dei Hospital, Msida, Malta
| | - Daniel Field
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, United Kingdom
| | - Antonella Lettieri
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Louise A. Ocaka
- Section of Molecular Basis of Rare Disease, Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Alyssa J.J. Paganoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Sakina H. Rajabali
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, United Kingdom
| | - Kimberley L.H. Riegman
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, United Kingdom
| | - Lisa B. De Martini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Taro Chaya
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | | | - Takahisa Furukawa
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - M. Albert Basson
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Mehul T. Dattani
- Section of Molecular Basis of Rare Disease, Genetics and Genomic Medicine Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
9
|
Biswas D, Shenoy SV, Chetanya C, Lachén-Montes M, Barpanda A, Athithyan AP, Ghosh S, Ausín K, Zelaya MV, Fernández-Irigoyen J, Manna A, Roy S, Talukdar A, Ball GR, Santamaría E, Srivastava S. Deciphering the Interregional and Interhemisphere Proteome of the Human Brain in the Context of the Human Proteome Project. J Proteome Res 2021; 20:5280-5293. [PMID: 34714085 DOI: 10.1021/acs.jproteome.1c00511] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This study, which performs an extensive mass spectrometry-based analysis of 19 brain regions from both left and right hemispheres, presents the first draft of the human brain interhemispheric proteome. This high-resolution proteomics data provides comprehensive coverage of 3300 experimentally measured (nonhypothetical) proteins across multiple regions, allowing the characterization of protein-centric interhemispheric differences and synapse biology, and portrays the regional mapping of specific regions for brain disorder biomarkers. In the context of the Human Proteome Project (HPP), the interhemispheric proteome data reveal specific markers like chimerin 2 (CHN2) in the cerebellar vermis, olfactory marker protein (OMP) in the olfactory bulb, and ankyrin repeat domain 63 (ANKRD63) in basal ganglia, in line with regional brain transcriptomes mapped in the Human Protein Atlas (HPA). In addition, an in silico analysis pipeline was used to predict the structure and function of the uncharacterized uPE1 protein ANKRD63, and parallel reaction monitoring (PRM) was applied to validate its region-specific expression. Finally, we have built the Interhemispheric Brain Proteome Map (IBPM) Portal (www.brainprot.org) to stimulate the scientific community's interest in the brain molecular landscape and accelerate and support research in neuroproteomics. Data are available via ProteomeXchange with identifier PXD019936.
Collapse
Affiliation(s)
- Deeptarup Biswas
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Sanjyot Vinayak Shenoy
- Department of Mathematics, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Chetanya Chetanya
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Mercedes Lachén-Montes
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Abhilash Barpanda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | | | - Susmita Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Karina Ausín
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - María Victoria Zelaya
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Akash Manna
- Medicine Department, Medical College Hospital Kolkata, 88 College Street, Kolkata 700072, India
| | - Sudesh Roy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Arunasu Talukdar
- Medicine Department, Medical College Hospital Kolkata, 88 College Street, Kolkata 700072, India
| | - Graham Roy Ball
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, United Kingdom
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
10
|
Identifying GPSM Family Members as Potential Biomarkers in Breast Cancer: A Comprehensive Bioinformatics Analysis. Biomedicines 2021; 9:biomedicines9091144. [PMID: 34572330 PMCID: PMC8471503 DOI: 10.3390/biomedicines9091144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
G-protein signaling modulators (GPSMs) are a class of proteins involved in the regulation of G protein-coupled receptors, the most abundant family of cell-surface receptors that are crucial in the development of various tumors, including breast cancer. This study aims to identify the potential therapeutic and prognostic roles of GPSMs in breast cancer. Oncomine and UALCAN databases were queried to determine GPSM expression levels in breast cancer tissues compared to normal samples. Survival analysis was conducted to reveal the prognostic significance of GPSMs in individuals with breast cancer. Functional enrichment analysis was performed using cBioPortal and MetaCore platforms. Finally, the association between GPSMs and immune infiltration cells in breast cancer was identified using the TIMER server. The experimental results then showed that all GPSM family members were significantly differentially expressed in breast cancer according to Oncomine and UALCAN data. Their expression levels were also associated with advanced tumor stages, and GPSM2 was found to be related to worse distant metastasis-free survival in patients with breast cancer. Functional enrichment analysis indicated that GPSMs were largely involved in cell division and cell cycle pathways. Finally, GPSM3 expression was correlated with the infiltration of several immune cells. Members of the GPSM class were differentially expressed in breast cancer. In conclusion, expression of GPSM2 was linked with worse distant metastasis-free outcomes, and hence could potentially serve as a prognostic biomarker. Furthermore, GPSM3 has potential to be a possible target for immunotherapy for breast cancer.
Collapse
|
11
|
Li S, He B, Yang C, Yang J, Wang L, Duan X, Deng X, Zhao J, Fang R. Comparative transcriptome analysis of normal and CD44-deleted mouse brain under chronic infection with Toxoplasma gondii. Acta Trop 2020; 210:105589. [PMID: 32544399 DOI: 10.1016/j.actatropica.2020.105589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022]
Abstract
Toxoplasma gondii is a globally-distributed intracellular parasitic protozoon with wide host range. Chronic infection is the most prevalent form of T. gondii infection, which can lead to significant damage. CD44 plays an important role in body's immune response, however, little is known about the function and mechanism of CD44 in T. gondii infection until now. In the present study, total RNA isolated from four groups including C57BL/6 mouse (C57), C57BL/6△CD44 mouse(C57△CD44), C57BL/6 mouse infected with T. gondii (C57-TG) and C57BL/6△CD44 infected with T. gondii (C57△CD44-TG)were subjected to comparative transcriptome analyses using RNA-seq techniques to explore the possible function of CD44 in mouse brain during chronic Toxoplasma infection. The results indicated a total of 35,908, 54,428, 51,473 and 22,387 unigenes were annotated in KOG, Swissprot, GO and KEGG databases by transcriptome analysis, respectively, and all the databases shared 9,833 unigenes. Subsequently, differentially expressed GO terms and enriched KEGG Pathways showed 20,303 unigenes were annotated belonging to three main GO categories (namely biological process, cellular component and molecular function) and six main KEGG categories (cellular processes, environmental information processing, genetic information processing, human diseases, metabolism and organismal systems) between normal C57 and C57△CD44 mice, as well as for C57-TG and C57△CD44-TG mice. For up-regulated genes, Mid1, Ttr and Cd4 were significantly up-regulated in the C57△CD44 mouse compared with the C57 mouse, and Pcp2, Ppp1r17 and Nrk were significantly up-regulated in the C57△CD44-TG mouse compared with the C57-TG mouse. As to down-regulated genes, AC114588.1, Cbln3 and Pmch were significantly down-regulated in the C57△CD44 the mouse compared with the C57 mouse, and down-regulated genes were enriched for immunoglobulins, major histocompatibility complex (MHC) class II antigens, chemokines ligands and interferon (IFN)-inducible GTPase families in the C57△CD44-TG mouse compared with the C57-TG mouse. The present study is the first trial for exploring the function of CD44 in the mouse brain during chronic infection with T. gondii at the transcriptional level, which can provide a basis for the study of the host immune defense mechanism against T. gondii infection.
Collapse
Affiliation(s)
- Senyang Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province 430070, PR China
| | - Bin He
- Wuhan Academy of Agricultural Sciences, PR China
| | - Chenghang Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province 430070, PR China
| | - Jing Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province 430070, PR China.
| | - Lixia Wang
- Hubei Provincial Center for Diseases Control and Prevention, Wuhan 430079, Hubei, PR China
| | - Xi Duan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province 430070, PR China.
| | - Xiaokun Deng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province 430070, PR China.
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province 430070, PR China.
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, Hubei Province 430070, PR China.
| |
Collapse
|
12
|
Francescatto M, Lizio M, Philippens I, Pardo LM, Bontrop R, Sakai M, Watanabe S, Itoh M, Hasegawa A, Lassmann T, Severin J, Harshbarger J, Abugessaisa I, Kasukawa T, Carninci P, Hayashizaki Y, Forrest ARR, Kawaji H, Rizzu P, Heutink P. Transcription start site profiling of 15 anatomical regions of the Macaca mulatta central nervous system. Sci Data 2017; 4:170163. [PMID: 29087374 PMCID: PMC5663209 DOI: 10.1038/sdata.2017.163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 08/29/2017] [Indexed: 01/10/2023] Open
Abstract
Rhesus macaque was the second non-human primate whose genome has been fully
sequenced and is one of the most used model organisms to study human biology and
disease, thanks to the close evolutionary relationship between the two species.
But compared to human, where several previously unknown RNAs have been
uncovered, the macaque transcriptome is less studied. Publicly available RNA
expression resources for macaque are limited, even for brain, which is highly
relevant to study human cognitive abilities. In an effort to complement those
resources, FANTOM5 profiled 15 distinct anatomical regions of the aged macaque
central nervous system using Cap Analysis of Gene Expression, a high-resolution,
annotation-independent technology that allows monitoring of transcription
initiation events with high accuracy. We identified 25,869 CAGE peaks,
representing bona fide promoters. For each peak we provide detailed annotation,
expanding the landscape of ‘known’ macaque genes, and we show
concrete examples on how to use the resulting data. We believe this data
represents a useful resource to understand the central nervous system in
macaque.
Collapse
Affiliation(s)
- Margherita Francescatto
- Italian Institute of Technology, Department of Neuroscience and Brain Technologies, Via Morego 30, Genova 16163, Italy
| | - Marina Lizio
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Ingrid Philippens
- Biomedical Primate Research Centre, Postbox 3306, Rijswijk 2280 GH, The Netherlands
| | | | - Ronald Bontrop
- Biomedical Primate Research Centre, Postbox 3306, Rijswijk 2280 GH, The Netherlands
| | - Mizuho Sakai
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Shoko Watanabe
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Masayoshi Itoh
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Preventive Medicine and Diagnosis Innovation Program, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Akira Hasegawa
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Timo Lassmann
- RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,Telethon Kids Institute, The University of Western Australia, 100 Roberts Road, Subiaco, Western Australia 6008, Australia
| | - Jessica Severin
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Jayson Harshbarger
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Imad Abugessaisa
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Takeya Kasukawa
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Piero Carninci
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Yoshihide Hayashizaki
- RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Preventive Medicine and Diagnosis Innovation Program, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Alistair R R Forrest
- RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,Harry Perkins Institute of Medical Research, 6 Verdun St, Nedlands, Western Australia 6009, Australia
| | - Hideya Kawaji
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Yokohama Institute, Omics Science Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Preventive Medicine and Diagnosis Innovation Program, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan.,RIKEN Advanced Center for Computing and Communication, Preventive Medicine and Applied Genomics Unit, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Patrizia Rizzu
- German Center for Neurodegenerative Diseases, Otfried-Müller Straße 23, Tübingen 72076, Germany
| | - Peter Heutink
- German Center for Neurodegenerative Diseases, Otfried-Müller Straße 23, Tübingen 72076, Germany
| |
Collapse
|
13
|
Somatostatin activates Ras and ERK1/2 via a G protein βγ-subunit-initiated pathway in thyroid cells. Mol Cell Biochem 2015; 411:253-60. [PMID: 26472731 DOI: 10.1007/s11010-015-2587-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/08/2015] [Indexed: 10/22/2022]
Abstract
Somatostatin (SST) is one of the main regulators of thyroid function. It acts by binding to its receptors, which lead to the dissociation of G proteins into Gαi and Gβγ subunits. However, much less is known about the function of Gβγ in thyroid cells. Here, we studied the role of SST and Gβγ dimers released upon SST stimulation on the Ras-ERK1/2 pathway in FTRL-5 thyroid cells. We demonstrate that SST activates Ras through Gi proteins, since SST-induced Ras activation is inhibited by pertussis toxin. Moreover, the specific sequestration of Gβγ dimers decreases Ras-GTP and phosphorylated ERK1/2 levels, and overexpression of Gβγ increases ERK1/2 phosphorylation induced by SST, indicating that Gβγ dimers released after SST treatment mediate activation of Ras and ERK1/2. On the other hand, SST treatment does not modify the expression of the thyroid differentiation marker sodium/iodide symporter (NIS) through ERK1/2 activation. However, SST increases AKT activation and the inhibition of the Src/PI3K/AKT pathway increases NIS levels in SST-treated cells. Thus, we conclude that, in thyroid cells, signalling from SST receptors to ERK1/2 involves a Gβγ-mediated signal acting on a Ras-dependent pathway. Moreover, we demonstrate that SST might regulates NIS expression through a Src/PI3K/AKT-dependent mechanism, but not through ERK1/2 signalling, showing the main role of this hormone in thyroid function.
Collapse
|
14
|
Greenlee JE, Clawson SA, Hill KE, Wood B, Clardy SL, Tsunoda I, Carlson NG. Anti-Yo antibody uptake and interaction with its intracellular target antigen causes Purkinje cell death in rat cerebellar slice cultures: a possible mechanism for paraneoplastic cerebellar degeneration in humans with gynecological or breast cancers. PLoS One 2015; 10:e0123446. [PMID: 25885452 PMCID: PMC4401511 DOI: 10.1371/journal.pone.0123446] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 03/03/2015] [Indexed: 02/05/2023] Open
Abstract
Anti-Yo antibodies are immunoglobulin G (IgG) autoantibodies reactive with a 62 kDa Purkinje cell cytoplasmic protein. These antibodies are closely associated with paraneoplastic cerebellar degeneration in the setting of gynecological and breast malignancies. We have previously demonstrated that incubation of rat cerebellar slice cultures with patient sera and cerebrospinal fluid containing anti-Yo antibodies resulted in Purkinje cell death. The present study addressed three fundamental questions regarding the role of anti-Yo antibodies in disease pathogenesis: 1) Whether the Purkinje cell cytotoxicity required binding of anti-Yo antibody to its intraneuronal 62 kDa target antigen; 2) whether Purkinje cell death might be initiated by antibody-dependent cellular cytotoxicity rather than intracellular antibody binding; and 3) whether Purkinje cell death might simply be a more general result of intracellular antibody accumulation, rather than of specific antibody-antigen interaction. In our study, incubation of rat cerebellar slice cultures with anti-Yo IgG resulted in intracellular antibody binding, and cell death. Infiltration of the Purkinje cell layer by cells of macrophage/microglia lineage was not observed until extensive cell death was already present. Adsorption of anti-Yo IgG with its 62 kDa target antigen abolished both antibody accumulation and cytotoxicity. Antibodies to other intracellular Purkinje cell proteins were also taken up by Purkinje cells and accumulated intracellularly; these included calbindin, calmodulin, PCP-2, and patient anti-Purkinje cell antibodies not reactive with the 62 kDa Yo antigen. However, intracellular accumulation of these antibodies did not affect Purkinje cell viability. The present study is the first to demonstrate that anti-Yo antibodies cause Purkinje cell death by binding to the intracellular 62 kDa Yo antigen. Anti-Yo antibody cytotoxicity did not involve other antibodies or factors present in patient serum and was not initiated by brain mononuclear cells. Purkinje cell death was not simply due to intraneuronal antibody accumulation.
Collapse
Affiliation(s)
- John E Greenlee
- Neurology Service, George E. Wahlen Veterans Affairs Health Care System, Salt Lake City, Utah, United States of America; Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Susan A Clawson
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Kenneth E Hill
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Blair Wood
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Stacey L Clardy
- Neurology Service, George E. Wahlen Veterans Affairs Health Care System, Salt Lake City, Utah, United States of America; Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Ikuo Tsunoda
- Department of Microbiology and Immunology Center for Molecular and Tumor Virology (CMTV), Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Noel G Carlson
- Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America; Geriatric Research, Education, and Care Center (GRECC), George E. Wahlen Veterans Affairs Health Care System, Salt Lake City, Utah, United States of America; Research Service, George E. Wahlen Veterans Affairs Health Care System, Salt Lake City, Utah, United States of America; Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| |
Collapse
|
15
|
Ju X, Wen Y, Metzger D, Jung M. The role of p38 in mitochondrial respiration in male and female mice. Neurosci Lett 2013; 544:152-6. [PMID: 23603578 DOI: 10.1016/j.neulet.2013.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 03/29/2013] [Accepted: 04/10/2013] [Indexed: 01/28/2023]
Abstract
p38 is a mitogen-activated protein kinase and mediates cell growth, cell differentiation, and synaptic plasticity. The aim of this study is to determine the extent to which p38 plays a role in maintaining mitochondrial respiration in male and female mice under a normal condition. To achieve this aim, we have generated transgenic mice that lack p38 in cerebellar Purkinje neurons by crossing Pcp2 (Purkinje cell protein 2)-Cre mice with p38(loxP/loxP) mice. Mitochondria from cerebellum were then isolated from the transgenic and wild-type mice to measure mitochondrial respiration using XF24 respirometer. The mRNA and protein expression of cytochrome c oxidase (COX) in cerebellum were also measured using RT-PCR and immunoblot methods. Separately, HT22 cells were used to determine the involvement of 17β-estradiol (E2) and COX in mitochondrial respiration. The genetic knockout of p38 in Purkinje neurons suppressed the mitochondrial respiration only in male mice and increased COX expression only in female mice. The inhibition of COX by sodium azide (SA) sharply suppressed mitochondrial respiration of HT22 cells in a manner that was protected by E2. These data suggest that p38 is required for the mitochondrial respiration of male mice. When p38 is below a normal level, females may maintain mitochondrial respiration through COX up-regulation.
Collapse
Affiliation(s)
- Xiaohua Ju
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| | | | | | | |
Collapse
|
16
|
Xu M, Sulkowski ZL, Parekh P, Khan A, Chen T, Midha S, Iwasaki T, Shimokawa N, Koibuchi N, Zavacki AM, Sajdel-Sulkowska EM. Effects of Perinatal Lipopolysaccharide (LPS) Exposure on the Developing Rat Brain; Modeling the Effect of Maternal Infection on the Developing Human CNS. THE CEREBELLUM 2013; 12:572-86. [DOI: 10.1007/s12311-013-0465-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
17
|
Jung M, Metzger D. Purkinje-neuron-specific down-regulation of p38 protects motoric function from the repeated use of benzodiazepine. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/abb.2013.46a009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
18
|
Liu DZ, Ander BP. Cell cycle inhibition without disruption of neurogenesis is a strategy for treatment of aberrant cell cycle diseases: an update. ScientificWorldJournal 2012; 2012:491737. [PMID: 22547985 PMCID: PMC3323905 DOI: 10.1100/2012/491737] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 11/17/2011] [Indexed: 12/12/2022] Open
Abstract
Since publishing our earlier report describing a strategy for the treatment of central nervous system (CNS) diseases by inhibiting the cell cycle and without disrupting neurogenesis (Liu et al. 2010), we now update and extend this strategy to applications in the treatment of cancers as well. Here, we put forth the concept of "aberrant cell cycle diseases" to include both cancer and CNS diseases, the two unrelated disease types on the surface, by focusing on a common mechanism in each aberrant cell cycle reentry. In this paper, we also summarize the pharmacological approaches that interfere with classical cell cycle molecules and mitogenic pathways to block the cell cycle of tumor cells (in treatment of cancer) as well as to block the cell cycle of neurons (in treatment of CNS diseases). Since cell cycle inhibition can also block proliferation of neural progenitor cells (NPCs) and thus impair brain neurogenesis leading to cognitive deficits, we propose that future strategies aimed at cell cycle inhibition in treatment of aberrant cell cycle diseases (i.e., cancers or CNS diseases) should be designed with consideration of the important side effects on normal neurogenesis and cognition.
Collapse
Affiliation(s)
- Da-Zhi Liu
- Department of Neurology and the MIND Institute, University of California at Davis, Sacramento, CA 95817, USA.
| | | |
Collapse
|
19
|
Excitatory and Mitogenic Signaling in Cell Death, Blood-brain Barrier Breakdown, and BBB Repair after Intracerebral Hemorrhage. Transl Stroke Res 2012; 3:62-9. [PMID: 24323862 DOI: 10.1007/s12975-012-0147-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Revised: 02/18/2012] [Accepted: 02/22/2012] [Indexed: 02/06/2023]
Abstract
Intracerebral hemorrhage (ICH) results in the release of a large number of endogenous molecules, including glutamate, Ca(2+), ROS, thrombin, heme, iron, TNF-α, and others. These molecules participate in excitatory and mitogenic signaling transduction in which N-methyl-D-aspartate (NMDA) receptors and Src family kinases (SFKs) are implicated. Mitogenic signaling initiates the cell cycle for normal cell division of microglia and neural progenitor cells, whereas aberrant mitogenic signaling causes toxicity, killing neurons, astrocytes, and brain microvascular endothelial cells in neurological diseases including ICH. In this review, we summarize (1) how SFKs modulate NMDA receptors to kill neurons following ICH and (2) how SFKs modulate mitogenic signaling transduction to kill neurons and play a role in disrupting the blood-brain barrier (BBB) immediately following ICH and in repairing the BBB during the recovery phases weeks following ICH.
Collapse
|
20
|
Blumer JB, Oner SS, Lanier SM. Group II activators of G-protein signalling and proteins containing a G-protein regulatory motif. Acta Physiol (Oxf) 2012; 204:202-18. [PMID: 21615707 DOI: 10.1111/j.1748-1716.2011.02327.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Beyond the core triad of receptor, Gαβγ and effector, there are multiple accessory proteins that provide alternative modes of signal input and regulatory adaptability to G-protein signalling systems. Such accessory proteins may segregate a signalling complex to microdomains of the cell, regulate the basal activity, efficiency and specificity of signal propagation and/or serve as alternative binding partners for Gα or Gβγ independent of the classical heterotrimeric Gαβγ complex. The latter concept led to the postulate that Gα and Gβγ regulate intracellular events distinct from their role as transducers for cell surface seven-transmembrane span receptors. One general class of such accessory proteins is defined by AGS proteins or activators of G-protein signalling that refer to mammalian cDNAs identified in a specific yeast-based functional screen. The discovery of AGS proteins and related entities revealed a number of unexpected mechanisms for regulation of G-protein signalling systems and expanded functional roles for this important signalling system.
Collapse
Affiliation(s)
- J B Blumer
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, 29425, USA
| | | | | |
Collapse
|
21
|
Zhang L, Sun YN, Li YM, Lin LX, Ye Y, Yan YQ, Chen ZP. Effect of different iodine nutrition on cerebellum Pcp-2 in rat offspring during lactation. Biol Trace Elem Res 2011; 143:1629-39. [PMID: 21344292 DOI: 10.1007/s12011-011-8991-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 02/01/2011] [Indexed: 12/29/2022]
Abstract
The thyroid functions of breastfed infants, as well as (indirectly) the development of their central nervous system, are dependent on the iodine status of the lactating mother. Purkinje cell protein-2 is a cell-specific marker of the cerebellum Purkinje cell and is a suitable indicator for observing the postnatal development of the cerebellum after birth. We measured the Purkinje cell protein-2 mRNA and protein levels in the rat cerebellum in the critical postnatal (14 days after birth) and maturation periods (28 days after birth) to determine the effect of different nutritional iodine levels on cerebellum growth in the offspring during lactation. We found that severe iodine deficiency resulted in thyroid dysfunction in lactating rats and their offspring on both 14 and 28 days, showing maternal total T(4) 16.7 ± 12.0 vs 36.4 ± 15.0, P < 0.05 (14 days) and 22.6 ± 18.7 vs 53.4 ± 9.4, P < 0.01 (28 days), and neonatal total T(4) 10.6 ± 2.3 vs 16.4 ± 4.7, P < 0.01(14 days) and 12.8 ± 2.9 vs 16.7 ± 3.4, P < 0.05 (28 days), respectively. The Purkinje cell protein-2 mRNA and its protein levels in offspring rats were significantly reduced that showed Purkinje cell protein-2 mRNA 1.12 ± 0.04 vs 2.25 ± 0.53, P < 0.05 (14 days) and 1.74 ± 0.94 vs 8.69 ± 2.71, P < 0.01 (28 days). However, mild iodine deficiency and excessive iodine maintained almost normal thyroid function in maternal and neonatal rats and normal Purkinje cell protein-2 mRNA and protein levels in offspring's cerebellum. We conclude that severe iodine deficiency could significantly reduce Purkinje cell protein-2 mRNA and its protein levels, indicating that the cerebellum development was retarded, but mild iodine deficiency and excessive iodine could maintain them at an approximately normal level by the mother's and offspring's compensations, especially by the mother's mammary glands.
Collapse
Affiliation(s)
- Lu Zhang
- Institute of Endocrinology, Tianjin Medical University, Key Lab of Hormones and Development, Ministry of Health, 22, Qi-Xiang-Tai Road, 300070, Tianjin, China
| | | | | | | | | | | | | |
Collapse
|
22
|
Zhou JY, Schepmoes AA, Zhang X, Moore RJ, Monroe ME, Lee JH, Camp DG, Smith RD, Qian WJ. Improved LC-MS/MS spectral counting statistics by recovering low-scoring spectra matched to confidently identified peptide sequences. J Proteome Res 2010; 9:5698-704. [PMID: 20812748 DOI: 10.1021/pr100508p] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Spectral counting has become a popular method for LC-MS/MS based proteome quantification; however, this methodology is often not reliable when proteins are identified by a small number of spectra. Here, we present a simple strategy to improve spectral counting based quantification for low-abundance proteins by recovering low-quality or low-scoring spectra for confidently identified peptides. In this approach, stringent data filtering criteria were initially applied to achieve confident peptide identifications with low false discovery rate (e.g., < 1% at peptide level) after LC-MS/MS analysis and database search by SEQUEST. Then, all low-scoring MS/MS spectra that matched to this set of confidently identified peptides were recovered, leading to more than 20% increase of total identified spectra. The validity of these recovered spectra was assessed by the parent ion mass measurement error distribution, retention time distribution, and by comparing the individual low score and high score spectra that correspond to the same peptides. The results support that the recovered low-scoring spectra have similar confidence levels in peptide identifications as the spectra passing the initial stringent filter. The application of this strategy of recovering low-scoring spectra significantly improved the spectral count quantification statistics for low-abundance proteins, as illustrated in the identification of mouse brain region specific proteins.
Collapse
Affiliation(s)
- Jian-Ying Zhou
- Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Ruether K, Feigenspan A, Pirngruber J, Leitges M, Baehr W, Strauss O. PKC{alpha} is essential for the proper activation and termination of rod bipolar cell response. Invest Ophthalmol Vis Sci 2010; 51:6051-8. [PMID: 20554612 DOI: 10.1167/iovs.09-4704] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Protein kinase (PKC)-α is abundant in retinal bipolar cells. This study was performed to explore its role in visual processing. METHODS PKCα-knockout (Prkca(-/-)) mice and control animals were examined by using electroretinography (ERG), light microscopy, and immunocytochemistry. RESULTS The Prkca(-/-) mice showed no signs of retinal degeneration up to 12 months of age, but ERG measurements indicated a decelerated increase in the ascending limb of the scotopic (rod-sensitive) b-wave as well as a delayed return to baseline. These results suggest that PKCα is an important modulator that affects bipolar cell signal transduction and termination. Confocal microscopy of retinal sections showed that PKCα co-localized with calbindin, which indicates a PKCα localization in close proximity to the horizontal cell terminals. In addition, the implicit time of the ERG c-wave originating from the retinal pigment epithelium (RPE) and the recovery of photoreceptors from bleaching conditions were substantially faster in the knockout mice than in the wild-type control animals. CONCLUSIONS These results suggest that PKCα is a modulator of rod-bipolar cell function by accelerating glutamate-driven signal transduction and termination. This modulation is of importance in the switch between scotopic and photopic vision. Furthermore, PKCα seems to play a role in RPE function.
Collapse
Affiliation(s)
- Klaus Ruether
- Charité- Eye-Hospital, Campus Virchow-Klinikum, Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
24
|
Zhang X, Zhou JY, Chin MH, Schepmoes AA, Petyuk VA, Weitz KK, Petritis BO, Monroe ME, Camp DG, Wood SA, Melega WP, Bigelow DJ, Smith DJ, Qian WJ, Smith RD. Region-specific protein abundance changes in the brain of MPTP-induced Parkinson's disease mouse model. J Proteome Res 2010; 9:1496-509. [PMID: 20155936 DOI: 10.1021/pr901024z] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Parkinson's disease (PD) is characterized by dopaminergic neurodegeneration in the nigrostriatal region of the brain; however, the neurodegeneration extends well beyond dopaminergic neurons. To gain a better understanding of the molecular changes relevant to PD, we applied two-dimensional LC-MS/MS to comparatively analyze the proteome changes in four brain regions (striatum, cerebellum, cortex, and the rest of brain) using a MPTP-induced PD mouse model with the objective to identify potential nigrostriatal-specific and other region-specific protein abundance changes. The combined analyses resulted in the identification of 4,895 nonredundant proteins with at least two unique peptides per protein. The relative abundance changes in each analyzed brain region were estimated based on the spectral count information. A total of 518 proteins were observed with substantial MPTP-induced abundance changes across different brain regions. A total of 270 of these proteins were observed with specific changes occurring either only in the striatum and/or in the rest of the brain region that contains substantia nigra, suggesting that these proteins are associated with the underlying nigrostriatal pathways. Many of the proteins that exhibit changes were associated with dopamine signaling, mitochondrial dysfunction, the ubiquitin system, calcium signaling, the oxidative stress response, and apoptosis. A set of proteins with either consistent change across all brain regions or with changes specific to the cortex and cerebellum regions were also detected. Ubiquitin specific protease (USP9X), a deubiquination enzyme involved in the protection of proteins from degradation and promotion of the TGF-beta pathway, exhibited altered abundance in all brain regions. Western blot validation showed similar spatial changes, suggesting that USP9X is potentially associated with neurodegeneration. Together, this study for the first time presents an overall picture of proteome changes underlying both nigrostriatal pathways and other brain regions potentially involved in MPTP-induced neurodegeneration. The observed molecular changes provide a valuable reference resource for future hypothesis-driven functional studies of PD.
Collapse
Affiliation(s)
- Xu Zhang
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, P.O. Box 999, Richland, Washington 99352, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sulaiman P, Fina M, Feddersen R, Vardi N. Ret-PCP2 colocalizes with protein kinase C in a subset of primate ON cone bipolar cells. J Comp Neurol 2010; 518:1098-112. [PMID: 20127818 DOI: 10.1002/cne.22266] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Purkinje cell protein 2 (PCP2), a member of the family of guanine dissociation inhibitors and a strong interactor with the G-protein subunit G alpha(o), localizes to retinal ON bipolar cells. The retina-specific splice variant of PCP2, Ret-PCP2, accelerates the light response of rod bipolar cells by modulating the mGluR6 transduction cascade. All ON cone bipolar cells express mGluR6 and G alpha(o), but only a subset expresses Ret-PCP2. Here we test the hypothesis that Ret-PCP2 contributes to shaping the various temporal bandwidths of ON cone bipolar cells in monkey retina. We found that the retinal splice variants in monkey and mouse are similar and longer than the cerebellar variants. Ret-PCP2 is strongly expressed by diffuse cone bipolar type 4 cells (DB4; marked with anti-PKCalpha) and weakly expressed by midget bipolar dendrites (labeled by antibodies against G alpha(o), G gamma 13, or mGluR6). Ret-PCP2 is absent from diffuse cone bipolar type 6 (DB6; marked with anti-CD15) and blue cone bipolar cells (marked with anti-CCK precursor). Thus, cone bipolar cells that terminate in stratum 3 of the inner plexiform layer (DB4) express more Ret-PCP2 than those that terminate in strata 3 + 4 (midget bipolar cells), and these in turn express more than those that terminate in stratum 5 (DB6 and blue cone bipolar cells). This expression pattern approximates the arborization of ganglion cells (GC) with different temporal bandwidths: parasol GCs stratifying near stratum 3 are faster than midget GCs stratifying in strata 3 + 4, and these are probably faster than the sluggish GCs that arborize in stratum 5.
Collapse
Affiliation(s)
- Pyroja Sulaiman
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
26
|
Liu DZ, Ander BP, Sharp FR. Cell cycle inhibition without disruption of neurogenesis is a strategy for treatment of central nervous system diseases. Neurobiol Dis 2009; 37:549-57. [PMID: 19944161 DOI: 10.1016/j.nbd.2009.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 11/10/2009] [Accepted: 11/18/2009] [Indexed: 12/12/2022] Open
Abstract
Classically, the cell cycle is regarded as the process leading to cellular proliferation. However, increasing evidence over the last decade supports the notion that neuronal cell cycle re-entry results in post-mitotic death. A mature neuron that re-enters the cell cycle can neither advance to a new G0 quiescent state nor revert to its earlier G0 state. This presents a critical dilemma to the neuron from which death may be an unavoidable but necessary outcome for adult neurons attempting to complete the cell cycle. In contrast, tumor cells that undergo aberrant cell cycle re-entry divide and can survive. Thus, cell cycle inhibition strategies are of interest in cancer treatment but may also represent an important means of protecting neurons. In this review, we put forth the concept of the "expanded cell cycle" and summarize the cell cycle proteins, signal transduction events and mitogenic molecules that can drive a neuron into the cell cycle in various CNS diseases. We also discuss the pharmacological approaches that interfere with the mitogenic pathways and prevent mature neurons from attempting cell cycle re-entry, protecting them from cell death. Lastly, future attempts at blocking the cell cycle to rescue mature neurons from injury should be designed so as to not block normal neurogenesis.
Collapse
Affiliation(s)
- Da-Zhi Liu
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, CA 95817, USA.
| | | | | |
Collapse
|
27
|
Retinal ON bipolar cells express a new PCP2 splice variant that accelerates the light response. J Neurosci 2008; 28:8873-84. [PMID: 18768681 DOI: 10.1523/jneurosci.0812-08.2008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
PCP2, a member of the GoLoco domain-containing family, is present exclusively in cerebellar Purkinje cells and retinal ON bipolar cells. Its function in these tissues is unknown. Biochemical and expression system studies suggest that PCP2 is a guanine nucleotide dissociation inhibitor, although a guanine nucleotide exchange factor has also been suggested. Here, we studied the function of PCP2 in ON bipolar cells because their light response depends on Galpha(o1), which is known to interact with PCP2. We identified a new splice variant of PCP2 (Ret-PCP2) and localized it to rod bipolar and ON cone bipolar cells. Electroretinogram recordings from PCP2-null mice showed a normal a-wave but a slower falling phase of the b-wave (generated by the activity of ON bipolar cells) relative to the wild type. Whole-cell recordings from rod bipolar cells showed, both under Ames medium and after blocking GABA(A/C) and glycine receptors, that PCP2-null rod bipolar cells were more depolarized than wild-type cells with greater inward current when clamped to -60 mV. Also under both conditions, the rise time of the response to intense light was slower by 28% (Ames) and 44% (inhibitory blockers) in the null cells. Under Ames medium, we also observed >30% longer decay time in the PCP2-null rod bipolar cells. We conclude that PCP2 facilitates cation channels closure in the dark, shortens the rise time of the light response directly, and accelerates the decay time indirectly via the inhibitory network. These data can most easily be explained if PCP2 serves as a guanine nucleotide exchange factor.
Collapse
|
28
|
Yung LY, Tso PH, Wu EH, Yu JC, Ip NY, Wong YH. Nerve growth factor-induced stimulation of p38 mitogen-activated protein kinase in PC12 cells is partially mediated via Gi/o proteins. Cell Signal 2008; 20:1538-44. [DOI: 10.1016/j.cellsig.2008.04.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 04/10/2008] [Accepted: 04/11/2008] [Indexed: 12/21/2022]
|
29
|
Gene expression profiling in rat cerebellum following in utero and lactational exposure to mixtures of methylmercury, polychlorinated biphenyls and organochlorine pesticides. Toxicol Lett 2008; 176:93-103. [DOI: 10.1016/j.toxlet.2007.08.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Revised: 08/30/2007] [Accepted: 08/30/2007] [Indexed: 11/20/2022]
|
30
|
Compe E, Malerba M, Soler L, Marescaux J, Borrelli E, Egly JM. Neurological defects in trichothiodystrophy reveal a coactivator function of TFIIH. Nat Neurosci 2007; 10:1414-22. [PMID: 17952069 DOI: 10.1038/nn1990] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Accepted: 09/06/2007] [Indexed: 11/09/2022]
Abstract
Mutations in the XPD subunit of the DNA repair/transcription factor TFIIH yield the rare genetic disorder trichothiodystrophy (TTD). Although this syndrome was initially associated with a DNA repair defect, individuals with TTD develop neurological features, such as microcephaly and hypomyelination that could be connected to transcriptional defects. Here we show that an XPD mutation in TTD mice results in a spatial and selective deregulation of thyroid hormone target genes in the brain. Molecular analyses performed on the mice brain tissue demonstrate that TFIIH is required for the stabilization of thyroid hormone receptors (TR) to their DNA-responsive elements. The limiting amounts of TFIIH found in individuals with TTD thus contribute to the deregulation of TR-responsive genes. The discovery of an unexpected stabilizing function for TFIIH deepens our understanding of the pathogenesis and neurological manifestations observed in TTD individuals.
Collapse
Affiliation(s)
- Emmanuel Compe
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 1 rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, C.U. Strasbourg, France
| | | | | | | | | | | |
Collapse
|
31
|
Kitatsuji C, Kurogochi M, Nishimura SI, Ishimori K, Wakasugi K. Molecular Basis of Guanine Nucleotide Dissociation Inhibitor Activity of Human Neuroglobin by Chemical Cross-linking and Mass Spectrometry. J Mol Biol 2007; 368:150-60. [PMID: 17337004 DOI: 10.1016/j.jmb.2007.02.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Revised: 02/01/2007] [Accepted: 02/01/2007] [Indexed: 11/24/2022]
Abstract
Oxidized human neuroglobin (Ngb), a heme protein expressed in the brain, has been proposed to act as a guanine nucleotide dissociation inhibitor (GDI) for the GDP-bound form of the heterotrimeric G protein alpha-subunit (Galpha(i)). Here, to elucidate the molecular mechanism underlying the GDI activity of Ngb, we used an glutathione-S-transferase pull-down assay to confirm that Ngb competes with G-protein betagamma-subunits (Gbetagamma) for binding to Galpha(i), and identified the Galpha(i)-binding site in Ngb by chemical cross-linking with 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride and sulfo-N-hydroxysuccinimide, coupled with mass spectrometry (MS). Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS analysis for tryptic peptides derived from the cross-linked Ngb-Galpha(i) complex revealed several binding regions in Ngb. Furthermore, MALDI-TOF/TOF MS analysis of the cross-linked Ngb and Galpha(i) peptides, together with the MS/MS scoring method, predicted cross-linking between Glu60 (Ngb) and Ser206 (Galpha(i)), and between Glu53 (Ngb) and Ser44 (Galpha(i)). Because Ser206 of Galpha(i) is located in the region that contacts Gbetagamma, binding of Ngb could facilitate the release of Gbetagamma from Galpha(i). Binding of Ngb to Galpha(i) would also inhibit the exchange of GDP for GTP, because Ser44 (Galpha(i)) is adjacent to the GDP-binding site and Glu53 (Ngb), which is cross-linked to Ser44 (Galpha(i)), could be located close to GDP. Thus, we have identified, for the first time, the sites of interaction between Ngb and Galpha(i), enabling us to discuss the functional significance of this binding on the GDI activity of Ngb.
Collapse
Affiliation(s)
- Chihiro Kitatsuji
- Department of Molecular Engineering, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | | | | | | | | |
Collapse
|
32
|
Blumer JB, Smrcka AV, Lanier S. Mechanistic pathways and biological roles for receptor-independent activators of G-protein signaling. Pharmacol Ther 2006; 113:488-506. [PMID: 17240454 PMCID: PMC1978177 DOI: 10.1016/j.pharmthera.2006.11.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2006] [Accepted: 11/10/2006] [Indexed: 01/14/2023]
Abstract
Signal processing via heterotrimeric G-proteins in response to cell surface receptors is a central and much investigated aspect of how cells integrate cellular stimuli to produce coordinated biological responses. The system is a target of numerous therapeutic agents and plays an important role in adaptive processes of organs; aberrant processing of signals through these transducing systems is a component of various disease states. In addition to G-protein coupled receptor (GPCR)-mediated activation of G-protein signaling, nature has evolved creative ways to manipulate and utilize the Galphabetagamma heterotrimer or Galpha and Gbetagamma subunits independent of the cell surface receptor stimuli. In such situations, the G-protein subunits (Galpha and Gbetagamma) may actually be complexed with alternative binding partners independent of the typical heterotrimeric Galphabetagamma. Such regulatory accessory proteins include the family of regulator of G-protein signaling (RGS) proteins that accelerate the GTPase activity of Galpha and various entities that influence nucleotide binding properties and/or subunit interaction. The latter group of proteins includes receptor-independent activators of G-protein signaling (AGS) proteins that play surprising roles in signal processing. This review provides an overview of our current knowledge regarding AGS proteins. AGS proteins are indicative of a growing number of accessory proteins that influence signal propagation, facilitate cross talk between various types of signaling pathways, and provide a platform for diverse functions of both the heterotrimeric Galphabetagamma and the individual Galpha and Gbetagamma subunits.
Collapse
Affiliation(s)
| | - Alan V. Smrcka
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Box 711, Rochester, NY 14642-8711
| | - S.M. Lanier
- ** Corresponding Author, Stephen M. Lanier, Ph.D., Department of Pharmacology, Medical University of South Carolina, Colcock Hall, 2nd Floor, PO Box 250002, 179 Ashley Avenue, Charleston, SC 29425, 843-792-0442, E-mail:
| |
Collapse
|
33
|
Chang C, Dong R, Miyazaki A, Sakashita N, Zhang Y, Liu J, Guo M, Li BL, Chang TY. Human acyl-CoA:cholesterol acyltransferase (ACAT) and its potential as a target for pharmaceutical intervention against atherosclerosis. Acta Biochim Biophys Sin (Shanghai) 2006; 38:151-6. [PMID: 16518538 DOI: 10.1111/j.1745-7270.2006.00154.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Acyl-CoA:cholesterol acyltransferase (ACAT) catalyzes the formation of cholesteryl esters from cholesterol and long-chain fatty-acyl-coenzyme A. At the single-cell level, ACAT serves as a regulator of intracellular cholesterol homeostasis. In addition, ACAT supplies cholesteryl esters for lipoprotein assembly in the liver and small intestine. Under pathological conditions, the accumulation of cholesteryl esters produced by ACAT in macrophages contributes to foam cell formation, a hallmark of the early stage of atherosclerosis. Several reviews addressing various aspects of ACAT and ACAT inhibitors are available. This review briefly outlines the current knowledge on the biochemical properties of human ACATs, and then focuses on discussing the merit of ACAT as a drug target for pharmaceutical interventions against atherosclerosis.
Collapse
Affiliation(s)
- Catherine Chang
- Department of Biochemistry, Dartmouth Medical School, Hanover, New Hampshire 03755, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Li BL, Chang TY, Chen J, Chang CCY, Zhao XN. Human ACAT1 gene expression and its involvement in the development of atherosclerosis. Future Cardiol 2006; 2:93-9. [DOI: 10.2217/14796678.2.1.93] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Atherosclerosis is caused by a series of pathologic changes at the cellular level, with formation of macrophage-derived foam cells occurring at an early stage. Most of the cholesteryl esters in macrophage foam cells are produced by the enzyme acyl-coenzyme A:cholesterol acyltransferase (ACAT). Two ACAT genes, Acat1 and Acat2, exist in mammals. In the monocyte–macrophages, ACAT1 is the major isoenzyme and is a drug target for atherosclerosis treatment. Various proatherogenic stimuli, including interferon-γ and dexamethasone, cause upregulation of human Acat1 expression in macrophages. Thus, it should be possible to find antagonist(s) to downregulate human Acat1 expression. A greater understanding of human Acat1 expression may provide scientists with opportunities for novel therapeutic approaches to combat atherosclerosis.
Collapse
Affiliation(s)
- Bo-Liang Li
- State Key Laboratory of Molecular Biology, 320 Yue-Yang Road, Shanghai 200031, China
| | - Ta-Yuan Chang
- Department of Biochemistry, Dartmouth Medical School, Hanover, NH 03755 USA
| | - Jia Chen
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Catherine CY Chang
- Department of Biochemistry, Dartmouth Medical School, Hanover, NH 03755, USA
| | - Xiao-Nan Zhao
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|