201
|
Abstract
p21-activated kinases (PAKs) act downstream of Rho-family GTPase and are linked to steps in both cancer initiation and progression. There are six mammalian PAK isoforms that are divided into two groups, and for different reasons both groups are attractive targets for cancer therapy. We describe the background and recent development of a PAK inhibitor, PF-3758309, which exhibits relatively good selectivity and high potency for PAKs. Experiments using PF-3758309 confirm that inhibiting PAK is a beneficial strategy to combat some tumors, and this activity is likely related to modulation of both cell proliferation and survival. The genetic loss of NF2 (neurofibromatosis type 2) leading to increased cell proliferation through a Ras-Rac-PAK pathway may represent a good test system to analyze this new PAK inhibitor.
Collapse
Affiliation(s)
- Zhuo-shen Zhao
- Small G-Protein Signalling and Kinases (sGSK) Group at Institute of Molecular and Cell Biology (IMCB), Neuroscience Research PartnershipProteos BuildingSingapore 138673
| | - Ed Manser
- Small G-Protein Signalling and Kinases (sGSK) Group at Institute of Molecular and Cell Biology (IMCB), Neuroscience Research PartnershipProteos BuildingSingapore 138673
- Institute of Medical Biology (IMB), A*STAR#06-34 Immunos BuildingSingapore 138648
| |
Collapse
|
202
|
Perreau VM, Orchard S, Adlard PA, Bellingham SA, Cappai R, Ciccotosto GD, Cowie TF, Crouch PJ, Duce JA, Evin G, Faux NG, Hill AF, Hung YH, James SA, Li QX, Mok SS, Tew DJ, White AR, Bush AI, Hermjakob H, Masters CL. A domain level interaction network of amyloid precursor protein and Abeta of Alzheimer's disease. Proteomics 2010; 10:2377-95. [PMID: 20391539 DOI: 10.1002/pmic.200900773] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The primary constituent of the amyloid plaque, beta-amyloid (Abeta), is thought to be the causal "toxic moiety" of Alzheimer's disease. However, despite much work focused on both Abeta and its parent protein, amyloid precursor protein (APP), the functional roles of APP and its cleavage products remain to be fully elucidated. Protein-protein interaction networks can provide insight into protein function, however, high-throughput data often report false positives and are in frequent disagreement with low-throughput experiments. Moreover, the complexity of the CNS is likely to be under represented in such databases. Therefore, we curated the published work characterizing both APP and Abeta to create a protein interaction network of APP and its proteolytic cleavage products, with annotation, where possible, to the level of APP binding domain and isoform. This is the first time that an interactome has been refined to domain level, essential for the interpretation of APP due to the presence of multiple isoforms and processed fragments. Gene ontology and network analysis were used to identify potentially novel functional relationships among interacting proteins.
Collapse
Affiliation(s)
- Victoria M Perreau
- Neuroproteomics and Neurogenomics Platform, National Neurosciences Facility, The University of Melbourne, Parkville, VIC, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Mergui X, Puiffe ML, Valteau-Couanet D, Lipinski M, Bénard J, Amor-Guéret M. p21Waf1 expression is regulated by nuclear intermediate filament vimentin in neuroblastoma. BMC Cancer 2010; 10:473. [PMID: 20813048 PMCID: PMC2939553 DOI: 10.1186/1471-2407-10-473] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 09/02/2010] [Indexed: 01/26/2023] Open
Abstract
Background Human neuroblastoma (NB) cell lines may present with either one of the so-called S-and N-subtypes. We have previously reported a strong correlation between protein expression levels of vimentin, an S-subtype marker, and the p21Waf1 cyclin-dependent kinase inhibitor. We here investigated whether this correlation extend to the mRNA level in NB cell lines as well as in patients' tumors. We also further explored the relationship between expression of vimentin and p21, by asking whether vimentin could regulate p21 expression. Methods Vimentin and p21 mRNA levels in NB cell lines as well as in patients' tumors (n = 77) were quantified using Q-PCR. Q-PCR data obtained from tumors of high risk NB patients (n = 40) were analyzed in relation with the overall survival using the Log-rank Kaplan-Meier estimation. siRNA-mediated depletion or overexpression of vimentin in highly or low expressing vimentin cell lines, respectively, followed by protein expression and promoter activation assays were used to assess the role of vimentin in modulating p21 expression. Results We extend the significant correlation between vimentin and p21 expression to the mRNA level in NB cell lines as well as in patients' tumors. Overall survival analysis from Q-PCR data obtained from tumors of high risk patients suggests that lower levels of p21 expression could be associated with a poorer outcome. Our data additionally indicate that the correlation observed between p21 and vimentin expression levels results from p21 transcriptional activity being regulated by vimentin. Indeed, downregulating vimentin resulted in a significant decrease in p21 mRNA and protein expression as well as in p21 promoter activity. Conversely, overexpressing vimentin triggered an increase in p21 promoter activity in cells with a nuclear expression of vimentin. Conclusion Our results suggest that p21 mRNA tumor expression level could represent a refined prognostic factor for high risk NB patients. Our data also show that vimentin regulates p21 transcription; this is the first demonstration of a gene regulating function for this type III-intermediate filament.
Collapse
Affiliation(s)
- Xénia Mergui
- Université Paris Sud-11, CNRS, UMR 8126, Institut de Cancérologie Gustave Roussy, 114 rue Edouard Vaillant, Villejuif F-94805, France.
| | | | | | | | | | | |
Collapse
|
204
|
Cause-effect relationships in medicine: a protein network perspective. Trends Pharmacol Sci 2010; 31:547-55. [PMID: 20810173 DOI: 10.1016/j.tips.2010.07.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Revised: 07/21/2010] [Accepted: 07/26/2010] [Indexed: 11/22/2022]
Abstract
Current target-based drug discovery platforms are not able to predict drug efficacy and the full spectrum of drug effects in organisms. Hence, many experimental drugs do not survive the lengthy and costly process of drug development. Understanding how drugs affect cellular network structures and how the resulting signals are translated into drug effects is extremely important for the discovery of new medicines. This requires a greater understanding of cause-effect relationships at the organism, organ, tissue, cellular, and molecular level. There is a growing recognition that this information must be integrated into discovery paradigms, but a 'road map' for obtaining and integrating information about heterogeneous networks into drug-discovery platforms currently does not exist. This review explores recent network-centered approaches developed to investigate the genesis of medicine and disease effects, specifically highlighting protein-protein interaction network models and their use in cause-effect analyses in medicine.
Collapse
|
205
|
van Ham TJ, Holmberg MA, van der Goot AT, Teuling E, Garcia-Arencibia M, Kim HE, Du D, Thijssen KL, Wiersma M, Burggraaff R, van Bergeijk P, van Rheenen J, Jerre van Veluw G, Hofstra RMW, Rubinsztein DC, Nollen EAA. Identification of MOAG-4/SERF as a regulator of age-related proteotoxicity. Cell 2010; 142:601-12. [PMID: 20723760 DOI: 10.1016/j.cell.2010.07.020] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 03/31/2010] [Accepted: 06/17/2010] [Indexed: 01/03/2023]
Abstract
Fibrillar protein aggregates are the major pathological hallmark of several incurable, age-related, neurodegenerative disorders. These aggregates typically contain aggregation-prone pathogenic proteins, such as amyloid-beta in Alzheimer's disease and alpha-synuclein in Parkinson's disease. It is, however, poorly understood how these aggregates are formed during cellular aging. Here we identify an evolutionarily highly conserved modifier of aggregation, MOAG-4, as a positive regulator of aggregate formation in C. elegans models for polyglutamine diseases. Inactivation of MOAG-4 suppresses the formation of compact polyglutamine aggregation intermediates that are required for aggregate formation. The role of MOAG-4 in driving aggregation extends to amyloid-beta and alpha-synuclein and is evolutionarily conserved in its human orthologs SERF1A and SERF2. MOAG-4/SERF appears to act independently from HSF-1-induced molecular chaperones, proteasomal degradation, and autophagy. Our results suggest that MOAG-4/SERF regulates age-related proteotoxicity through a previously unexplored pathway, which will open up new avenues for research on age-related, neurodegenerative diseases.
Collapse
Affiliation(s)
- Tjakko J van Ham
- Department of Genetics, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Godin JD, Colombo K, Molina-Calavita M, Keryer G, Zala D, Charrin BC, Dietrich P, Volvert ML, Guillemot F, Dragatsis I, Bellaiche Y, Saudou F, Nguyen L, Humbert S. Huntingtin is required for mitotic spindle orientation and mammalian neurogenesis. Neuron 2010; 67:392-406. [PMID: 20696378 DOI: 10.1016/j.neuron.2010.06.027] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2010] [Indexed: 01/06/2023]
Abstract
Huntingtin is the protein mutated in Huntington's disease, a devastating neurodegenerative disorder. We demonstrate here that huntingtin is essential to control mitosis. Huntingtin is localized at spindle poles during mitosis. RNAi-mediated silencing of huntingtin in cells disrupts spindle orientation by mislocalizing the p150(Glued) subunit of dynactin, dynein, and the large nuclear mitotic apparatus NuMA protein. This leads to increased apoptosis following mitosis of adherent cells in vitro. In vivo inactivation of huntingtin by RNAi or by ablation of the Hdh gene affects spindle orientation and cell fate of cortical progenitors of the ventricular zone in mouse embryos. This function is conserved in Drosophila, the specific disruption of Drosophila huntingtin in neuroblast precursors leading to spindle misorientation. Moreover, Drosophila huntingtin restores spindle misorientation in mammalian cells. These findings reveal an unexpected role for huntingtin in dividing cells, with potential important implications in health and disease.
Collapse
|
207
|
Dowie MJ, Scotter EL, Molinari E, Glass M. The therapeutic potential of G-protein coupled receptors in Huntington's disease. Pharmacol Ther 2010; 128:305-23. [PMID: 20708032 DOI: 10.1016/j.pharmthera.2010.07.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 07/14/2010] [Indexed: 01/29/2023]
Abstract
Huntington's disease is a late-onset autosomal dominant inherited neurodegenerative disease characterised by increased symptom severity over time and ultimately premature death. An expanded CAG repeat sequence in the huntingtin gene leads to a polyglutamine expansion in the expressed protein, resulting in complex dysfunctions including cellular excitotoxicity and transcriptional dysregulation. Symptoms include cognitive deficits, psychiatric changes and a movement disorder often referred to as Huntington's chorea, which involves characteristic involuntary dance-like writhing movements. Neuropathologically Huntington's disease is characterised by neuronal dysfunction and death in the striatum and cortex with an overall decrease in cerebral volume (Ho et al., 2001). Neuronal dysfunction begins prior to symptom presentation, and cells of particular vulnerability include the striatal medium spiny neurons. Huntington's is a devastating disease for patients and their families and there is currently no cure, or even an effective therapy for disease symptoms. G-protein coupled receptors are the most abundant receptor type in the central nervous system and are linked to complex downstream pathways, manipulation of which may have therapeutic application in many neurological diseases. This review will highlight the potential of G-protein coupled receptor drug targets as emerging therapies for Huntington's disease.
Collapse
Affiliation(s)
- Megan J Dowie
- Centre for Brain Research, University of Auckland, Private Bag 92019 Auckland, New Zealand
| | | | | | | |
Collapse
|
208
|
Wang W, Han G, Ye M, Shi H, Zou H, Huo K. Mapping of phosphorylation sites in human MSK1 activated by a novel interaction with MRK-beta. Electrophoresis 2010; 31:1283-93. [PMID: 20408143 DOI: 10.1002/elps.200900637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The desire to map reliable phosphorylation signaling network has motivated the development of high-performance techniques. Targeted biochemical studies and updated methods employing MS techniques are most used in mapping the phosphorylation sites and verifying novel interactions of kinases. Previously, we have established a novel method to efficiently facilitate more comprehensive, accurate phosphorylation site mapping of individual phosphoproteins by using combination of multiple stage MS analysis with target-decoy database search against the much smaller targeted database. In this study, by applying this method, we have identified the phosphorylation sites in human MSK1 mitogen- and stress-activated protein kinase 1, which has been proved to be a multi-phosphorylated kinase that plays key roles in various cell functions, activated by a novel interaction with MRK-beta. The results show that this method can find out not only those previously identified active sites in MSK1, but also some novel phosphorylated sites, which correlates with biochemical evidence that, besides p38 and extracellular signal-regulated kinase, MRK-beta could also activate MSK1 through direct interaction. Hence, we conclude this method is sensitive and reliable as expected and it can be further combined with automated screening and biochemical study in efficiently building up a more comprehensive phosphoprotein network.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | | | | | | | | | | |
Collapse
|
209
|
Sylvester M, Kliche S, Lange S, Geithner S, Klemm C, Schlosser A, Großmann A, Stelzl U, Schraven B, Krause E, Freund C. Adhesion and degranulation promoting adapter protein (ADAP) is a central hub for phosphotyrosine-mediated interactions in T cells. PLoS One 2010; 5:e11708. [PMID: 20661443 PMCID: PMC2908683 DOI: 10.1371/journal.pone.0011708] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 06/08/2010] [Indexed: 01/13/2023] Open
Abstract
TCR stimulation leads to an increase in cellular adhesion among other outcomes. The adhesion and degranulation promoting adapter protein (ADAP) is known to be rapidly phosphorylated after T cell stimulation and relays the TCR signal to adhesion molecules of the integrin family. While three tyrosine phosphorylation sites have been characterized biochemically, the binding capabilities and associated functions of several other potential phosphotyrosine motifs remain unclear. Here, we utilize in vitro phosphorylation and mass spectrometry to map novel phosphotyrosine sites in the C-terminal part of human ADAP (486–783). Individual tyrosines were then mutated to phenylalanine and their relevance for cellular adhesion and migration was tested experimentally. Functionally important tyrosine residues include two sites within the folded hSH3 domains of ADAP and two at the C-terminus. Furthermore, using a peptide pulldown approach in combination with stable isotope labeling in cell culture (SILAC) we identified SLP-76, PLCγ, PIK3R1, Nck, CRK, Gads, and RasGAP as phospho-dependent binding partners of a central YDDV motif of ADAP. The phosphorylation-dependent interaction between ADAP and Nck was confirmed by yeast two-hybrid analysis, immunoprecipitation and binary pulldown experiments, indicating that ADAP directly links integrins to modulators of the cytoskeleton independent of SLP-76.
Collapse
Affiliation(s)
- Marc Sylvester
- Protein Engineering Group, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Fachbereich Biologie, Chemie, Pharmazie, Freie Universität Berlin, Berlin, Germany
| | - Stefanie Kliche
- Institut für Molekulare und Klinische Immunologie, Otto-von-Guericke-Universität, Magdeburg, Germany
| | - Sabine Lange
- Mass Spectrometry Group, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Sabine Geithner
- Protein Engineering Group, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Fachbereich Biologie, Chemie, Pharmazie, Freie Universität Berlin, Berlin, Germany
| | - Clementine Klemm
- Mass Spectrometry Group, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Andreas Schlosser
- Institut für Medizinische Immunologie CCM, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Arndt Großmann
- Otto-Warburg-Laboratorium, Max-Planck-Institut für Molekulare Genetik, Berlin, Germany
| | - Ulrich Stelzl
- Otto-Warburg-Laboratorium, Max-Planck-Institut für Molekulare Genetik, Berlin, Germany
| | - Burkhart Schraven
- Institut für Molekulare und Klinische Immunologie, Otto-von-Guericke-Universität, Magdeburg, Germany
| | - Eberhard Krause
- Mass Spectrometry Group, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Christian Freund
- Protein Engineering Group, Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Fachbereich Biologie, Chemie, Pharmazie, Freie Universität Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
210
|
Zuccato C, Valenza M, Cattaneo E. Molecular mechanisms and potential therapeutical targets in Huntington's disease. Physiol Rev 2010; 90:905-81. [PMID: 20664076 DOI: 10.1152/physrev.00041.2009] [Citation(s) in RCA: 617] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the gene encoding for huntingtin protein. A lot has been learned about this disease since its first description in 1872 and the identification of its causative gene and mutation in 1993. We now know that the disease is characterized by several molecular and cellular abnormalities whose precise timing and relative roles in pathogenesis have yet to be understood. HD is triggered by the mutant protein, and both gain-of-function (of the mutant protein) and loss-of-function (of the normal protein) mechanisms are involved. Here we review the data that describe the emergence of the ancient huntingtin gene and of the polyglutamine trait during the last 800 million years of evolution. We focus on the known functions of wild-type huntingtin that are fundamental for the survival and functioning of the brain neurons that predominantly degenerate in HD. We summarize data indicating how the loss of these beneficial activities reduces the ability of these neurons to survive. We also review the different mechanisms by which the mutation in huntingtin causes toxicity. This may arise both from cell-autonomous processes and dysfunction of neuronal circuitries. We then focus on novel therapeutical targets and pathways and on the attractive option to counteract HD at its primary source, i.e., by blocking the production of the mutant protein. Strategies and technologies used to screen for candidate HD biomarkers and their potential application are presented. Furthermore, we discuss the opportunities offered by intracerebral cell transplantation and the likely need for these multiple routes into therapies to converge at some point as, ideally, one would wish to stop the disease process and, at the same time, possibly replace the damaged neurons.
Collapse
Affiliation(s)
- Chiara Zuccato
- Department of Pharmacological Sciences and Centre for Stem Cell Research, Università degli Studi di Milano, Milan, Italy
| | | | | |
Collapse
|
211
|
Enokido Y, Tamura T, Ito H, Arumughan A, Komuro A, Shiwaku H, Sone M, Foulle R, Sawada H, Ishiguro H, Ono T, Murata M, Kanazawa I, Tomilin N, Tagawa K, Wanker EE, Okazawa H. Mutant huntingtin impairs Ku70-mediated DNA repair. ACTA ACUST UNITED AC 2010; 189:425-43. [PMID: 20439996 PMCID: PMC2867301 DOI: 10.1083/jcb.200905138] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mutant huntingtin prevents interaction of the DNA damage repair complex component Ku70 with damaged DNA, blocking repair of double-strand breaks. DNA repair defends against naturally occurring or disease-associated DNA damage during the long lifespan of neurons and is implicated in polyglutamine disease pathology. In this study, we report that mutant huntingtin (Htt) expression in neurons causes double-strand breaks (DSBs) of genomic DNA, and Htt further promotes DSBs by impairing DNA repair. We identify Ku70, a component of the DNA damage repair complex, as a mediator of the DNA repair dysfunction in mutant Htt–expressing neurons. Mutant Htt interacts with Ku70, impairs DNA-dependent protein kinase function in nonhomologous end joining, and consequently increases DSB accumulation. Expression of exogenous Ku70 rescues abnormal behavior and pathological phenotypes in the R6/2 mouse model of Huntington’s disease (HD). These results collectively suggest that Ku70 is a critical regulator of DNA damage in HD pathology.
Collapse
Affiliation(s)
- Yasushi Enokido
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Neuroinflammation in Huntington's disease. J Neural Transm (Vienna) 2010; 117:1001-8. [PMID: 20535620 DOI: 10.1007/s00702-010-0430-7] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 05/22/2010] [Indexed: 01/17/2023]
Abstract
Huntington's disease (HD) is a monogenic neurodegenerative disease characterized by abnormal motor movements, personality changes and early death. In contrast to other neurodegenerative diseases, very little is known about the role of neuroinflammation in HD. While the current data clearly demonstrate the existence of inflammatory processes in HD pathophysiology, the question of whether neuroinflammation is purely reactive or might actively participate in disease pathogenesis is currently a matter of ongoing research and debate. This review will try to shed some light on the current state of research in this area and provide an outlook on potential future developments.
Collapse
|
213
|
Pardo R, Molina-Calavita M, Poizat G, Keryer G, Humbert S, Saudou F. pARIS-htt: an optimised expression platform to study huntingtin reveals functional domains required for vesicular trafficking. Mol Brain 2010; 3:17. [PMID: 20515468 PMCID: PMC2887845 DOI: 10.1186/1756-6606-3-17] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 06/01/2010] [Indexed: 01/02/2023] Open
Abstract
Background Huntingtin (htt) is a multi-domain protein of 350 kDa that is mutated in Huntington's disease (HD) but whose function is yet to be fully understood. This absence of information is due in part to the difficulty of manipulating large DNA fragments by using conventional molecular cloning techniques. Consequently, few studies have addressed the cellular function(s) of full-length htt and its dysfunction(s) associated with the disease. Results We describe a flexible synthetic vector encoding full-length htt called pARIS-htt (Adaptable, RNAi Insensitive &Synthetic). It includes synthetic cDNA coding for full-length human htt modified so that: 1) it is improved for codon usage, 2) it is insensitive to four different siRNAs allowing gene replacement studies, 3) it contains unique restriction sites (URSs) dispersed throughout the entire sequence without modifying the translated amino acid sequence, 4) it contains multiple cloning sites at the N and C-ter ends and 5) it is Gateway compatible. These modifications facilitate mutagenesis, tagging and cloning into diverse expression plasmids. Htt regulates dynein/dynactin-dependent trafficking of vesicles, such as brain-derived neurotrophic factor (BDNF)-containing vesicles, and of organelles, including reforming and maintenance of the Golgi near the cell centre. We used tests of these trafficking functions to validate various pARIS-htt constructs. We demonstrated, after silencing of endogenous htt, that full-length htt expressed from pARIS-htt rescues Golgi apparatus reformation following reversible microtubule disruption. A mutant form of htt that contains a 100Q expansion and a htt form devoid of either HAP1 or dynein interaction domains are both unable to rescue loss of endogenous htt. These mutants have also an impaired capacity to promote BDNF vesicular trafficking in neuronal cells. Conclusion We report the validation of a synthetic gene encoding full-length htt protein that will facilitate analyses of its structure/function. This may help provide relevant information about the cellular dysfunctions operating during the disease. As proof of principle, we show that either polyQ expansion or deletion of key interacting domains within full-length htt protein impairs its function in transport indicating that HD mutation induces defects on intrinsic properties of the protein and further demonstrating the importance of studying htt in its full-length context.
Collapse
|
214
|
Wang J, Zhou X, Zhu J, Zhou C, Guo Z. Revealing and avoiding bias in semantic similarity scores for protein pairs. BMC Bioinformatics 2010; 11:290. [PMID: 20509916 PMCID: PMC2903568 DOI: 10.1186/1471-2105-11-290] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 05/28/2010] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Semantic similarity scores for protein pairs are widely applied in functional genomic researches for finding functional clusters of proteins, predicting protein functions and protein-protein interactions, and for identifying putative disease genes. However, because some proteins, such as those related to diseases, tend to be studied more intensively, annotations are likely to be biased, which may affect applications based on semantic similarity measures. Thus, it is necessary to evaluate the effects of the bias on semantic similarity scores between proteins and then find a method to avoid them. RESULTS First, we evaluated 14 commonly used semantic similarity scores for protein pairs and demonstrated that they significantly correlated with the numbers of annotation terms for the proteins (also known as the protein annotation length). These results suggested that current applications of the semantic similarity scores between proteins might be unreliable. Then, to reduce this annotation bias effect, we proposed normalizing the semantic similarity scores between proteins using the power transformation of the scores. We provide evidence that this improves performance in some applications. CONCLUSIONS Current semantic similarity measures for protein pairs are highly dependent on protein annotation lengths, which are subject to biological research bias. This affects applications that are based on these semantic similarity scores, especially in clustering studies that rely on score magnitudes. The normalized scores proposed in this paper can reduce the effects of this bias to some extent.
Collapse
Affiliation(s)
- Jing Wang
- Bioinformatics Centre, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Xianxiao Zhou
- Bioinformatics Centre, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Jing Zhu
- Bioinformatics Centre, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Chenggui Zhou
- Bioinformatics Centre, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Zheng Guo
- Bioinformatics Centre, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| |
Collapse
|
215
|
Abu-Helo A, Simonin F. Identification and biological significance of G protein-coupled receptor associated sorting proteins (GASPs). Pharmacol Ther 2010; 126:244-50. [PMID: 20394773 DOI: 10.1016/j.pharmthera.2010.03.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 03/23/2010] [Indexed: 12/25/2022]
Abstract
G protein-coupled receptors (GPCRs) represent one of the most abundant protein families encoded by the human genome. They are involved in the modulation of numerous physiological functions and represent major drug targets. Their activity is tightly controlled by a vast array of interacting partners that modulate their membrane targeting, intracellular trafficking and signalling properties. Among them, several proteins from the same family, G protein-coupled receptor associated sorting proteins (GASP), have been shown to display a broad spectrum of interactions with GPCRs. In addition to their postulated role in the modulation of the post-endocytic sorting of these receptors, recent data indicate that several GASPs may modulate the transcriptional activity of the cell through their interaction with transcription factors. However, no clear molecular function has been assigned yet to this protein family. In this review, we describe the discovery of GASPs, their major features, interacting partners, functions and possible involvement in pathological situations including neurodegenerative diseases and cancer.
Collapse
Affiliation(s)
- Alaa Abu-Helo
- Institut de Recherche de l'ESBS, CNRS - Université de Strasbourg, FRE3211, Bld Sébastien Brant, 67412 Illkirch, France
| | | |
Collapse
|
216
|
Ravache M, Abou-Sleymane G, Trottier Y. [Neurodegenerative polyglutamine expansion diseases: physiopathology and therapeutic strategies]. ACTA ACUST UNITED AC 2010; 58:357-66. [PMID: 20299163 DOI: 10.1016/j.patbio.2009.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 12/29/2009] [Indexed: 10/19/2022]
Abstract
Polyglutamine expansion diseases are adult-onset inherited neurodegenerative disorders that lead to death 10 to 20 years after the first symptoms. Currently, there is no therapy to fight against these diseases. They include Huntington's disease, spinobulbar muscular atrophy, dentatorubral-pallido-luysian atrophy and six types of spino-cerebellar ataxia. The diseases are caused by a unique mutational mechanism: an expansion of the CAG trinucleotide in the corresponding genes coding for an expanded tract of glutamine in the mutated proteins. Polyglutamine expansion confers to the mutant proteins toxic properties that cause neuronal cell death in brain regions specific to each disease. Thanks to cellular and animal models (fly, fish, mouse and rat) of these diseases, we have considerably improved our understanding of the toxic nature of polyglutamine expansion and the physiopathology, and we are now in position to design and test therapeutic strategies to prevent or delay the disease process.
Collapse
Affiliation(s)
- M Ravache
- Département de Neurobiologie et Génétique, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/Inserm, université de Strasbourg, BP 10142, 67404 Illkirch cedex, France
| | | | | |
Collapse
|
217
|
Eriguchi M, Mizuta H, Luo S, Kuroda Y, Hara H, Rubinsztein DC. α Pix enhances mutant huntingtin aggregation. J Neurol Sci 2010; 290:80-5. [DOI: 10.1016/j.jns.2009.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 11/03/2009] [Accepted: 11/09/2009] [Indexed: 11/28/2022]
|
218
|
Lemmens I, Lievens S, Tavernier J. Strategies towards high-quality binary protein interactome maps. J Proteomics 2010; 73:1415-20. [PMID: 20153845 DOI: 10.1016/j.jprot.2010.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 02/01/2010] [Accepted: 02/05/2010] [Indexed: 01/03/2023]
Abstract
Many processes in a cell depend on protein-protein interactions (PPIs) and perturbations of these interactions can lead to diseases. Comprehensive knowledge of PPI networks will not only give us information on how the cell is organized, but will also provide new drug targets. Current binary PPI networks are mainly generated by high-throughput yeast two-hybrid. Due to the small overlap of these maps, it has long been assumed that these maps are of low quality containing many false positives. However, by using an orthogonal two-hybrid method, MAPPIT (mammalian protein-protein interaction trap), these maps were shown to be of high quality suggesting that the limited overlap is likely due to low sensitivity and not to low specificity.
Collapse
Affiliation(s)
- Irma Lemmens
- Department of Medical Protein Research, VIB, Ghent, Belgium
| | | | | |
Collapse
|
219
|
F-actin binding regions on the androgen receptor and huntingtin increase aggregation and alter aggregate characteristics. PLoS One 2010; 5:e9053. [PMID: 20140226 PMCID: PMC2816219 DOI: 10.1371/journal.pone.0009053] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 12/30/2009] [Indexed: 11/19/2022] Open
Abstract
Protein aggregation is associated with neurodegeneration. Polyglutamine expansion diseases such as spinobulbar muscular atrophy and Huntington disease feature proteins that are destabilized by an expanded polyglutamine tract in their N-termini. It has previously been reported that intracellular aggregation of these target proteins, the androgen receptor (AR) and huntingtin (Htt), is modulated by actin-regulatory pathways. Sequences that flank the polyglutamine tract of AR and Htt might influence protein aggregation and toxicity through protein-protein interactions, but this has not been studied in detail. Here we have evaluated an N-terminal 127 amino acid fragment of AR and Htt exon 1. The first 50 amino acids of ARN127 and the first 14 amino acids of Htt exon 1 mediate binding to filamentous actin in vitro. Deletion of these actin-binding regions renders the polyglutamine-expanded forms of ARN127 and Htt exon 1 less aggregation-prone, and increases the SDS-solubility of aggregates that do form. These regions thus appear to alter the aggregation frequency and type of polyglutamine-induced aggregation. These findings highlight the importance of flanking sequences in determining the propensity of unstable proteins to misfold.
Collapse
|
220
|
Nibbe RK, Koyutürk M, Chance MR. An integrative -omics approach to identify functional sub-networks in human colorectal cancer. PLoS Comput Biol 2010; 6:e1000639. [PMID: 20090827 PMCID: PMC2797084 DOI: 10.1371/journal.pcbi.1000639] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Accepted: 12/11/2009] [Indexed: 02/03/2023] Open
Abstract
Emerging evidence indicates that gene products implicated in human cancers often cluster together in “hot spots” in protein-protein interaction (PPI) networks. Additionally, small sub-networks within PPI networks that demonstrate synergistic differential expression with respect to tumorigenic phenotypes were recently shown to be more accurate classifiers of disease progression when compared to single targets identified by traditional approaches. However, many of these studies rely exclusively on mRNA expression data, a useful but limited measure of cellular activity. Proteomic profiling experiments provide information at the post-translational level, yet they generally screen only a limited fraction of the proteome. Here, we demonstrate that integration of these complementary data sources with a “proteomics-first” approach can enhance the discovery of candidate sub-networks in cancer that are well-suited for mechanistic validation in disease. We propose that small changes in the mRNA expression of multiple genes in the neighborhood of a protein-hub can be synergistically associated with significant changes in the activity of that protein and its network neighbors. Further, we hypothesize that proteomic targets with significant fold change between phenotype and control may be used to “seed” a search for small PPI sub-networks that are functionally associated with these targets. To test this hypothesis, we select proteomic targets having significant expression changes in human colorectal cancer (CRC) from two independent 2-D gel-based screens. Then, we use random walk based models of network crosstalk and develop novel reference models to identify sub-networks that are statistically significant in terms of their functional association with these proteomic targets. Subsequently, using an information-theoretic measure, we evaluate synergistic changes in the activity of identified sub-networks based on genome-wide screens of mRNA expression in CRC. Cross-classification experiments to predict disease class show excellent performance using only a few sub-networks, underwriting the strength of the proposed approach in discovering relevant and reproducible sub-networks. Intensive research on cancer has led to an understanding of many individual genes that may be important for the initiation and progression of tumors. However, since cancer is a progressive disease that results from accumulation of multiple mutations likely acting in concert, individual markers can only provide limited insights into cellular mechanisms that underlie tumorigenesis. For this reason, recent studies focus on identification of “sub-network markers”, that is, functionally associated genes that exhibit coordinate changes in molecular expression during cancer progression. However, expression of genes is most frequently interrogated at the mRNA level, which captures functional activity of genes only to a limited extent. Screening of protein expression, on the other hand, provides information on the abundance of functional gene products, but its scale is often limited compared to screening of mRNA expression. In this article, we develop a proteomics-driven computational method that searches for sub-network markers in human colorectal cancer, based on a seed of differentially expressed proteins identified by proteomic screening. Our results show that significant changes in the expression of these proteins is likely to be associated with coordinate changes in the expression of the genes whose products are functionally associated with these proteins. This analysis leads to novel insights in the synergistic processes that underlie tumorigenesis.
Collapse
Affiliation(s)
- Rod K Nibbe
- Center for Proteomics & Bioinformatics, Case Western Reserve University, Cleveland, Ohio, United States of America.
| | | | | |
Collapse
|
221
|
Menon P, Deane R, Sagare A, Lane SM, Zarcone TJ, O'Dell MR, Yan C, Zlokovic BV, Berk BC. Impaired spine formation and learning in GPCR kinase 2 interacting protein-1 (GIT1) knockout mice. Brain Res 2010; 1317:218-26. [PMID: 20043896 DOI: 10.1016/j.brainres.2009.11.084] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 11/18/2009] [Accepted: 11/18/2009] [Indexed: 11/29/2022]
Abstract
The G-protein coupled receptor (GPCR)-kinase interacting proteins 1 and 2 (GIT1 and GIT2) are scaffold proteins with ADP-ribosylating factor GTPase activity. GIT1 and GIT2 control numerous cellular functions and are highly expressed in neurons, endothelial cells and vascular smooth muscle cells. GIT1 promotes dendritic spine formation, growth and motility in cultured neurons, but its role in brain in vivo is unknown. By using global GIT1 knockout mice (GIT1 KO), we show that compared to WT controls, deletion of GIT1 results in markedly reduced dendritic length and spine density in the hippocampus by 36.7% (p<0.0106) and 35.1% (p<0.0028), respectively. This correlated with their poor adaptation to new environments as shown by impaired performance on tasks dependent on learning. We also studied the effect of GIT1 gene deletion on brain microcirculation. In contrast to findings in systemic circulation, GIT1 KO mice had an intact blood-brain barrier and normal regional cerebral blood flow as determined with radiotracers. Thus, our data suggest that GIT1 plays an important role in brain in vivo by regulating spine density involved in synaptic plasticity that is required for processes involved in learning.
Collapse
Affiliation(s)
- Prashanthi Menon
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Nguyen L, Humbert S, Saudou F, Chariot A. Elongator - an emerging role in neurological disorders. Trends Mol Med 2009; 16:1-6. [PMID: 20036197 DOI: 10.1016/j.molmed.2009.11.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 11/04/2009] [Accepted: 11/17/2009] [Indexed: 10/20/2022]
Abstract
Neurological disorders are becoming a major public health issue in our aging society. An important objective is to understand the molecular events that underlie these diseases to prevent their onset and/or halt their progression. Acetylation of alpha-tubulin is a post-translational modification of microtubules that serves as a recognition signal for the anchoring of molecular motors and, as such, underlies the transport of various proteins or organelles in neurons. This process is affected in striatal and cortical neurons from Huntington's disease patients. Recent studies have shown that Elp3, the catalytic subunit of the Elongator complex, promotes the acetylation of alpha-tubulin in microtubules. Elongator complex activity is impaired in patients with familial dysautonomia. Based on converging experimental and clinical evidence, we propose that Elongator might be commonly targeted in different neurological disorders, and thus might represent a strong candidate for research and development efforts to design drug-based therapies.
Collapse
Affiliation(s)
- Laurent Nguyen
- Interdisciplinary Cluster for Applied Genoproteomics (GIGA-Research), University of Liège, C.H.U. Sart Tilman, Liège, Belgium.
| | | | | | | |
Collapse
|
223
|
Li M, Huang Y, Ma AAK, Lin E, Diamond MI. Y-27632 improves rotarod performance and reduces huntingtin levels in R6/2 mice. Neurobiol Dis 2009; 36:413-20. [DOI: 10.1016/j.nbd.2009.06.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 06/24/2009] [Accepted: 06/28/2009] [Indexed: 01/12/2023] Open
|
224
|
Lievens S, Lemmens I, Tavernier J. Mammalian two-hybrids come of age. Trends Biochem Sci 2009; 34:579-88. [PMID: 19786350 DOI: 10.1016/j.tibs.2009.06.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 06/12/2009] [Accepted: 06/12/2009] [Indexed: 12/22/2022]
Abstract
A diverse series of mammalian two-hybrid technologies for the detection of protein-protein interactions have emerged in the past few years, complementing the established yeast two-hybrid approach. Given the mammalian background in which they operate, these assays open new avenues to study the dynamics of mammalian protein interaction networks, i.e. the temporal, spatial and functional modulation of protein-protein associations. In addition, novel assay formats are available that enable high-throughput mammalian two-hybrid applications, facilitating their use in large-scale interactome mapping projects. Finally, as they can be applied in drug discovery and development programs, these techniques also offer exciting new opportunities for biomedical research.
Collapse
Affiliation(s)
- Sam Lievens
- Department of Medical Protein Research, VIB, A. Baertsoenkaai 3, 9000 Ghent, Belgium
| | | | | |
Collapse
|
225
|
Pegoraro G, Kubben N, Wickert U, Göhler H, Hoffmann K, Misteli T. Ageing-related chromatin defects through loss of the NURD complex. Nat Cell Biol 2009; 11:1261-7. [PMID: 19734887 PMCID: PMC2779731 DOI: 10.1038/ncb1971] [Citation(s) in RCA: 216] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 06/25/2009] [Indexed: 12/16/2022]
Abstract
Physiological and premature aging are characterized by multiple defects in chromatin structure and accumulation of persistent DNA damage. Here we identify the NURD remodeling complex as a key modulator of these aging-associated chromatin defects. We demonstrate loss of several NURD components during premature and normal aging and we find aging-associated reduction of HDAC1 activity. Silencing of individual NURD subunits recapitulates some chromatin defects associated with aging and we provide evidence that structural chromatin defects precede DNA damage accumulation. These results outline a molecular mechanism for chromatin defects during aging.
Collapse
|
226
|
Linghu B, Snitkin ES, Hu Z, Xia Y, Delisi C. Genome-wide prioritization of disease genes and identification of disease-disease associations from an integrated human functional linkage network. Genome Biol 2009; 10:R91. [PMID: 19728866 PMCID: PMC2768980 DOI: 10.1186/gb-2009-10-9-r91] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2009] [Revised: 07/09/2009] [Accepted: 09/03/2009] [Indexed: 11/16/2022] Open
Abstract
An evidence-weighted functional-linkage network of human genes reveals associations among diseases that share no known disease genes and have dissimilar phenotypes
We integrate 16 genomic features to construct an evidence-weighted functional-linkage network comprising 21,657 human genes. The functional-linkage network is used to prioritize candidate genes for 110 diseases, and to reliably disclose hidden associations between disease pairs having dissimilar phenotypes, such as hypercholesterolemia and Alzheimer's disease. Many of these disease-disease associations are supported by epidemiology, but with no previous genetic basis. Such associations can drive novel hypotheses on molecular mechanisms of diseases and therapies.
Collapse
Affiliation(s)
- Bolan Linghu
- Bioinformatics Program, Boston University, 24 Cummington Street, Boston, MA 02215, USA.
| | | | | | | | | |
Collapse
|
227
|
Neuroproteomics: understanding the molecular organization and complexity of the brain. Nat Rev Neurosci 2009; 10:635-46. [DOI: 10.1038/nrn2701] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
228
|
Zhang S, Hisatsune C, Matsu-Ura T, Mikoshiba K. G-protein-coupled receptor kinase-interacting proteins inhibit apoptosis by inositol 1,4,5-triphosphate receptor-mediated Ca2+ signal regulation. J Biol Chem 2009; 284:29158-69. [PMID: 19706611 DOI: 10.1074/jbc.m109.041509] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The inositol 1,4,5-trisphosphate (IP(3)) receptor (IP(3)R) is an intracellular IP(3)-gated calcium (Ca(2+)) release channel and plays important roles in regulation of numerous Ca(2+)-dependent cellular responses. Many intracellular modulators and IP(3)R-binding proteins regulate the IP(3)R channel function. Here we identified G-protein-coupled receptor kinase-interacting proteins (GIT), GIT1 and GIT2, as novel IP(3)R-binding proteins. We found that both GIT1 and GIT2 directly bind to all three subtypes of IP(3)R. The interaction was favored by the cytosolic Ca(2+) concentration and it functionally inhibited IP(3)R activity. Knockdown of GIT induced and accelerated caspase-dependent apoptosis in both unstimulated and staurosporine-treated cells, which was attenuated by wild-type GIT1 overexpression or pharmacological inhibitors of IP(3)R, but not by a mutant form of GIT1 that abrogates the interaction. Thus, we conclude that GIT inhibits apoptosis by modulating the IP(3)R-mediated Ca(2+) signal through a direct interaction with IP(3)R in a cytosolic Ca(2+)-dependent manner.
Collapse
Affiliation(s)
- Songbai Zhang
- Calcium Oscillation Project, International Cooperative Research Project-Solution Oriented Research for Science and Technology, Japan Science and Technology Agency, 2-1 Hirosawa, Wako City, Saitama 351-0198, Japan.
| | | | | | | |
Collapse
|
229
|
Rudinskiy N, Kaneko YA, Beesen AA, Gokce O, Régulier E, Déglon N, Luthi-Carter R. Diminished hippocalcin expression in Huntington's disease brain does not account for increased striatal neuron vulnerability as assessed in primary neurons. J Neurochem 2009; 111:460-72. [PMID: 19686238 DOI: 10.1111/j.1471-4159.2009.06344.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hippocalcin is a neuronal calcium sensor protein previously implicated in regulating neuronal viability and plasticity. Hippocalcin is the most highly expressed neuronal calcium sensor in the medium spiny striatal output neurons that degenerate selectively in Huntington's disease (HD). We have previously shown that decreased hippocalcin expression occurs in parallel with the onset of disease phenotype in mouse models of HD. Here we show by in situ hybridization histochemistry that hippocalcin RNA is also diminished by 63% in human HD brain. These findings lead us to hypothesize that diminished hippocalcin expression might contribute to striatal neurodegeneration in HD. We tested this hypothesis by assessing whether restoration of hippocalcin expression would decrease striatal neurodegeneration in cellular models of HD comprising primary striatal neurons exposed to mutant huntingtin, the mitochondrial toxin 3-nitropropionic acid or an excitotoxic concentration of glutamate. Counter to our hypothesis, hippocalcin expression did not improve the survival of striatal neurons under these conditions. Likewise, expression of hippocalcin together with interactor proteins including the neuronal apoptosis inhibitory protein did not increase the survival of striatal cells in cellular models of HD. These results indicate that diminished hippocalcin expression does not contribute to HD-related neurodegeneration.
Collapse
Affiliation(s)
- Nikita Rudinskiy
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
230
|
Abstract
The integration of medical information into gene and protein networks could lead to a better understanding of complex diseases. Molecular networks are being used to reconcile genotypes and phenotypes by integrating medical information. In this context, networks will be instrumental for the interpretation of disease at the personalized medicine level.
Collapse
Affiliation(s)
- Anaïs Baudot
- Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), C/Melchor Fernández Almagro 3, E-28029 Madrid, Spain
| | | | | |
Collapse
|
231
|
Layman W, McEwen D, Beyer L, Lalani S, Fernbach S, Oh E, Swaroop A, Hegg C, Raphael Y, Martens J, Martin D. Defects in neural stem cell proliferation and olfaction in Chd7 deficient mice indicate a mechanism for hyposmia in human CHARGE syndrome. Hum Mol Genet 2009; 18:1909-23. [PMID: 19279158 PMCID: PMC2678924 DOI: 10.1093/hmg/ddp112] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 03/02/2009] [Accepted: 03/09/2009] [Indexed: 11/14/2022] Open
Abstract
Mutations in CHD7, a chromodomain gene, are present in a majority of individuals with CHARGE syndrome, a multiple anomaly disorder characterized by ocular Coloboma, Heart defects, Atresia of the choanae, Retarded growth and development, Genital hypoplasia and Ear anomalies. The clinical features of CHARGE syndrome are highly variable and incompletely penetrant. Olfactory dysfunction is a common feature in CHARGE syndrome and has been potentially linked to primary olfactory bulb defects, but no data confirming this mechanistic link have been reported. On the basis of these observations, we hypothesized that loss of Chd7 disrupts mammalian olfactory tissue development and function. We found severe defects in olfaction in individuals with CHD7 mutations and CHARGE, and loss of odor evoked electro-olfactogram responses in Chd7 deficient mice, suggesting reduced olfaction is due to a dysfunctional olfactory epithelium. Chd7 expression was high in basal olfactory epithelial neural stem cells and down-regulated in mature olfactory sensory neurons. We observed smaller olfactory bulbs, reduced olfactory sensory neurons, and disorganized epithelial ultrastructure in Chd7 mutant mice, despite apparently normal functional cilia and sustentacular cells. Significant reductions in the proliferation of neural stem cells and regeneration of olfactory sensory neurons in the mature Chd7(Gt/+) olfactory epithelium indicate critical roles for Chd7 in regulating neurogenesis. These studies provide evidence that mammalian olfactory dysfunction due to Chd7 haploinsufficiency is linked to primary defects in olfactory neural stem cell proliferation and may influence olfactory bulb development.
Collapse
Affiliation(s)
| | | | | | - S.R. Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - S.D. Fernbach
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - E. Oh
- Neurobiology Neurodegeneration and Repair Laboratory (N-NRL), National Eye Institute / NIH, Bethesda, MD 20892, USA
| | - A. Swaroop
- Neurobiology Neurodegeneration and Repair Laboratory (N-NRL), National Eye Institute / NIH, Bethesda, MD 20892, USA
| | - C.C. Hegg
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | | | | | - D.M. Martin
- Department of Human Genetics
- Department of Pediatrics, University of Michigan Medical Center, 3520A Medical Science Research Building I, Ann Arbor, MI 48109-5652, USA
| |
Collapse
|
232
|
Raychaudhuri S, Dey S, Bhattacharyya NP, Mukhopadhyay D. The role of intrinsically unstructured proteins in neurodegenerative diseases. PLoS One 2009; 4:e5566. [PMID: 19440375 PMCID: PMC2679209 DOI: 10.1371/journal.pone.0005566] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Accepted: 03/24/2009] [Indexed: 11/19/2022] Open
Abstract
The number and importance of intrinsically disordered proteins (IUP), known to be involved in various human disorders, are growing rapidly. To test for the generalized implications of intrinsic disorders in proteins involved in Neurodegenerative diseases, disorder prediction tools have been applied to three datasets comprising of proteins involved in Huntington Disease (HD), Parkinson's disease (PD), Alzheimer's disease (AD). Results show, in general, proteins in disease datasets possess significantly enhanced intrinsic unstructuredness. Most of these disordered proteins in the disease datasets are found to be involved in neuronal activities, signal transduction, apoptosis, intracellular traffic, cell differentiation etc. Also these proteins are found to have more number of interactors and hence as the proportion of disorderedness (i.e., the length of the unfolded stretch) increased, the size of the interaction network simultaneously increased. All these observations reflect that, “Moonlighting” i.e. the contextual acquisition of different structural conformations (transient), eventually may allow these disordered proteins to act as network “hubs” and thus they may have crucial influences in the pathogenecity of neurodegenerative diseases.
Collapse
Affiliation(s)
- Swasti Raychaudhuri
- Structural Genomics Section, Saha Institute of Nuclear Physics, Kolkata, India
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - Sucharita Dey
- Structural Genomics Section, Saha Institute of Nuclear Physics, Kolkata, India
| | - Nitai P. Bhattacharyya
- Structural Genomics Section, Saha Institute of Nuclear Physics, Kolkata, India
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - Debashis Mukhopadhyay
- Structural Genomics Section, Saha Institute of Nuclear Physics, Kolkata, India
- * E-mail:
| |
Collapse
|
233
|
A dynamic network approach for the study of human phenotypes. PLoS Comput Biol 2009; 5:e1000353. [PMID: 19360091 PMCID: PMC2661364 DOI: 10.1371/journal.pcbi.1000353] [Citation(s) in RCA: 437] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 03/09/2009] [Indexed: 12/11/2022] Open
Abstract
The use of networks to integrate different genetic, proteomic, and metabolic
datasets has been proposed as a viable path toward elucidating the origins of
specific diseases. Here we introduce a new phenotypic database summarizing
correlations obtained from the disease history of more than 30 million patients
in a Phenotypic Disease Network (PDN). We present evidence that the structure of
the PDN is relevant to the understanding of illness progression by showing that
(1) patients develop diseases close in the network to those they already have;
(2) the progression of disease along the links of the network is different for
patients of different genders and ethnicities; (3) patients diagnosed with
diseases which are more highly connected in the PDN tend to die sooner than
those affected by less connected diseases; and (4) diseases that tend to be
preceded by others in the PDN tend to be more connected than diseases that
precede other illnesses, and are associated with higher degrees of mortality.
Our findings show that disease progression can be represented and studied using
network methods, offering the potential to enhance our understanding of the
origin and evolution of human diseases. The dataset introduced here, released
concurrently with this publication, represents the largest relational phenotypic
resource publicly available to the research community. To help the understanding of physiological failures, diseases are defined as
specific sets of phenotypes affecting one or several physiological systems. Yet,
the complexity of biological systems implies that our working definitions of
diseases are careful discretizations of a complex phenotypic space. To reconcile
the discrete nature of diseases with the complexity of biological organisms, we
need to understand how diseases are connected, as connections between these
different discrete categories can be informative about the mechanisms causing
physiological failures. Here we introduce the Phenotypic Disease Network (PDN)
as a map summarizing phenotypic connections between diseases and show that
diseases progress preferentially along the links of this map. Furthermore, we
show that this progression is different for patients with different genders and
racial backgrounds and that patients affected by diseases that are connected to
many other diseases in the PDN tend to die sooner than those affected by less
connected diseases. Additionally, we have created a queryable online database
(http://hudine.neu.edu/) of the 18 different datasets generated
from the more than 31 million patients in this study. The disease associations
can be explored online or downloaded in bulk.
Collapse
|
234
|
Saleh N, Moutereau S, Durr A, Krystkowiak P, Azulay JP, Tranchant C, Broussolle E, Morin F, Bachoud-Lévi AC, Maison P. Neuroendocrine disturbances in Huntington's disease. PLoS One 2009; 4:e4962. [PMID: 19319184 PMCID: PMC2655649 DOI: 10.1371/journal.pone.0004962] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 02/02/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Huntington's disease (HD) is a severe inherited neurodegenerative disorder characterized, in addition to neurological impairment, by weight loss suggesting endocrine disturbances. The aims of this study were to look for neuroendocrine disturbances in patients with Huntington's disease (HD) and to determine the relationship with weight loss seen in HD METHODS AND FINDING: We compared plasma levels of hormones from the five pituitary axes in 219 patients with genetically documented HD and in 71 sex- and age-matched controls. Relationships between hormone levels and disease severity, including weight-loss severity, were evaluated. Growth hormone (GH) and standard deviation score of insulin-like growth factor 1 (SDS IGF-1) were significantly higher in patients than in controls (0.25 (0.01-5.89) vs. 0.15 (0.005-4.89) ng/ml, p = 0.013 and 0.16+/-1.02 vs. 0.06+/-0.91, p = 0.039; respectively). Cortisol was higher (p = 0.002) in patients (399.14+/-160.5 nmol/L vs. 279.8+/-130.1 nmol/L), whereas no differences were found for other hormone axes. In patients, elevations in GH and IGF-1 and decreases in thyroid-stimulating hormone, free triiodothyronine and testosterone (in men) were associated with severity of impairments (Independence scale, Functional score, Total Functional Capacity, Total Motor score, Behavioral score). Only GH was independently associated with body mass index (beta = -0.26, p = 0.001). CONCLUSION Our data suggest that the thyrotropic and in men gonadotropic axes are altered in HD according to the severity of the disease. The somatotropic axis is overactive even in patients with early disease, and could be related to the weight loss seen in HD patients.
Collapse
Affiliation(s)
- Nadine Saleh
- Centre de référence maladie de Huntington, AP-HP, Groupe Henri-Mondor Albert-Chenevier/Pitié Salpétrière, Créteil, Paris, France
- Service Pharmacologie Clinique, AP-HP, Groupe Henri-Mondor Albert-Chenevier, Creteil, France
- Universite Paris 12, Faculte de Medecine, Creteil, France
- INSERM, Unite U955, Equipe 1, Creteil, France
| | - Stéphane Moutereau
- Service de Biochimie, AP-HP, Groupe Henri-Mondor Albert-Chenevier, Creteil, France
| | - Alexandra Durr
- Centre de référence maladie de Huntington, AP-HP, Groupe Henri-Mondor Albert-Chenevier/Pitié Salpétrière, Créteil, Paris, France
- INSERM U289, Department of Genetics, Cytogenetics and Embryology, Pitié Salpétrière, Paris, France
| | | | | | | | | | | | - Anne-Catherine Bachoud-Lévi
- Centre de référence maladie de Huntington, AP-HP, Groupe Henri-Mondor Albert-Chenevier/Pitié Salpétrière, Créteil, Paris, France
- Universite Paris 12, Faculte de Medecine, Creteil, France
- INSERM, Unite U955, Equipe 1, Creteil, France
- Ecole normale Supérieure, Département d'études Cognitives, Paris, France
- * E-mail:
| | - Patrick Maison
- Service Pharmacologie Clinique, AP-HP, Groupe Henri-Mondor Albert-Chenevier, Creteil, France
- Universite Paris 12, Faculte de Medecine, Creteil, France
- INSERM, Unite U955, Equipe 1, Creteil, France
- Unité de recherche clinique, AP-HP, Groupe Henri-Mondor Albert-Chenevier, Creteil, France
| |
Collapse
|
235
|
Yosef N, Ungar L, Zalckvar E, Kimchi A, Kupiec M, Ruppin E, Sharan R. Toward accurate reconstruction of functional protein networks. Mol Syst Biol 2009; 5:248. [PMID: 19293828 PMCID: PMC2671920 DOI: 10.1038/msb.2009.3] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Accepted: 01/07/2009] [Indexed: 01/04/2023] Open
Abstract
Genome-scale screening studies are gradually accumulating a wealth of data on the putative involvement of hundreds of genes/proteins in various cellular responses or functions. A fundamental challenge is to chart out the protein pathways that underlie these systems. Previous approaches to the problem have either employed a local optimization criterion, aiming to infer each pathway independently, or a global criterion, searching for the overall most parsimonious subnetwork. Here, we study the trade-off between the two approaches and present a new intermediary scheme that provides explicit control over it. We demonstrate its utility in the analysis of the apoptosis network in humans, and the telomere length maintenance (TLM) system in yeast. Our results show that in the majority of real-life cases, the intermediary approach provides the most plausible solutions. We use a new set of perturbation experiments measuring the role of essential genes in telomere length regulation to further study the TLM network. Surprisingly, we find that the proteasome plays an important role in telomere length regulation through its associations with transcription and DNA repair circuits.
Collapse
Affiliation(s)
- Nir Yosef
- The Blavatnik School of Computer Science, Tel-Aviv University, Tel-Aviv, Israel.
| | | | | | | | | | | | | |
Collapse
|
236
|
Bell R, Hubbard A, Chettier R, Chen D, Miller JP, Kapahi P, Tarnopolsky M, Sahasrabuhde S, Melov S, Hughes RE. A human protein interaction network shows conservation of aging processes between human and invertebrate species. PLoS Genet 2009; 5:e1000414. [PMID: 19293945 PMCID: PMC2657003 DOI: 10.1371/journal.pgen.1000414] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Accepted: 02/10/2009] [Indexed: 01/08/2023] Open
Abstract
We have mapped a protein interaction network of human homologs of proteins that modify longevity in invertebrate species. This network is derived from a proteome-scale human protein interaction Core Network generated through unbiased high-throughput yeast two-hybrid searches. The longevity network is composed of 175 human homologs of proteins known to confer increased longevity through loss of function in yeast, nematode, or fly, and 2,163 additional human proteins that interact with these homologs. Overall, the network consists of 3,271 binary interactions among 2,338 unique proteins. A comparison of the average node degree of the human longevity homologs with random sets of proteins in the Core Network indicates that human homologs of longevity proteins are highly connected hubs with a mean node degree of 18.8 partners. Shortest path length analysis shows that proteins in this network are significantly more connected than would be expected by chance. To examine the relationship of this network to human aging phenotypes, we compared the genes encoding longevity network proteins to genes known to be changed transcriptionally during aging in human muscle. In the case of both the longevity protein homologs and their interactors, we observed enrichments for differentially expressed genes in the network. To determine whether homologs of human longevity interacting proteins can modulate life span in invertebrates, homologs of 18 human FRAP1 interacting proteins showing significant changes in human aging muscle were tested for effects on nematode life span using RNAi. Of 18 genes tested, 33% extended life span when knocked-down in Caenorhabditis elegans. These observations indicate that a broad class of longevity genes identified in invertebrate models of aging have relevance to human aging. They also indicate that the longevity protein interaction network presented here is enriched for novel conserved longevity proteins. Studies of longevity in model organisms such as baker's yeast, roundworm, and fruit fly have clearly demonstrated that a diverse array of genetic mutations can result in increased life span. In fact, large-scale genetic screens have identified hundreds of genes that when mutated, knocked down, or deleted will significantly enhance longevity in these organisms. Despite great progress in understanding genetic and genomic determinants of life span in model organisms, the general relevance of invertebrate longevity genes to human aging and longevity has yet to be fully established. In this study, we show that human homologs of invertebrate longevity genes change in their expression levels during aging in human tissue. We also show that human genes encoding proteins that interact with human longevity homolog proteins are also changed in expression during human aging. These observations taken together indicate that the broad patterns underlying genetic control of life span in invertebrates is highly relevant to human aging and longevity. We also present a collection of novel candidate genes and proteins that may influence human life span.
Collapse
Affiliation(s)
- Russell Bell
- Prolexys Pharmaceuticals, Salt Lake City, Utah, United States of America
| | - Alan Hubbard
- School of Public Health, University of California Berkeley, Berkeley, California, United States of America
- Buck Institute for Age Research, Novato, California, United States of America
| | - Rakesh Chettier
- Prolexys Pharmaceuticals, Salt Lake City, Utah, United States of America
| | - Di Chen
- Buck Institute for Age Research, Novato, California, United States of America
| | - John P. Miller
- Buck Institute for Age Research, Novato, California, United States of America
| | - Pankaj Kapahi
- Buck Institute for Age Research, Novato, California, United States of America
| | | | | | - Simon Melov
- Buck Institute for Age Research, Novato, California, United States of America
| | - Robert E. Hughes
- Buck Institute for Age Research, Novato, California, United States of America
- * E-mail:
| |
Collapse
|
237
|
Yeger-Lotem E, Riva L, Su LJ, Gitler AD, Cashikar A, King OD, Auluck PK, Geddie ML, Valastyan JS, Karger DR, Lindquist S, Fraenkel E. Bridging high-throughput genetic and transcriptional data reveals cellular responses to alpha-synuclein toxicity. Nat Genet 2009; 41:316-23. [PMID: 19234470 PMCID: PMC2733244 DOI: 10.1038/ng.337] [Citation(s) in RCA: 226] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 01/27/2009] [Indexed: 02/07/2023]
Abstract
Cells respond to stimuli by changes in various processes, including signaling pathways and gene expression. Efforts to identify components of these responses increasingly depend on mRNA profiling and genetic library screens. By comparing the results of these two assays across various stimuli, we found that genetic screens tend to identify response regulators, whereas mRNA profiling frequently detects metabolic responses. We developed an integrative approach that bridges the gap between these data using known molecular interactions, thus highlighting major response pathways. We used this approach to reveal cellular pathways responding to the toxicity of alpha-synuclein, a protein implicated in several neurodegenerative disorders including Parkinson's disease. For this we screened an established yeast model to identify genes that when overexpressed alter alpha-synuclein toxicity. Bridging these data and data from mRNA profiling provided functional explanations for many of these genes and identified previously unknown relations between alpha-synuclein toxicity and basic cellular pathways.
Collapse
Affiliation(s)
- Esti Yeger-Lotem
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142 USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Laura Riva
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Linhui Julie Su
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142 USA
| | - Aaron D. Gitler
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142 USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anil Cashikar
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142 USA
| | - Oliver D. King
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142 USA
| | - Pavan K. Auluck
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142 USA
- Department of Pathology and Neurology, Massachusetts General Hospital, Boston, MA 02114 and Harvard Medical School, Boston MA 02115 USA
| | - Melissa L. Geddie
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142 USA
| | - Julie S. Valastyan
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142 USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David R. Karger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142 USA
- Howard Hughes Medical Institute, Cambridge, MA 02142 USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
238
|
Chen J, Aronow BJ, Jegga AG. Disease candidate gene identification and prioritization using protein interaction networks. BMC Bioinformatics 2009; 10:73. [PMID: 19245720 PMCID: PMC2657789 DOI: 10.1186/1471-2105-10-73] [Citation(s) in RCA: 240] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Accepted: 02/27/2009] [Indexed: 12/22/2022] Open
Abstract
Background Although most of the current disease candidate gene identification and prioritization methods depend on functional annotations, the coverage of the gene functional annotations is a limiting factor. In the current study, we describe a candidate gene prioritization method that is entirely based on protein-protein interaction network (PPIN) analyses. Results For the first time, extended versions of the PageRank and HITS algorithms, and the K-Step Markov method are applied to prioritize disease candidate genes in a training-test schema. Using a list of known disease-related genes from our earlier study as a training set ("seeds"), and the rest of the known genes as a test list, we perform large-scale cross validation to rank the candidate genes and also evaluate and compare the performance of our approach. Under appropriate settings – for example, a back probability of 0.3 for PageRank with Priors and HITS with Priors, and step size 6 for K-Step Markov method – the three methods achieved a comparable AUC value, suggesting a similar performance. Conclusion Even though network-based methods are generally not as effective as integrated functional annotation-based methods for disease candidate gene prioritization, in a one-to-one comparison, PPIN-based candidate gene prioritization performs better than all other gene features or annotations. Additionally, we demonstrate that methods used for studying both social and Web networks can be successfully used for disease candidate gene prioritization.
Collapse
Affiliation(s)
- Jing Chen
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | | | | |
Collapse
|
239
|
Zabel C, Mao L, Woodman B, Rohe M, Wacker MA, Kläre Y, Koppelstätter A, Nebrich G, Klein O, Grams S, Strand A, Luthi-Carter R, Hartl D, Klose J, Bates GP. A large number of protein expression changes occur early in life and precede phenotype onset in a mouse model for huntington disease. Mol Cell Proteomics 2008; 8:720-34. [PMID: 19043139 DOI: 10.1074/mcp.m800277-mcp200] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Huntington disease (HD) is fatal in humans within 15-20 years of symptomatic disease. Although late stage HD has been studied extensively, protein expression changes that occur at the early stages of disease and during disease progression have not been reported. In this study, we used a large two-dimensional gel/mass spectrometry-based proteomics approach to investigate HD-induced protein expression alterations and their kinetics at very early stages and during the course of disease. The murine HD model R6/2 was investigated at 2, 4, 6, 8, and 12 weeks of age, corresponding to absence of disease and early, intermediate, and late stage HD. Unexpectedly the most HD stage-specific protein changes (71-100%) as well as a drastic alteration (almost 6% of the proteome) in protein expression occurred already as early as 2 weeks of age. Early changes included mainly the up-regulation of proteins involved in glycolysis/gluconeogenesis and the down-regulation of the actin cytoskeleton. This suggests a period of highly variable protein expression that precedes the onset of HD phenotypes. Although an up-regulation of glycolysis/gluconeogenesis-related protein alterations remained dominant during HD progression, late stage alterations at 12 weeks showed an up-regulation of proteins involved in proteasomal function. The early changes in HD coincide with a peak in protein alteration during normal mouse development at 2 weeks of age that may be responsible for these massive changes. Protein and mRNA data sets showed a large overlap on the level of affected pathways but not single proteins/mRNAs. Our observations suggest that HD is characterized by a highly dynamic disease pathology not represented by linear protein concentration alterations over the course of disease.
Collapse
Affiliation(s)
- Claus Zabel
- Institute for Human Genetics, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
McCue PP, Phang JM. Identification of human intracellular targets of the medicinal Herb St. John's Wort by chemical-genetic profiling in yeast. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2008; 56:11011-7. [PMID: 18975972 PMCID: PMC2645918 DOI: 10.1021/jf801593a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
St. John's wort (SJW; Hypericum perforatum L.) is commonly known for its antidepressant properties and was traditionally used to promote wound healing, but its molecular mechanism of action is not known. Here, chemical-genetic profiling in yeast was used to predict the human intracellular targets of an aqueous extract of SJW. SJW source material was authenticated by TLC, digital microscopy, and HPLC and further characterized by colorimetric methods for antioxidant activity, protein content, and total soluble phenolic content. SJW extract contained 1.76 microg/mL hyperforin, 10.14 microg/mL hypericin, and 46.05 microg/mL pseudohypericin. The effect of SJW extract on approximately 5900 barcoded heterozygous diploid deletion strains of Saccharomyces cerevisiae was investigated using high-density oligonucleotide microarrays. Seventy-eight yeast genes were identified as sensitive to SJW and were primarily associated with vesicle-mediated transport and signal transduction pathways. Potential human intracellular targets were identified using sequence-based comparisons and included proteins associated with neurological disease and angiogenesis-related pathways. Selected human targets were confirmed by cell-based immunocytochemical assays. The comprehensive and systematic nature of chemical-genetic profiling in yeast makes this technique attractive for elucidating the potential molecular mechanisms of action of botanical medicines and other bioactive dietary plants.
Collapse
Affiliation(s)
- Patrick P McCue
- National Center for Complementary and Alternative Medicine, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
241
|
Wall DP, Esteban FJ, Deluca TF, Huyck M, Monaghan T, Velez de Mendizabal N, Goñí J, Kohane IS. Comparative analysis of neurological disorders focuses genome-wide search for autism genes. Genomics 2008; 93:120-9. [PMID: 18950700 DOI: 10.1016/j.ygeno.2008.09.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2008] [Revised: 09/19/2008] [Accepted: 09/23/2008] [Indexed: 11/30/2022]
Abstract
The behaviors of autism overlap with a diverse array of other neurological disorders, suggesting common molecular mechanisms. We conducted a large comparative analysis of the network of genes linked to autism with those of 432 other neurological diseases to circumscribe a multi-disorder subcomponent of autism. We leveraged the biological process and interaction properties of these multi-disorder autism genes to overcome the across-the-board multiple hypothesis corrections that a purely data-driven approach requires. Using prior knowledge of biological process, we identified 154 genes not previously linked to autism of which 42% were significantly differentially expressed in autistic individuals. Then, using prior knowledge from interaction networks of disorders related to autism, we uncovered 334 new genes that interact with published autism genes, of which 87% were significantly differentially regulated in autistic individuals. Our analysis provided a novel picture of autism from the perspective of related neurological disorders and suggested a model by which prior knowledge of interaction networks can inform and focus genome-scale studies of complex neurological disorders.
Collapse
Affiliation(s)
- D P Wall
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
242
|
Chaurasia G, Malhotra S, Russ J, Schnoegl S, Hänig C, Wanker EE, Futschik ME. UniHI 4: new tools for query, analysis and visualization of the human protein-protein interactome. Nucleic Acids Res 2008; 37:D657-60. [PMID: 18984619 PMCID: PMC2686569 DOI: 10.1093/nar/gkn841] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Human protein interaction maps have become important tools of biomedical research for the elucidation of molecular mechanisms and the identification of new modulators of disease processes. The Unified Human Interactome database (UniHI, http://www.unihi.org) provides researchers with a comprehensive platform to query and access human protein–protein interaction (PPI) data. Since its first release, UniHI has considerably increased in size. The latest update of UniHI includes over 250 000 interactions between ∼22 300 unique proteins collected from 14 major PPI sources. However, this wealth of data also poses new challenges for researchers due to the complexity of interaction networks retrieved from the database. We therefore developed several new tools to query, analyze and visualize human PPI networks. Most importantly, UniHI allows now the construction of tissue-specific interaction networks and focused querying of canonical pathways. This will enable researchers to target their analysis and to prioritize candidate proteins for follow-up studies.
Collapse
Affiliation(s)
- Gautam Chaurasia
- Institute for Theoretical Biology, Charité, Humboldt-University, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
243
|
Bahri SM, Choy JM, Manser E, Lim L, Yang X. The Drosophila homologue of Arf-GAP GIT1, dGIT, is required for proper muscle morphogenesis and guidance during embryogenesis. Dev Biol 2008; 325:15-23. [PMID: 18996366 DOI: 10.1016/j.ydbio.2008.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 08/29/2008] [Accepted: 09/01/2008] [Indexed: 11/18/2022]
Abstract
GIT1-like proteins are GTPase-activating proteins (GAPs) for Arfs and interact with a variety of signaling molecules to function as integrators of pathways controlling cytoskeletal organization and cell motility. In this report, we describe the characterization of a Drosophila homologue of GIT1, dGIT, and show that it is required for proper muscle morphogenesis and myotube guidance in the fly embryo. The dGIT protein is concentrated at the termini of growing myotubes and localizes to muscle attachment sites in late stage embryos. dgit mutant embryos show muscle patterning defects and aberrant targeting in subsets of their muscles. dgit mutant muscles fail to localize the p21-activated kinase, dPak, to their termini. dPak and dGIT form a complex in the presence of dPIX and dpak mutant embryos show similar muscle morphogenesis and targeting phenotypes to that of dgit. We propose that dGIT and dPak are part of a complex that promotes proper muscle morphogenesis and myotube targeting during embryogenesis.
Collapse
Affiliation(s)
- Sami M Bahri
- Institute of Molecular and Cell Biology, Singapore 138673, Singapore.
| | | | | | | | | |
Collapse
|
244
|
Dortay H, Gruhn N, Pfeifer A, Schwerdtner M, Schmülling T, Heyl A. Toward an interaction map of the two-component signaling pathway of Arabidopsis thaliana. J Proteome Res 2008; 7:3649-60. [PMID: 18642946 DOI: 10.1021/pr0703831] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Among the signal transduction pathways in higher eukaryotes, the two-component system (TCS) is unique to plants. In the model plant Arabidopsis thaliana, it consists of more than 30 proteins, including eight receptors, five phosphotransmitters and 23 response regulators. One of its important functions is to perceive and transduce the signal of the plant hormone cytokinin. The basic signal flow within the TCS is well-understood, but it is unclear how this pathway is integrated with the remainder of the proteome. Thus, knowledge about the interactions of TCS proteins should contribute to the understanding of their mode of action. Therefore, we conducted medium-scale yeast two-hybrid screens focusing on those members of the TCS, which are thought to be involved in cytokinin signaling. In total, more than 6.3 x 10 (7) transformants were screened resulting in the identification of 160 different interactions, of which 136 were novel. Most of the interacting proteins belong to the functional categories of signal transduction and protein metabolism. TCS proteins and their interactors localized to the same subcellular compartment in many cases, a prerequisite to being of biological relevance. The resulting interaction network map revealed large differences in the connectivity. Cytokinin receptors (AHK2, CRE1/AHK4) showed the highest numbers of different interaction partners. This study is the first systematic protein-protein interaction experiment for a plant signal system and provides numerous starting points for further analysis of the molecular mechanisms used to convert the signal carried by the TCS into biological processes.
Collapse
Affiliation(s)
- Hakan Dortay
- Institute of Biology/Applied Genetics, Free University of Berlin, Albrecht-Thaer-Weg 6, 14195 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
245
|
Bhattacharyya NP, Banerjee M, Majumder P. Huntington’s disease: roles of huntingtin-interacting protein 1 (HIP-1) and its molecular partner HIPPI in the regulation of apoptosis and transcription. FEBS J 2008; 275:4271-9. [DOI: 10.1111/j.1742-4658.2008.06563.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
246
|
Zolghadr K, Mortusewicz O, Rothbauer U, Kleinhans R, Goehler H, Wanker EE, Cardoso MC, Leonhardt H. A fluorescent two-hybrid assay for direct visualization of protein interactions in living cells. Mol Cell Proteomics 2008; 7:2279-87. [PMID: 18622019 DOI: 10.1074/mcp.m700548-mcp200] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Genetic high throughput screens have yielded large sets of potential protein-protein interactions now to be verified and further investigated. Here we present a simple assay to directly visualize protein-protein interactions in single living cells. Using a modified lac repressor system, we tethered a fluorescent bait at a chromosomal lac operator array and assayed for co-localization of fluorescent prey fusion proteins. With this fluorescent two-hybrid assay we successfully investigated the interaction of proteins from different subcellular compartments including nucleus, cytoplasm, and mitochondria. In combination with an S phase marker we also studied the cell cycle dependence of protein-protein interactions. These results indicate that the fluorescent two-hybrid assay is a powerful tool to investigate protein-protein interactions within their cellular environment and to monitor the response to external stimuli in real time.
Collapse
Affiliation(s)
- Kourosh Zolghadr
- Munich Center for Integrated Protein Science (CiPSM) and Department of Biology, Ludwig Maximilians University Munich, 82152 Planegg-Martinsried, Germany
| | | | | | | | | | | | | | | |
Collapse
|
247
|
Abstract
Y-27632, an inhibitor of the Rho-associated kinase ROCK, is a therapeutic lead for Huntington disease (HD). The downstream targets that mediate its inhibitory effects on huntingtin (Htt) aggregation and toxicity are unknown. We have identified profilin, a small actin-binding factor that also interacts with Htt, as being a direct target of the ROCK1 isoform. The overexpression of profilin reduces the aggregation of polyglutamine-expanded Htt and androgen receptor (AR) peptides. This requires profilin's G-actin binding activity and its direct interaction with Htt, which are both inhibited by the ROCK1-mediated phosphorylation of profilin at Ser-137. Y-27632 blocks the phosphorylation of profilin in HEK293 cells and primary neurons, which maintains profilin in an active state. The knockdown of profilin blocks the inhibitory effect of Y-27632 on both AR and Htt aggregation. A signaling pathway from ROCK1 to profilin thus controls polyglutamine protein aggregation and is targeted by a promising therapeutic lead for HD.
Collapse
|
248
|
Korolainen MA, Corthals GL, Kouvonen P, Kurkinen KMA, Nyman TA. 3rd Annual FinnProt Meeting: from cells to systems. Expert Rev Proteomics 2008; 5:389-91. [PMID: 18532906 DOI: 10.1586/14789450.5.3.389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The Finnish Proteomics Society, FinnProt ( www.finnprot.org ), was founded in November 2004 as the Proteomics Division of the Societas biochemica, biophysica et microbiologica Fenniae ( www.biobio.org ). The mission of FinnProt is to make proteomics research readily available for the large scientific community in Finland, promote research and education in proteomics and protein chemistry, and act as the official Finnish collaborative body to international proteomics organizations such as the European Proteomics Association.
Collapse
Affiliation(s)
- Minna A Korolainen
- Department of Neurology, University of Kuopio, PO Box 1627, FIN-70211, Kuopio, Finland.
| | | | | | | | | |
Collapse
|
249
|
Cavet ME, Pang J, Yin G, Berk BC. An epidermal growth factor (EGF) -dependent interaction between GIT1 and sorting nexin 6 promotes degradation of the EGF receptor. FASEB J 2008; 22:3607-16. [PMID: 18523162 DOI: 10.1096/fj.07-094086] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
G-protein coupled receptor (GPCR) kinase-2 interacting protein 1 (GIT1) is a multifunctional scaffolding protein that regulates epidermal growth factor receptor (EGFR) signaling pathways. We demonstrate that GIT1 interacts with sorting nexin 6 (SNX6), a member of the SNX family that increases EGFR trafficking between endosomes and lysosomes, thereby enhancing EGFR degradation. The GIT1-SNX6 interaction is increased 3-fold after treatment with EGF for 60 min. The second coiled-coil domain (CC2; aa 424-474) of GIT1 mediates binding to SNX6. Subcellular fractionation and confocal microscopy data indicate that GIT1 and SNX6 interact in endosomes. Knockdown of GIT1 expression by small interfering RNA decreased the rate of EGF-induced EGFR degradation. Expression of exogenous GIT1 or SNX6 alone did not alter EGFR degradation; however, coexpression of GIT1 and SNX6 decreased EGFR levels both basally and in response to EGF. In contrast, expression of GIT1(CC2 deleted) and SNX6 did not reduce EGFR levels, demonstrating that the interaction between GIT1 and SNX6 was required to regulate EGFR trafficking. Phosphorylation of the EGFR substrate phospholipase C-gamma was decreased by coexpression of GIT1 and SNX6. These data demonstrate an endosomal, EGF-regulated interaction between SNX6 and GIT1 that enhances degradation of the EGFR, and thereby alters EGFR signaling. Our findings suggest a new role for GIT1 in tyrosine kinase receptor trafficking.
Collapse
Affiliation(s)
- Megan E Cavet
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine, Rochester, New York 14642, USA
| | | | | | | |
Collapse
|
250
|
Frank SR, Hansen SH. The PIX-GIT complex: a G protein signaling cassette in control of cell shape. Semin Cell Dev Biol 2008; 19:234-44. [PMID: 18299239 PMCID: PMC2394276 DOI: 10.1016/j.semcdb.2008.01.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Accepted: 01/16/2008] [Indexed: 01/24/2023]
Abstract
Arf and Rho GTP-binding proteins coordinately regulate membrane dynamics and cytoskeletal rearrangements. The Cdc42/Rac guanine nucleotide exchange factor PIX and the Arf GTPase-activating protein GIT form a stable complex in cells. The PIX-GIT complex functions to integrate signaling among Arf, Cdc42, and Rac proteins in response to cues emanating from integrins, heterotrimeric G proteins, receptor tyrosine kinases, and cell-cell interactions. A concept that emerges from the literature is that the PIX-GIT complex serves as a cassette to elicit changes in cell shape essential for polarized cell responses in a wide range of biological contexts.
Collapse
Affiliation(s)
- Scott R. Frank
- GI Cell Biology Laboratory, The Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Steen H. Hansen
- GI Cell Biology Laboratory, The Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|