1
|
Zhang X, Liu Y, Yang R, Guo Y, Yan M, Xiao Y, Dong Y, Zhang R, Qin Y, Bu Y, Zhang Y, Gao H. Phosphorylation of RasGRP1 by Shc3 prevents RasGRP1 degradation and contributes to Ras/c-Jun activation in hepatocellular carcinoma. Mol Cell Biochem 2024; 479:2307-2321. [PMID: 37646951 DOI: 10.1007/s11010-023-04839-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
Ras guanine nucleotide-releasing protein 1 (RasGRP1), a Ras activator, is upregulated in hepatocellular carcinoma (HCC) and other kinds of cancer and is associated with the poor prognosis of patients. However, little is known about the underlying regulatory mechanisms of RasGRP1 in the context of cancer. Here, we report that RasGRP1 physically interacted with the adaptor protein Src homolog and collagen homolog 3 (Shc3). Moreover, RasGRP1 C-terminus domain (aa 607-797) bound to the central collagen-homology 1 (CH1) domain of Shc3. Subsequently, Shc3 enhanced the RasGRP1 tyrosine phosphorylation rate and stability by inhibiting its ubiquitination. Notably, the phosphorylation-mimicking mutants of RasGRP1, RasGRP1 Y704A, and Y748A, rescued the phosphorylation and ubiquitination levels of RasGRP1 in HCC cells. Further investigation showed that the RasGRP1 and Shc3 interaction induced activation of Ras and c-Jun, resulting in cell proliferation in vitro. Moreover, the regulation of Shc3/RasGRP1/Ras/c-Jun signal transduction was confirmed in vivo using the subcutaneous xenograft mouse model. Thus, we propose that continuous Shc3 overexpression may be a possible mechanism for maintaining RasGRP1 stability and that persistent activation of Ras/c-Jun signaling through the interaction of RasGRP1 and Shc3 is a key event increasing cell proliferation. Our findings suggest that the interaction of RasGRP1 and Shc3 plays an important role in HCC tumorigenesis and suggests the potential clinical usage of novel biomarkers and therapeutic targets in HCC.
Collapse
Affiliation(s)
- Xinran Zhang
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yun Liu
- Key Laboratory of Cancer Prevention and Therapy, Department of Pediatric Oncology, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Rui Yang
- Department of Critical Care Medicine, Tianjin First Central Hospital, Tianjin Institute of Emergency Medicine, Tianjin, 300192, China
| | - Yuanyuan Guo
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Meiling Yan
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Ying Xiao
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yunzhuo Dong
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Ruixia Zhang
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yinpeng Qin
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yishan Bu
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yi Zhang
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Huier Gao
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China.
| |
Collapse
|
2
|
Zhou Y, Sun G, Li C, Bai X. Grb2 Expression in Acute Spinal Cord Injury After Methylprednisolone Intrathecal Injection in Rats. Int J Spine Surg 2023; 17:678-683. [PMID: 37884335 PMCID: PMC10623675 DOI: 10.14444/8527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023] Open
Abstract
OBJECTIVE This study aims to investigate the effect of methylprednisolone (MP) intrathecal injection on a rat model of acute spinal cord injury (ASCI). METHODS Allen's frame was used to establish a rat model of ASCI. MP and normal saline were intrathecally injected to Sprague-Dawley rats at 0, 3, 6, 8, 12, and 24 hours after ASCI, and injured spinal cord tissues were sterilely extracted after 24 hours of treatment. Isobaric tags for relative and absolute quantitation (iTRAQ) were coupled with 2-dimensional liquid chromatography tandem mass spectrometry to separate and identify differentially expressed proteins. RESULTS The expression of growth factor receptor-bound protein 2 (Grb2) was downregulated in the MP groups at 0 hours (iTRAQ ratio = 0.996), 3 hours (iTRAQ ratio = 0.737), 8 hours (iTRAQ ratio = 0.763), and 24 hours (iTRAQ ratio = 0.908) after injury compared with that in the control groups. No significant difference in Grb2 expression was observed between the control groups at 6 and 12 hours after ASCI. CONCLUSIONS Standardized MP intrathecal injection after ASCI treatment reduces Grb2 activation in a rat ASCI model. Further studies should determine whether or not the same effect can be observed in human ASCIs.
Collapse
Affiliation(s)
- Yijun Zhou
- The First People's Hospital of Changde, Changde Hospital Affiliated to Xiangya Medical College of Central South University, Hunan, People's Republic of China
| | - Guanwen Sun
- The First People's Hospital of Changde, Changde Hospital Affiliated to Xiangya Medical College of Central South University, Hunan, People's Republic of China
| | - Changhong Li
- The First People's Hospital of Changde, Changde Hospital Affiliated to Xiangya Medical College of Central South University, Hunan, People's Republic of China
| | - Xiaoan Bai
- The First People's Hospital of Changde, Changde Hospital Affiliated to Xiangya Medical College of Central South University, Hunan, People's Republic of China
| |
Collapse
|
3
|
Martins RS, Jesus TT, Cardoso L, Soares P, Vinagre J. Personalized Medicine in Medullary Thyroid Carcinoma: A Broad Review of Emerging Treatments. J Pers Med 2023; 13:1132. [PMID: 37511745 PMCID: PMC10381735 DOI: 10.3390/jpm13071132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Medullary thyroid carcinoma (MTC) arises from parafollicular cells in the thyroid gland, and although rare, it represents an aggressive type of thyroid cancer. MTC is recognized for its low mutational burden, with point mutations in RET or RAS genes being the most common oncogenic events. MTC can be resistant to cytotoxic chemotherapy, and multitarget kinase inhibitors (MKIs) have been considered a treatment option. They act by inhibiting the activities of specific tyrosine kinase receptors involved in tumor growth and angiogenesis. Several tyrosine kinase inhibitors are approved in the treatment of advanced MTC, including vandetanib and cabozantinib. However, due to the significant number of adverse events, debatable efficiency and resistance, there is a need for novel RET-specific TKIs. Newer RET-specific TKIs are expected to overcome previous limitations and improve patient outcomes. Herein, we aim to review MTC signaling pathways, the most recent options for treatment and the applications for personalized medicine.
Collapse
Affiliation(s)
- Rui Sousa Martins
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Faculdade de Ciências da Universidade do Porto (FCUP), 4169-007 Porto, Portugal
| | - Tito Teles Jesus
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
| | - Luís Cardoso
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Departamento de Endocrinologia, Diabetes e Metabolismo do Centro Hospitalar Universitário de Coimbra, 3000-075 Coimbra, Portugal
| | - Paula Soares
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| | - João Vinagre
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), 4200-135 Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| |
Collapse
|
4
|
Liu Y, Konopka G. An integrative understanding of comparative cognition: lessons from human brain evolution. Integr Comp Biol 2020; 60:991-1006. [PMID: 32681799 PMCID: PMC7608741 DOI: 10.1093/icb/icaa109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A comprehensive understanding of animal cognition requires the integration of studies on behavior, electrophysiology, neuroanatomy, development, and genomics. Although studies of comparative cognition are receiving increasing attention from organismal biologists, most current studies focus on the comparison of behaviors and anatomical structures to understand their adaptative values. However, to understand the most potentially complex cognitive program of the human brain a greater synthesis of a multitude of disciplines is needed. In this review, we start with extensive neuroanatomic comparisons between humans and other primates. One likely specialization of the human brain is the expansion of neocortex, especially in regions for high-order cognition (e.g., prefrontal cortex). We then discuss how such an expansion can be linked to heterochrony of the brain developmental program, resulting in a greater number of neurons and enhanced computational capacity. Furthermore, alteration of gene expression in the human brain has been associated with positive selection in DNA sequences of gene regulatory regions. These results not only imply that genes associated with brain development are a major factor in the evolution of cognition, but also that high-quality whole-genome sequencing and gene manipulation techniques are needed for an integrative and functional understanding of comparative cognition in non-model organisms.
Collapse
Affiliation(s)
- Yuxiang Liu
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Genevieve Konopka
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
5
|
Criscuoli M, Ulivieri C, Filippi I, Monaci S, Guerrini G, Crifò B, De Tommaso D, Pelicci G, Baldari CT, Taylor CT, Carraro F, Naldini A. The Shc protein Rai enhances T-cell survival under hypoxia. J Cell Physiol 2020; 235:8058-8070. [PMID: 31944299 DOI: 10.1002/jcp.29461] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/07/2020] [Indexed: 12/18/2022]
Abstract
Hypoxia occurs in physiological and pathological conditions. T cells experience hypoxia in pathological and physiological conditions as well as in lymphoid organs. Indeed, hypoxia-inducible factor 1α (HIF-1α) affects T cell survival and functions. Rai, an Shc family protein member, exerts pro-survival effects in hypoxic neuroblastoma cells. Since Rai is also expressed in T cells, we here investigated its role in hypoxic T cells. In this work, hypoxia differently affected cell survival, proapoptotic, and metabolic programs in T cells, depending upon Rai expression. By using Jurkat cells stably expressing Rai and splenocytes from Rai-/- mice, we demonstrated that Rai promotes T cell survival and affects cell metabolism under hypoxia. Upon exposure to hypoxia, Jurkat T cells expressing Rai show (a) higher HIF-1α protein levels; (b) a decreased cell death and increased Akt/extracellular-signal-regulated kinase phosphorylation; (c) a decreased expression of proapoptotic markers, including caspase activities and poly(ADP-ribose) polymerase cleavage; (d) an increased glucose and lactate metabolism; (e) an increased activation of nuclear factor-kB pathway. The opposite effects were observed in hypoxic splenocytes from Rai-/- mice. Thus, Rai plays an important role in hypoxic signaling and may be relevant in the protection of T cells against hypoxia.
Collapse
Affiliation(s)
- Mattia Criscuoli
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Irene Filippi
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.,Istituto Toscano Tumori, Firenze, Italy
| | - Sara Monaci
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuditta Guerrini
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Bianca Crifò
- Department of Systems Biology, UCD Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | | | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.,Department of Translational Medicine, Piemonte Orientale University "Amedeo Avogadro", Novara, Italy
| | | | - Cormac T Taylor
- Department of Systems Biology, UCD Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Fabio Carraro
- Istituto Toscano Tumori, Firenze, Italy.,Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Antonella Naldini
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
6
|
Ulivieri C, De Tommaso D, Finetti F, Ortensi B, Pelicci G, D'Elios MM, Ballerini C, Baldari CT. A T Cell Suppressive Circuitry Mediated by CD39 and Regulated by ShcC/Rai Is Induced in Astrocytes by Encephalitogenic T Cells. Front Immunol 2019; 10:1041. [PMID: 31134091 PMCID: PMC6524536 DOI: 10.3389/fimmu.2019.01041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis is an autoimmune disease caused by autoreactive immune cell infiltration into the central nervous system leading to inflammation, demyelination, and neuronal loss. While myelin-reactive Th1 and Th17 are centrally implicated in multiple sclerosis pathogenesis, the local CNS microenvironment, which is shaped by both infiltrated immune cells and central nervous system resident cells, has emerged a key player in disease onset and progression. We have recently demonstrated that ShcC/Rai is as a novel astrocytic adaptor whose loss in mice protects from experimental autoimmune encephalomyelitis. Here, we have explored the mechanisms that underlie the ability of Rai-/- astrocytes to antagonize T cell-dependent neuroinflammation. We show that Rai deficiency enhances the ability of astrocytes to upregulate the expression and activity of the ectonucleotidase CD39, which catalyzes the conversion of extracellular ATP to the immunosuppressive metabolite adenosine, through both contact-dependent and-independent mechanisms. As a result, Rai-deficient astrocytes acquire an enhanced ability to suppress T-cell proliferation, which involves suppression of T cell receptor signaling and upregulation of the inhibitory receptor CTLA-4. Additionally, Rai-deficient astrocytes preferentially polarize to the neuroprotective A2 phenotype. These results identify a new mechanism, to which Rai contributes to a major extent, by which astrocytes modulate the pathogenic potential of autoreactive T cells.
Collapse
Affiliation(s)
| | | | | | - Barbara Ortensi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.,Department of Translational Medicine, Piemonte Orientale University "Amedeo Avogadro", Novara, Italy
| | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.,Department of Translational Medicine, Piemonte Orientale University "Amedeo Avogadro", Novara, Italy
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Clara Ballerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | |
Collapse
|
7
|
Shi G, Chen L, Chen G, Zou C, Li J, Li M, Fang C, Li C. Identification and Functional Prediction of Long Intergenic Non-coding RNAs Related to Subcutaneous Adipose Development in Pigs. Front Genet 2019; 10:160. [PMID: 30886630 PMCID: PMC6409335 DOI: 10.3389/fgene.2019.00160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/14/2019] [Indexed: 12/19/2022] Open
Abstract
An increasing number of studies have shown that long intergenic non-coding RNAs (lincRNAs) are a very important class of non-coding RNAs that plays a vital role in many biological processes. Adipose tissue is an important place for storing energy, but few studies on lincRNAs were related to pig subcutaneous fat development. Here, we used published RNA-seq data from subcutaneous adipose tissue of Italian Large White pigs and identified 252 putative lincRNAs, wherein 34 were unannotated. These lincRNAs had relatively shorter length, lower number of exons, and lower expression level compared with protein-coding transcripts. Gene ontology and pathway analysis indicated that the adjacent genes of lincRNAs were involved in lipid metabolism. In addition, differentially expressed lincRNAs (DELs) between low and high backfat thickness pigs were identified. Through the detection of quantitative trait locus (QTL), DELs were mainly located in QTLs related to adipose development. Based on the expression correlation of DEL genes and their differentially expressed potential target genes, we constructed a co-expression network and a potential pathway of DEL's effect on lipid metabolism. Our study identified and analyzed lincRNAs in subcutaneous adipose tissue, and results suggested that lincRNAs may be involved in the regulation of subcutaneous fat development. Our findings provided new insights into the biological function of porcine lincRNAs.
Collapse
Affiliation(s)
- Gaoli Shi
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Lin Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Guoting Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Cheng Zou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Jingxuan Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Mengxun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Chengchi Fang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Changchun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
8
|
Weissmann S, Cloos PA, Sidoli S, Jensen ON, Pollard S, Helin K. The Tumor Suppressor CIC Directly Regulates MAPK Pathway Genes via Histone Deacetylation. Cancer Res 2018; 78:4114-4125. [PMID: 29844126 PMCID: PMC6076439 DOI: 10.1158/0008-5472.can-18-0342] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/25/2018] [Accepted: 05/14/2018] [Indexed: 12/18/2022]
Abstract
Oligodendrogliomas are brain tumors accounting for approximately 10% of all central nervous system cancers. CIC is a transcription factor that is mutated in most patients with oligodendrogliomas; these mutations are believed to be a key oncogenic event in such cancers. Analysis of the Drosophila melanogaster ortholog of CIC, Capicua, indicates that CIC loss phenocopies activation of the EGFR/RAS/MAPK pathway, and studies in mammalian cells have demonstrated a role for CIC in repressing the transcription of the PEA3 subfamily of ETS transcription factors. Here, we address the mechanism by which CIC represses transcription and assess the functional consequences of CIC inactivation. Genome-wide binding patterns of CIC in several cell types revealed that CIC target genes were enriched for MAPK effector genes involved in cell-cycle regulation and proliferation. CIC binding to target genes was abolished by high MAPK activity, which led to their transcriptional activation. CIC interacted with the SIN3 deacetylation complex and, based on our results, we suggest that CIC functions as a transcriptional repressor through the recruitment of histone deacetylases. Independent single amino acid substitutions found in oligodendrogliomas prevented CIC from binding its target genes. Taken together, our results show that CIC is a transcriptional repressor of genes regulated by MAPK signaling, and that ablation of CIC function leads to increased histone acetylation levels and transcription at these genes, ultimately fueling mitogen-independent tumor growth.Significance: Inactivation of CIC inhibits its direct repression of MAPK pathway genes, leading to their increased expression and mitogen-independent growth.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/15/4114/F1.large.jpg Cancer Res; 78(15); 4114-25. ©2018 AACR.
Collapse
Affiliation(s)
- Simon Weissmann
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Paul A Cloos
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
- Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Simone Sidoli
- Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
- Department of Biochemistry and Molecular Biology, VILLUM Centre for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Ole N Jensen
- Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
- Department of Biochemistry and Molecular Biology, VILLUM Centre for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Steven Pollard
- MRC Centre for Regenerative Medicine and Edinburgh Cancer Research UK Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - Kristian Helin
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
- Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), University of Copenhagen, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Tsutsui Y, Hays FA. A Link Between Alzheimer's and Type II Diabetes Mellitus? Ca +2 -Mediated Signal Control and Protein Localization. Bioessays 2018; 40:e1700219. [PMID: 29694668 PMCID: PMC6166406 DOI: 10.1002/bies.201700219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/16/2018] [Indexed: 01/28/2023]
Abstract
We propose protein localization dependent signal activation (PLDSA) as a model to describe pre-existing protein partitioning between the cytosol, and membrane surface, as a means to modulate signal activation, specificity, and robustness. We apply PLDSA to explain possible molecular links between type II diabetes mellitus (T2DM) and Alzheimer's disease (AD) by describing Ca+2 -mediated interactions between the Src non-receptor tyrosine kinase and p52Shc adaptor protein. We suggest that these interactions may serve as a contributing factor to disease development and progression. In particular, we propose that signaling response is regulated, in part, by Ca+2 -mediated partitioning of lipid-bound and soluble forms of Src and p52shc. Thus, protein-protein interactions that drive signaling in response to extracellular ligand binding are also mediated by partitioning of signaling proteins between membrane-bound and soluble populations. We propose that PLDSA effects may explain, in part, the evolutionary basis of promiscuous protein interaction domains and their importance in cellular function.
Collapse
Affiliation(s)
- Yuko Tsutsui
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Franklin A. Hays
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, United States
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, United States
| |
Collapse
|
10
|
Mabruk ZA, Ahmed SBM, Thomas AC, Prigent SA. The role of the ShcD and RET interaction in neuroblastoma survival and migration. Biochem Biophys Rep 2018; 13:99-108. [PMID: 29556564 PMCID: PMC5857170 DOI: 10.1016/j.bbrep.2018.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 01/02/2018] [Accepted: 01/11/2018] [Indexed: 01/15/2023] Open
Abstract
Preliminary screening data showed that the ShcD adaptor protein associates with the proto-oncogene RET receptor tyrosine kinase. In the present study, we aimed to investigate the molecular interaction between ShcD and RET in human neuroblastoma cells and study the functional impact of this interaction. We were able to show that ShcD immunoprecipitated with RET from SK-N-AS neuroblastoma cell lysates upon GDNF treatment. This result was validated by ShcD-RET co-localization, which was visualized using a fluorescence microscope. ShcD-RET coexpression promoted ShcD and RET endosomal localization, resulting in unexpected inhibition of the downstream ERK and AKT pathways. Interestingly, ShcD-RET association reduced the viability and migration of SK-N-AS cells. Although ShcD was previously shown to trigger melanoma cell migration and tumorigenesis, our data showed an opposite role for ShcD in neuroblastoma SK-N-AS cells via its association with RET in GDNF-treated cells. In conclusion, ShcD acts as a switch molecule that promotes contrasting biological responses depending on the stimulus ad cell type. The melanoma associated Shc adaptor, ShcD, is found to interact with Ret oncogene receptor in SK-N-AS neuroblastoma cells. ShcD and Ret coexpression favoures their endosomal localization. ShcD-Ret association has suppressed ERK and AKT signalling. The functional consequence of ShcD and Ret interaction was shown to negatively affect cell survival and cellular migration in.
Collapse
Key Words
- ALK,, Anaplastic Lymphoma Kinase
- Akt,, Protein kinase B;
- CMV,, Cytomegalovirus
- DMEM,, Dulbecoo Modified Eagle's Medium;
- DNA,, Deoxyribonucleic Acid
- ECL,, Enhanced Chemiluminescence;
- EGF,, Epidermal Growth Factor;
- EGFR,, Epidermal Growth Factor Receptor;
- ERK,, Extracellular Signal–Regulated Kinases;
- Endosomes
- FBS,, Fetal Bovine Serum
- FGFR,, fibroblast growth factor receptors
- GDNF
- GDNF,, Glial Cell Line-Derived Neurotropic Factor;
- GFLs,, GDNF Family Ligands;
- GFP,, Green Fluorescent Protein
- GPCR,, G-Protein Coupled Receptor
- GRB2,, Growth Factor Receptor-Bound Protein 2;
- HGFR,, hepatocyte growth factor receptor;
- HRP,, Horseradish Peroxidase
- IGF,, Insulin Growth Factor;
- LB,, Luria-Bertani
- MAP,, Mitogen-Activated Protein;
- MAPK,, Mitogen-Activated Protein Kinases
- MuSK,, Muscle Specific Kinase
- NFDM,, Non-Fat Dry Milk
- Neuroblastoma
- PBS,, Phosphate-Buffered Saline
- PBST,, Phosphate-Buffered Saline Tween
- PDGF,, Platelet-Derived Growth Factor;
- PI3K,, Phosphoinositide 3-Kinase
- PMSF,, Phenylmethylsulfonyl Fluoride
- PVDF,, Polyvinylidene Fluoride
- RET
- RET,, Rearranged During Transfection
- RT,, Room Temperature;
- RTKs,, Receptor Tyrosine Kinase
- SDS-PAGE,, Sodium Dodecylsulphate Polyacrylamide Gel Electrophoresis
- ShcD
- ShcD,, Src Homology And Collagen D
- Src,, Proto-Oncogene Tyrosine-Protein Kinase Src
- TKRs,, Tyrosine Kinase Receptor;
- TrkA/B/C,, Tropomyosin-Related Kinase Receptor A/B/C
- hrs,, Hours
- mAb,, Monoclonal Antibody
- min,, Minute
- pAb,, Polyclonal Antibody
- pTyr,, Phospho-Tyrosine
- rpm,, revolution per minute;
Collapse
Affiliation(s)
- Zeanap A Mabruk
- Sharjah Institute for Medical Research and College of Medicine University of Sharjah, United Arab Emirates
| | - Samrein B M Ahmed
- Sharjah Institute for Medical Research and College of Medicine University of Sharjah, United Arab Emirates
| | - Asha Caroline Thomas
- Sharjah Institute for Medical Research and College of Medicine University of Sharjah, United Arab Emirates
| | - Sally A Prigent
- Department of Molecular and Cellular Biology, University of Leicester, UK
| |
Collapse
|
11
|
Triaca V, Coccurello R, Giacovazzo G. The neuronal Shc adaptor in Alzheimer's Disease. Aging (Albany NY) 2018; 10:5-6. [PMID: 29362290 PMCID: PMC5811251 DOI: 10.18632/aging.101368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 01/21/2018] [Indexed: 01/17/2023]
Affiliation(s)
- Viviana Triaca
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Rome, Italy
| | - Roberto Coccurello
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Rome, Italy.,European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, Rome, Italy
| | - Giacomo Giacovazzo
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Rome, Italy.,European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, Rome, Italy
| |
Collapse
|
12
|
Liu Y, Zhang X, Yang B, Zhuang H, Guo H, Wei W, Li Y, Chen R, Li Y, Zhang N. Demethylation-Induced Overexpression of Shc3 Drives c-Raf-Independent Activation of MEK/ERK in HCC. Cancer Res 2018; 78:2219-2232. [PMID: 29330146 DOI: 10.1158/0008-5472.can-17-2432] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 12/14/2017] [Accepted: 01/08/2018] [Indexed: 11/16/2022]
Abstract
Invasion and intrahepatic metastasis are major factors of poor prognosis in patients with hepatocellular carcinoma (HCC). In this study, we show that increased Src homolog and collagen homolog 3 (Shc3) expression in malignant HCC cell lines associate with HCC invasion and metastasis. Shc3 (N-Shc) was significantly upregulated in tumors of 33 HCC patient samples as compared with adjacent normal tissues. Further analysis of 52 HCC patient samples showed that Shc3 expression correlated with microvascular invasion, cancer staging, and poor prognosis. Shc3 interacted with major vault protein, resulting in activation of MEK1/2 and ERK1/2 independently of Shc1 and c-Raf; this interaction consequently induced epithelial-mesenchymal transition and promoted HCC cell proliferation and metastasis. The observed increase in Shc3 levels was due to demethylation of its upstream promoter, which allowed c-Jun binding. In turn, Shc3 expression promoted c-Jun phosphorylation in a positive feedback loop. Analysis of metastasis using a tumor xenograft mouse model further confirmed the role of Shc3 in vivo Taken together, our results indicate the importance of Shc3 in HCC progression and identify Shc3 as a novel biomarker and potential therapeutic target in HCC.Significance: Ectopic expression of Shc3 forms a complex with MVP/MEK/ERK to potentiate ERK activation and plays an important role in sorafinib resistance in HCC. Cancer Res; 78(9); 2219-32. ©2018 AACR.
Collapse
Affiliation(s)
- Yun Liu
- Key Laboratory of Breast Cancer Prevention and Therapy, Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Xinran Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy, Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Baicai Yang
- Key Laboratory of Breast Cancer Prevention and Therapy, Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Hao Zhuang
- Key Laboratory of Breast Cancer Prevention and Therapy, Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.,Department of Hepatic Biliary Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hua Guo
- Key Laboratory of Breast Cancer Prevention and Therapy, Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Wen Wei
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yuan Li
- Department of Laboratory Animal Sciences, Tianjin Medical University, Tianjin, China
| | - Ruibing Chen
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yongmei Li
- Department of Pathogen Biology, Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Ning Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy, Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
13
|
Criscuoli M, Filippi I, Osti D, Aldinucci C, Guerrini G, Pelicci G, Carraro F, Naldini A. The Shc protein RAI promotes an adaptive cell survival program in hypoxic neuroblastoma cells. J Cell Physiol 2017; 233:4282-4293. [DOI: 10.1002/jcp.26247] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 10/13/2017] [Indexed: 01/25/2023]
Affiliation(s)
- Mattia Criscuoli
- Cellular and Molecular Physiology UnitDepartment of Molecular and Developmental MedicineUniversity of SienaSienaItaly
| | - Irene Filippi
- Cellular and Molecular Physiology UnitDepartment of Molecular and Developmental MedicineUniversity of SienaSienaItaly
- Istituto Toscano TumoriFirenzeItaly
| | - Daniela Osti
- Department of Experimental OncologyEuropean Institute of OncologyMilanItaly
| | - Carlo Aldinucci
- Cellular and Molecular Physiology UnitDepartment of Molecular and Developmental MedicineUniversity of SienaSienaItaly
| | - Giuditta Guerrini
- Cellular and Molecular Physiology UnitDepartment of Molecular and Developmental MedicineUniversity of SienaSienaItaly
| | - Giuliana Pelicci
- Department of Experimental OncologyEuropean Institute of OncologyMilanItaly
- Department of Translational MedicinePiemonte Orientale University “Amedeo Avogadro”NovaraItaly
| | - Fabio Carraro
- Cellular and Molecular Physiology UnitDepartment of Molecular and Developmental MedicineUniversity of SienaSienaItaly
- Istituto Toscano TumoriFirenzeItaly
| | - Antonella Naldini
- Cellular and Molecular Physiology UnitDepartment of Molecular and Developmental MedicineUniversity of SienaSienaItaly
| |
Collapse
|
14
|
Kumasaka MY, Yajima I, Ohgami N, Ninomiya H, Iida M, Li X, Oshino R, Tanihata H, Yoshinaga M, Kato M. Manganese-Mediated Decrease in Levels of c-RET and Tyrosine Hydroxylase Expression In Vitro. Neurotox Res 2017; 32:661-670. [PMID: 28730349 DOI: 10.1007/s12640-017-9783-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 06/29/2017] [Accepted: 07/05/2017] [Indexed: 10/19/2022]
Abstract
Previous studies showed that overexposure to manganese causes parkinsonism, a disorder of dopaminergic neurons. Previous studies also showed that activity of c-RET kinase controls dopamine production through regulation of tyrosine hydroxylase (TH) expression, suggesting the involvement of c-RET in the development of parkinsonism. To our knowledge, however, there is no report showing a correlation between manganese-mediated parkinsonism and c-RET. In this study, we examined the effect of manganese on the expression and/or activation levels of c-RET and TH in human TH-expressing cells (TGW cells). We first found that treatment with 30 and 100 μM manganese resulted in reduction of c-RET transcript level and degradation of c-RET protein through promotion of ubiquitination. We then examined the biological significance of manganese-mediated decrease of c-RET protein expression. Decreased TH expression with decreased c-RET kinase activity was observed in c-RET protein-depleted TGW cells by treatment with manganese (30 μM) as well as by c-RET siRNA transfection. Since TH protein has been shown to be involved in the dopamine-producing pathway in previous studies, our results indicate the possibility that manganese-mediated reduction of TH expression and phosphorylation via decreased expression of c-RET protein in neural cells is involved in parkinsonism induced by manganese.
Collapse
Affiliation(s)
- Mayuko Y Kumasaka
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.,Unit of Environmental Health Sciences, Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai-shi, Aichi, 487-8501, Japan.,Voluntary Body for International Health Care in Universities, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Ichiro Yajima
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.,Unit of Environmental Health Sciences, Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai-shi, Aichi, 487-8501, Japan.,Voluntary Body for International Health Care in Universities, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Nobutaka Ohgami
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.,Unit of Environmental Health Sciences, Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai-shi, Aichi, 487-8501, Japan.,Voluntary Body for International Health Care in Universities, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Hiromasa Ninomiya
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.,Voluntary Body for International Health Care in Universities, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Machiko Iida
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.,Unit of Environmental Health Sciences, Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai-shi, Aichi, 487-8501, Japan.,Voluntary Body for International Health Care in Universities, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Xiang Li
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Reina Oshino
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.,Voluntary Body for International Health Care in Universities, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Hiroko Tanihata
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Masafumi Yoshinaga
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.,Voluntary Body for International Health Care in Universities, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Masashi Kato
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan. .,Unit of Environmental Health Sciences, Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai-shi, Aichi, 487-8501, Japan. .,Voluntary Body for International Health Care in Universities, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
15
|
Canu N, Amadoro G, Triaca V, Latina V, Sposato V, Corsetti V, Severini C, Ciotti MT, Calissano P. The Intersection of NGF/TrkA Signaling and Amyloid Precursor Protein Processing in Alzheimer's Disease Neuropathology. Int J Mol Sci 2017. [PMID: 28632177 PMCID: PMC5486140 DOI: 10.3390/ijms18061319] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Dysfunction of nerve growth factor (NGF) and its high-affinity Tropomyosin receptor kinase A (TrkA) receptor has been suggested to contribute to the selective degeneration of basal forebrain cholinergic neurons (BFCN) associated with the progressive cognitive decline in Alzheimer's disease (AD). The aim of this review is to describe our progress in elucidating the molecular mechanisms underlying the dynamic interplay between NGF/TrkA signaling and amyloid precursor protein (APP) metabolism within the context of AD neuropathology. This is mainly based on the finding that TrkA receptor binding to APP depends on a minimal stretch of ~20 amino acids located in the juxtamembrane/extracellular domain of APP that carries the α- and β-secretase cleavage sites. Here, we provide evidence that: (i) NGF could be one of the “routing” proteins responsible for modulating the metabolism of APP from amyloidogenic towards non-amyloidogenic processing via binding to the TrkA receptor; (ii) the loss of NGF/TrkA signaling could be linked to sporadic AD contributing to the classical hallmarks of the neuropathology, such as synaptic loss, β-amyloid peptide (Aβ) deposition and tau abnormalities. These findings will hopefully help to design therapeutic strategies for AD treatment aimed at preserving cholinergic function and anti-amyloidogenic activity of the physiological NGF/TrkA pathway in the septo-hippocampal system.
Collapse
Affiliation(s)
- Nadia Canu
- Department of System Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00137 Rome, Italy.
- Institute of Cellular Biology and Neurobiology, National Council of Research Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Giuseppina Amadoro
- Institute of Translational Pharmacology, National Research Council (CNR) Rome, Via Fosso del Cavaliere 100, 00133 Rome, Italy.
| | - Viviana Triaca
- Institute of Cellular Biology and Neurobiology, National Council of Research Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Valentina Latina
- Institute of Translational Pharmacology, National Research Council (CNR) Rome, Via Fosso del Cavaliere 100, 00133 Rome, Italy.
| | - Valentina Sposato
- European Brain Research Institute Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Veronica Corsetti
- European Brain Research Institute Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Cinzia Severini
- Institute of Cellular Biology and Neurobiology, National Council of Research Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Maria Teresa Ciotti
- Institute of Cellular Biology and Neurobiology, National Council of Research Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| | - Pietro Calissano
- European Brain Research Institute Rome, Via del Fosso del Fiorano 64, 00143 Rome, Italy.
| |
Collapse
|
16
|
Abstract
The Shc family of adaptor proteins is a group of proteins that lacks intrinsic enzymatic activity. Instead, Shc proteins possess various domains that allow them to recruit different signalling molecules. Shc proteins help to transduce an extracellular signal into an intracellular signal, which is then translated into a biological response. The Shc family of adaptor proteins share the same structural topography, CH2-PTB-CH1-SH2, which is more than an isoform of Shc family proteins; this structure, which includes multiple domains, allows for the posttranslational modification of Shc proteins and increases the functional diversity of Shc proteins. The deregulation of Shc proteins has been linked to different disease conditions, including cancer and Alzheimer’s, which indicates their key roles in cellular functions. Accordingly, a question might arise as to whether Shc proteins could be targeted therapeutically to correct their disturbance. To answer this question, thorough knowledge must be acquired; herein, we aim to shed light on the Shc family of adaptor proteins to understand their intracellular role in normal and disease states, which later might be applied to connote mechanisms to reverse the disease state.
Collapse
|
17
|
Wills MKB, Keyvani Chahi A, Lau HR, Tilak M, Guild BD, New LA, Lu P, Jacquet K, Meakin SO, Bisson N, Jones N. Signaling adaptor ShcD suppresses extracellular signal-regulated kinase (Erk) phosphorylation distal to the Ret and Trk neurotrophic receptors. J Biol Chem 2017; 292:5748-5759. [PMID: 28213521 DOI: 10.1074/jbc.m116.770511] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/06/2017] [Indexed: 11/06/2022] Open
Abstract
Proteins of the Src homology and collagen (Shc) family are typically involved in signal transduction events involving Ras/MAPK and PI3K/Akt pathways. In the nervous system, they function proximal to the neurotrophic factors that regulate cell survival, differentiation, and neuron-specific characteristics. The least characterized homolog, ShcD, is robustly expressed in the developing and mature nervous system, but its contributions to neural cell circuitry are largely uncharted. We now report that ShcD binds to active Ret, TrkA, and TrkB neurotrophic factor receptors predominantly via its phosphotyrosine-binding (PTB) domain. However, in contrast to the conventional Shc adaptors, ShcD suppresses distal phosphorylation of the Erk MAPK. Accordingly, genetic knock-out of mouse ShcD enhances Erk phosphorylation in the brain. In cultured cells, this capacity is tightly aligned to phosphorylation of ShcD CH1 region tyrosine motifs, which serve as docking platforms for signal transducers, such as Grb2. Erk suppression is relieved through independent mutagenesis of the PTB domain and the CH1 tyrosine residues, and successive substitution of these tyrosines breaks the interaction between ShcD and Grb2, thereby promoting TrkB-Grb2 association. Erk phosphorylation can also be restored in the presence of wild type ShcD through Grb2 overexpression. Conversely, mutation of the ShcD SH2 domain results in enhanced repression of Erk. Although the SH2 domain is a less common binding interface in Shc proteins, we demonstrate that it associates with the Ptpn11 (Shp2) phosphatase, which in turn regulates ShcD tyrosine phosphorylation. We therefore propose a model whereby ShcD competes with neurotrophic receptors for Grb2 binding and opposes activation of the MAPK cascade.
Collapse
Affiliation(s)
- Melanie K B Wills
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Ava Keyvani Chahi
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Hayley R Lau
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Manali Tilak
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Brianna D Guild
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Laura A New
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Peihua Lu
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Kévin Jacquet
- Cancer Research Centre, Quebec Network for Research on Protein Function, Engineering, and Applications (PROTEO) and Centre Hospitalier Universitaire de Québec Research Centre-Université Laval, Québec City, Québec G1R 2J6, Canada, and
| | - Susan O Meakin
- Department of Biochemistry, Western University, London, Ontario N6A 5B7, Canada
| | - Nicolas Bisson
- Cancer Research Centre, Quebec Network for Research on Protein Function, Engineering, and Applications (PROTEO) and Centre Hospitalier Universitaire de Québec Research Centre-Université Laval, Québec City, Québec G1R 2J6, Canada, and
| | - Nina Jones
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada,
| |
Collapse
|
18
|
Triaca V, Sposato V, Bolasco G, Ciotti MT, Pelicci P, Bruni AC, Cupidi C, Maletta R, Feligioni M, Nisticò R, Canu N, Calissano P. NGF controls APP cleavage by downregulating APP phosphorylation at Thr668: relevance for Alzheimer's disease. Aging Cell 2016; 15:661-72. [PMID: 27076121 PMCID: PMC4933663 DOI: 10.1111/acel.12473] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/29/2016] [Indexed: 12/17/2022] Open
Abstract
NGF has been implicated in forebrain neuroprotection from amyloidogenesis and Alzheimer's disease (AD). However, the underlying molecular mechanisms are still poorly understood. Here, we investigated the role of NGF signalling in the metabolism of amyloid precursor protein (APP) in forebrain neurons using primary cultures of septal neurons and acute septo-hippocampal brain slices. In this study, we show that NGF controls the basal level of APP phosphorylation at Thr668 (T668) by downregulating the activity of the Ser/Thr kinase JNK(p54) through the Tyr kinase signalling adaptor SH2-containing sequence C (ShcC). We also found that the specific NGF receptor, Tyr kinase A (TrkA), which is known to bind to APP, fails to interact with the fraction of APP molecules phosphorylated at T668 (APP(pT668) ). Accordingly, the amount of TrkA bound to APP is significantly reduced in the hippocampus of ShcC KO mice and of patients with AD in which elevated APP(pT668) levels are detected. NGF promotes TrkA binding to APP and APP trafficking to the Golgi, where APP-BACE interaction is hindered, finally resulting in reduced generation of sAPPβ, CTFβ and amyloid-beta (1-42). These results demonstrate that NGF signalling directly controls basal APP phosphorylation, subcellular localization and BACE cleavage, and pave the way for novel approaches specifically targeting ShcC signalling and/or the APP-TrkA interaction in AD therapy.
Collapse
Affiliation(s)
- Viviana Triaca
- Institute of Cell Biology and Neuroscience National Research Council (CNR) Rome Italy
- European Brain Research Institute (EBRI Foundation) Rome Italy
| | - Valentina Sposato
- Institute of Cell Biology and Neuroscience National Research Council (CNR) Rome Italy
- European Brain Research Institute (EBRI Foundation) Rome Italy
| | - Giulia Bolasco
- European Molecular Biology Laboratory (EMBL) Monterotondo Italy
| | - Maria Teresa Ciotti
- Institute of Cell Biology and Neuroscience National Research Council (CNR) Rome Italy
| | | | - Amalia C. Bruni
- Regional Neurogenetic Center (CRN) ASP Catanzaro Lamezia Terme Italy
| | - Chiara Cupidi
- Regional Neurogenetic Center (CRN) ASP Catanzaro Lamezia Terme Italy
| | - Raffaele Maletta
- Regional Neurogenetic Center (CRN) ASP Catanzaro Lamezia Terme Italy
| | - Marco Feligioni
- European Brain Research Institute (EBRI Foundation) Rome Italy
| | - Robert Nisticò
- European Brain Research Institute (EBRI Foundation) Rome Italy
| | - Nadia Canu
- Institute of Cell Biology and Neuroscience National Research Council (CNR) Rome Italy
- Department of System Medicine University of Rome “Tor Vergata” Rome Italy
| | | |
Collapse
|
19
|
Ulivieri C, Savino MT, Luccarini I, Fanigliulo E, Aldinucci A, Bonechi E, Benagiano M, Ortensi B, Pelicci G, D'Elios MM, Ballerini C, Baldari CT. The Adaptor Protein Rai/ShcC Promotes Astrocyte-Dependent Inflammation during Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2016; 197:480-90. [PMID: 27288534 DOI: 10.4049/jimmunol.1502063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 05/13/2016] [Indexed: 01/13/2023]
Abstract
Th17 cells have been casually associated to the pathogenesis of autoimmune disease. We have previously demonstrated that Rai/ShcC, a member of the Shc family of adaptor proteins, negatively regulates Th17 cell differentiation and lupus autoimmunity. In this study, we have investigated the pathogenic outcome of the Th17 bias associated with Rai deficiency on multiple sclerosis development, using the experimental autoimmune encephalomyelitis (EAE) mouse model. We found that, unexpectedly, EAE was less severe in Rai(-/-) mice compared with their wild-type counterparts despite an enhanced generation of myelin-specific Th17 cells that infiltrated into the CNS. Nevertheless, when adoptively transferred into immunodeficient Rai(+/+) mice, these cells promoted a more severe disease compared with wild-type encephalitogenic Th17 cells. This paradoxical phenotype was caused by a dampened inflammatory response of astrocytes, which were found to express Rai, to IL-17. The results provide evidence that Rai plays opposite roles in Th17 cell differentiation and astrocyte activation, with the latter dominant over the former in EAE, highlighting this adaptor as a potential novel target for the therapy of multiple sclerosis.
Collapse
Affiliation(s)
- Cristina Ulivieri
- Department of Life Sciences, University of Siena, 2 53100, Siena, Italy;
| | | | | | | | | | - Elena Bonechi
- Department of Neurosciences, 6 50134 Florence, Italy
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, 3 50134 Florence, Italy; and
| | - Barbara Ortensi
- Department of Experimental Oncology, European Institute of Oncology, 16 20139 Milan, Italy
| | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology, 16 20139 Milan, Italy
| | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, 3 50134 Florence, Italy; and
| | | | | |
Collapse
|
20
|
Muthusamy BP, Budi EH, Katsuno Y, Lee MK, Smith SM, Mirza AM, Akhurst RJ, Derynck R. ShcA Protects against Epithelial-Mesenchymal Transition through Compartmentalized Inhibition of TGF-β-Induced Smad Activation. PLoS Biol 2015; 13:e1002325. [PMID: 26680585 PMCID: PMC4682977 DOI: 10.1371/journal.pbio.1002325] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/10/2015] [Indexed: 12/15/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a normal cell differentiation event during development and contributes pathologically to carcinoma and fibrosis progression. EMT often associates with increased transforming growth factor-β (TGF-β) signaling, and TGF-β drives EMT, in part through Smad-mediated reprogramming of gene expression. TGF-β also activates the Erk MAPK pathway through recruitment and Tyr phosphorylation of the adaptor protein ShcA by the activated TGF-β type I receptor. We found that ShcA protects the epithelial integrity of nontransformed cells against EMT by repressing TGF-β-induced, Smad-mediated gene expression. p52ShcA competed with Smad3 for TGF-β receptor binding, and down-regulation of ShcA expression enhanced autocrine TGF-β/Smad signaling and target gene expression, whereas increased p52ShcA expression resulted in decreased Smad3 binding to the TGF-β receptor, decreased Smad3 activation, and increased Erk MAPK and Akt signaling. Furthermore, p52ShcA sequestered TGF-β receptor complexes to caveolin-associated membrane compartments, and reducing ShcA expression enhanced the receptor localization in clathrin-associated membrane compartments that enable Smad activation. Consequently, silencing ShcA expression induced EMT, with increased cell migration, invasion, and dissemination, and increased stem cell generation and mammosphere formation, dependent upon autocrine TGF-β signaling. These findings position ShcA as a determinant of the epithelial phenotype by repressing TGF-β-induced Smad activation through differential partitioning of receptor complexes at the cell surface. The adaptor protein ShcA protects epithelial cells from transitioning toward a mesenchymal phenotype by controlling partitioning of the TGF-β receptor and repressing downstream Smad2/3 activation. TGF-β family proteins control cell differentiation and various cell functions. Increased TGF-β signaling, acting through heteromeric receptor complexes, contributes to carcinoma progression and fibrosis. TGF-β drives epithelial–mesenchymal transdifferentiation (EMT), which enables cell migration and invasion. Upon TGF-β binding, “type I” receptors activate, through phosphorylation, Smad2 and Smad3 that control target gene transcription. In EMT, Smad complexes activate the expression of EMT “master” transcription factors and cooperate with these to repress the epithelial phenotype and activate mesenchymal gene expression. TGF-β receptors also activate Erk MAPK signaling, involving association of the adaptor protein ShcA and Tyr phosphorylation of ShcA by type I receptors. We now show that the predominant ShcA isoform, p52ShcA, competes with Smad2/3 for binding to type I TGF-β receptors, thus repressing Smad2/3 activation in response to TGF-β and localizing the receptors to caveolar compartments. Consequently, decreased ShcA expression enhanced TGF-β receptor localization in clathrin compartments and autocrine Smad2/3 signaling, repressed the epithelial phenotype, and promoted EMT. The changes following decreased ShcA expression resulted in increased cell migration and invasion, as well as increased stem cell generation, dependent upon autocrine TGF-β signaling. These findings position ShcA as a determinant of the epithelial phenotype by repressing TGF-β-induced Smad activation through differential partitioning of receptor complexes at the cell surface.
Collapse
Affiliation(s)
- Baby Periyanayaki Muthusamy
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
| | - Erine H. Budi
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
| | - Yoko Katsuno
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
| | - Matthew K. Lee
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Susan M. Smith
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Amer M. Mirza
- XOMA Corp., Berkeley, California, United States of America
| | - Rosemary J. Akhurst
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
- Department of Anatomy, University of California, San Francisco, San Francisco, California, United States of America
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, United States of America
| | - Rik Derynck
- Departments of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
- Department of Anatomy, University of California, San Francisco, San Francisco, California, United States of America
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
21
|
Tang N, Lyu D, Liu T, Chen F, Jing S, Hao T, Liu S. Different Effects of p52SHC1 and p52SHC3 on the Cell Cycle of Neurons and Neural Stem Cells. J Cell Physiol 2015; 231:172-80. [DOI: 10.1002/jcp.25069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 04/27/2015] [Accepted: 06/05/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Ning Tang
- State Key Laboratory of Proteomics and; Department of Neurobiology; Institute of Basic Medical Sciences; Beijing P. R. China
- Reproductive Medicine Center; Jinan Military General Hospital; Jinan P. R. China
| | - Dan Lyu
- State Key Laboratory of Proteomics and; Department of Neurobiology; Institute of Basic Medical Sciences; Beijing P. R. China
- Dan Lyu is currently working in Department of Pain Management; Tianjin First Center Hospital; Tianjin P. R. China
| | - Tao Liu
- State Key Laboratory of Proteomics and; Department of Neurobiology; Institute of Basic Medical Sciences; Beijing P. R. China
| | - Fangjin Chen
- State Key Laboratory of Proteomics and; Department of Neurobiology; Institute of Basic Medical Sciences; Beijing P. R. China
| | - Shuqian Jing
- State Key Laboratory of Proteomics and; Department of Neurobiology; Institute of Basic Medical Sciences; Beijing P. R. China
| | - Tianyu Hao
- Reproductive Medicine Center; Jinan Military General Hospital; Jinan P. R. China
| | - Shaojun Liu
- State Key Laboratory of Proteomics and; Department of Neurobiology; Institute of Basic Medical Sciences; Beijing P. R. China
| |
Collapse
|
22
|
Prescott JD, Zeiger MA. TheREToncogene in papillary thyroid carcinoma. Cancer 2015; 121:2137-46. [DOI: 10.1002/cncr.29044] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/21/2014] [Accepted: 08/26/2014] [Indexed: 01/19/2023]
Affiliation(s)
- Jason D. Prescott
- Endocrine Surgery, Department of Surgery; The Johns Hopkins University School of Medicine; Baltimore Maryland
| | - Martha A. Zeiger
- Endocrine Surgery, Department of Surgery; The Johns Hopkins University School of Medicine; Baltimore Maryland
| |
Collapse
|
23
|
Sagi O, Budovsky A, Wolfson M, Fraifeld VE. ShcC proteins: brain aging and beyond. Ageing Res Rev 2015; 19:34-42. [PMID: 25462193 DOI: 10.1016/j.arr.2014.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 11/08/2014] [Accepted: 11/17/2014] [Indexed: 02/02/2023]
Abstract
To date, most studies of Shc family of signaling adaptor proteins have been focused on the near-ubiquitously expressed ShcA, indicating its relevance to age-related diseases and longevity. Although the role of the neuronal ShcC protein is much less investigated, accumulated evidence suggests its importance for neuroprotection against such aging-associated conditions as brain ischemia and oxidative stress. Here, we summarize more than decade of studies on the ShcC expression and function in normal brain, age-related brain pathologies and immune disorders with a focus on the interactions of ShcC with signaling proteins/pathways, and the possible implications of these interactions for changes associated with aging.
Collapse
Affiliation(s)
- Orli Sagi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Arie Budovsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; Judea Regional Research & Development Center, Carmel 90404, Israel
| | - Marina Wolfson
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Vadim E Fraifeld
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
24
|
Cell density modulates SHC3 expression and survival of human glioblastoma cells through Fak activation. J Neurooncol 2014; 120:245-56. [PMID: 25062668 DOI: 10.1007/s11060-014-1551-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 07/05/2014] [Indexed: 10/25/2022]
Abstract
Shc3 protein levels are high in human glioblastoma but they decrease in vitro. We found that SHC3 mRNA and protein increased when glioblastoma cells grew as multicellular tumor spheroid (MTS). Shc3 expression was also induced in adherent cultures by increasing cell density. Among the Shc family members, only Shc2 and Shc3 increased with cell density. Shc3 and focal adhesion kinase (Fak) interact as shown by co-immunoprecipitation. Inhibition of Fak activation reduced Shc3 increase and MTS formation and changed Shc3 phosphorylation pattern. Our results suggest that in gliomas cell density modulates Shc3 protein levels and its activity, at least in part, through Fak activation.
Collapse
|
25
|
Wills MKB, Tong J, Tremblay SL, Moran MF, Jones N. The ShcD signaling adaptor facilitates ligand-independent phosphorylation of the EGF receptor. Mol Biol Cell 2014; 25:739-52. [PMID: 24430869 PMCID: PMC3952845 DOI: 10.1091/mbc.e13-08-0434] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 12/06/2013] [Accepted: 01/08/2014] [Indexed: 11/12/2022] Open
Abstract
Proto-oncogenic Src homology and collagen (Shc) proteins have been considered archetypal adaptors of epidermal growth factor receptor (EGFR)-mediated signaling. We report that in addition to its role as an EGFR-binding partner and Grb2 platform, ShcD acts noncanonically to promote phosphorylation of select EGFR residues. Unexpectedly, Y1068, Y1148, and Y1173 are subject to ShcD-induced, cell-autonomous hyperphosphorylation in the absence of external stimuli. This response is not elicited by other Shc proteins and requires the intrinsic EGFR kinase, as well as the ShcD phosphotyrosine-binding (PTB) domain. Assessments of Erk, Akt, phospholipase C 1γ, and FAK pathways reveal no apparent distal signaling targets of ShcD. Nevertheless, the capacity of cultured cells to repopulate a wounded monolayer is markedly accelerated by ShcD in an EGFR kinase-dependent manner. Furthermore, detection of overexpressed ShcD coincident with EGFR phosphorylation in human gliomas suggests a clinical application for these findings. We thus demonstrate unique and relevant synergy between ShcD and EGFR that is unprecedented among signaling adaptors.
Collapse
Affiliation(s)
- Melanie K. B. Wills
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Jiefei Tong
- Program in Molecular Structure and Function, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Sylvie L. Tremblay
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Michael F. Moran
- Program in Molecular Structure and Function, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Molecular Genetics and Banting and Best Department of Medical Research, University of Toronto, Toronto, ON M5G 1L6, Canada
| | - Nina Jones
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
26
|
DeNardo BD, Holloway MP, Ji Q, Nguyen KT, Cheng Y, Valentine MB, Salomon A, Altura RA. Quantitative phosphoproteomic analysis identifies activation of the RET and IGF-1R/IR signaling pathways in neuroblastoma. PLoS One 2013; 8:e82513. [PMID: 24349301 PMCID: PMC3859635 DOI: 10.1371/journal.pone.0082513] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 10/24/2013] [Indexed: 12/20/2022] Open
Abstract
Neuroblastoma is an embryonal tumor of childhood with a heterogenous clinical presentation that reflects differences in activation of complex biological signaling pathways. Protein phosphorylation is a key component of cellular signal transduction and plays a critical role in processes that control cancer cell growth and survival. We used shotgun LC/MS to compare phosphorylation between a human MYCN amplified neuroblastoma cell line (NB10), modeling a resistant tumor, and a human neural precursor cell line (NPC), modeling a normal baseline neural crest cell. 2181 unique phosphorylation sites representing 1171 proteins and 2598 phosphopeptides were found. Protein kinases accounted for 6% of the proteome, with a predominance of tyrosine kinases, supporting their prominent role in oncogenic signaling pathways. Highly abundant receptor tyrosine kinase (RTK) phosphopeptides in the NB10 cell line relative to the NPC cell line included RET, insulin-like growth factor 1 receptor/insulin receptor (IGF-1R/IR), and fibroblast growth factor receptor 1 (FGFR1). Multiple phosphorylated peptides from downstream mediators of the PI3K/AKT/mTOR and RAS pathways were also highly abundant in NB10 relative to NPC. Our analysis highlights the importance of RET, IGF-1R/IR and FGFR1 as RTKs in neuroblastoma and suggests a methodology that can be used to identify potential novel biological therapeutic targets. Furthermore, application of this previously unexploited technology in the clinic opens the possibility of providing a new wide-scale molecular signature to assess disease progression and prognosis.
Collapse
Affiliation(s)
- Bradley D. DeNardo
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Michael P. Holloway
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Qinqin Ji
- Department of Chemistry, Brown University, Providence, Rhode Island, United States of America
| | - Kevin T. Nguyen
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Yan Cheng
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Marcus B. Valentine
- St. Jude Comprehensive Cancer Center Cytogenetic Shared Resource, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Arthur Salomon
- Department of Molecular and Cellular Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Rachel A. Altura
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
27
|
Turco MY, Furia L, Dietze A, Fernandez Diaz L, Ronzoni S, Sciullo A, Simeone A, Constam D, Faretta M, Lanfrancone L. Cellular heterogeneity during embryonic stem cell differentiation to epiblast stem cells is revealed by the ShcD/RaLP adaptor protein. Stem Cells 2013; 30:2423-36. [PMID: 22948967 PMCID: PMC3533801 DOI: 10.1002/stem.1217] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The Shc family of adaptor proteins are crucial mediators of a plethora of receptors such as the tyrosine kinase receptors, cytokine receptors, and integrins that drive signaling pathways governing proliferation, differentiation, and migration. Here, we report the role of the newly identified family member, ShcD/RaLP, whose expression in vitro and in vivo suggests a function in embryonic stem cell (ESC) to epiblast stem cells (EpiSCs) transition. The transition from the naïve (ESC) to the primed (EpiSC) pluripotent state is the initial important step for ESCs to commit to differentiation and the mechanisms underlying this process are still largely unknown. Using a novel approach to simultaneously assess pluripotency, apoptosis, and proliferation by multiparameter flow cytometry, we show that ESC to EpiSC transition is a process involving a tight coordination between the modulation of the Oct4 expression, cell cycle progression, and cell death. We also describe, by high-content immunofluorescence analysis and time-lapse microscopy, the emergence of cells expressing caudal-related homeobox 2 (Cdx2) transcription factor during ESC to EpiSC transition. The use of the ShcD knockout ESCs allowed the unmasking of this process as they presented deregulated Oct4 modulation and an enrichment in Oct4-negative Cdx2-positive cells with increased MAPK/extracellular-regulated kinases 1/2 activation, within the differentiating population. Collectively, our data reveal ShcD as an important modulator in the switch of key pathway(s) involved in determining EpiSC identity.
Collapse
Affiliation(s)
- Margherita Y Turco
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Savino MT, Ulivieri C, Emmi G, Prisco D, De Falco G, Ortensi B, Beccastrini E, Emmi L, Pelicci G, D'Elios MM, Baldari CT. The Shc family protein adaptor, Rai, acts as a negative regulator of Th17 and Th1 cell development. J Leukoc Biol 2013; 93:549-59. [PMID: 23345394 DOI: 10.1189/jlb.0712331] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rai, a Shc adapter family member, acts as a negative regulator of antigen receptor signaling in T and B cells. Rai(-/-) mice develop lupus-like autoimmunity associated to the spontaneous activation of self-reactive lymphocytes. Here, we have addressed the potential role of Rai in the development of the proinflammatory Th1 and Th17 subsets, which are centrally implicated in the pathogenesis of a number of autoimmune diseases, including lupus. We show that Rai(-/-) mice display a spontaneous Th1/Th17 bias. In vitro polarization experiments on naive and effector/memory CD4(+) T cells demonstrate that Rai(-/-) favors the development and expansion of Th17 but not Th1 cells, indicating that Rai modulates TCR signaling to antagonize the pathways driving naive CD4(+) T cell differentiation to the Th17 lineage, while indirectly limiting Th1 cell development in vivo. Th1 and Th17 cell infiltrates were found in the kidneys of Rai(-/-) mice, providing evidence that Rai(-/-) contributes to the development of lupus nephritis, not only by enhancing lymphocyte activation but also by promoting the development and expansion of proinflammatory effector T cells. Interestingly, T cells from SLE patients were found to have a defect in Rai expression, suggesting a role for Rai in disease pathogenesis.
Collapse
|
29
|
Giunti S, Antonelli A, Amorosi A, Santarpia L. Cellular signaling pathway alterations and potential targeted therapies for medullary thyroid carcinoma. Int J Endocrinol 2013; 2013:803171. [PMID: 23509459 PMCID: PMC3594951 DOI: 10.1155/2013/803171] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 01/08/2013] [Accepted: 01/10/2013] [Indexed: 12/12/2022] Open
Abstract
Parafollicular C-cell-derived medullary thyroid cancer (MTC) comprises 3% to 4% of all thyroid cancers. While cytotoxic treatments have been shown to have limited efficacy, targeted molecular therapies that inhibit rearranged during transfection (RET) and other tyrosine kinase receptors that are mainly involved in angiogenesis have shown great promise in the treatment of metastatic or locally advanced MTC. Multi-tyrosine kinase inhibitors such as vandetanib, which is already approved for the treatment of progressive MTC, and cabozantinib have shown distinct advantages with regard to rates of disease response and control. However, these types of tyrosine kinase inhibitor compounds are able to concurrently block several types of targets, which limits the understanding of RET as a specific target. Moreover, important resistances to tyrosine kinase inhibitors can occur, which limit the long-term efficacy of these treatments. Deregulated cellular signaling pathways and genetic alterations in MTC, particularly the activation of the RAS/mammalian target of rapamycin (mTOR) cascades and RET crosstalk signaling, are now emerging as novel and potentially promising therapeutic treatments for aggressive MTC.
Collapse
Affiliation(s)
- Serena Giunti
- Department of Pathology, Centro Oncologico Fiorentino, Sesto Fiorentino, 50019 Firenze, Italy
| | - Alessandro Antonelli
- Department of Internal Medicine, University of Pisa School of Medicine, 56100 Pisa, Italy
| | - Andrea Amorosi
- Department of Pathology, Centro Oncologico Fiorentino, Sesto Fiorentino, 50019 Firenze, Italy
| | - Libero Santarpia
- Translational Research Unit, Department of Oncology, Istituto Toscano Tumori, 59100 Prato, Italy
- *Libero Santarpia:
| |
Collapse
|
30
|
Abstract
Shc (Src homology and collagen homology) proteins are considered prototypical signalling adaptors in mammalian cells. Consisting of four unique members, ShcA, B, C and D, and multiple splice isoforms, the family is represented in nearly every cell type in the body, where it engages in an array of fundamental processes to transduce environmental stimuli. Two decades of investigation have begun to illuminate the mechanisms of the flagship ShcA protein, whereas much remains to be learned about the newest discovery, ShcD. It is clear, however, that the distinctive modular architecture of Shc proteins, their promiscuous phosphotyrosine-based interactions with a multitude of membrane receptors, involvement in central cascades including MAPK (mitogen-activated protein kinase) and Akt, and unconventional contributions to oxidative stress and apoptosis all require intricate regulation, and underlie diverse physiological function. From early cardiovascular development and neuronal differentiation to lifespan determination and tumorigenesis, Shc adaptors have proven to be more ubiquitous, versatile and dynamic than their structures alone suggest.
Collapse
|
31
|
Ortensi B, Osti D, Pellegatta S, Pisati F, Brescia P, Fornasari L, Levi D, Gaetani P, Colombo P, Ferri A, Nicolis S, Finocchiaro G, Pelicci G. Rai is a New Regulator of Neural Progenitor Migration and Glioblastoma Invasion. Stem Cells 2012; 30:817-32. [DOI: 10.1002/stem.1056] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
32
|
Abstract
The risk of patients with Hirschsprung's disease later developing multiple endocrine neoplasia remains a matter of concern. The multiple endocrine neoplasia 2-Hirschsprung's disease association has been shown to cosegregate in Hirschsprung's disease patients with both short- and long-segment aganglionosis, although patients with long-segment aganglionosis a to carry the greatest risk. The Hirschsprung's disease-medullary thyroid carcinoma relationship also appears to be bi-directional, and activation or suppression of the rearranged during transfection gene appeared to vary over succeeding generations within the same family. Rearranged during transfection gene variations are associated with both conditions. The cosegregation of Hirschsprung's disease and multiple endocrine neoplasia 2 is particularly interesting as it involves both "switch off" and "switch on" of the rearranged during transfection proto-oncogene in the same patient. This cosegregation mostly relates to the cysteine-rich area on RET620 (the "Janus gene"). The mechanism whereby rearranged during transfection influences gene activation in multiple endocrine neoplasia 2 is complex, but genetic variations impair the rearranged during transfection tyrosine kinase response to tyrosine kinase activation, thus appearing to dictate downstream signaling cascade responses. Better understanding of the RET-620 relationship allows for a more cost-effective method of identifying those at risk by focusing rearranged during transfection gene testing to this specific area as a "hot spot". The clinical awareness of possible medullary thyroid carcinoma has led to timely intervention and early treatment of this chemo- and radioresistant tumor with poor prognosis. Establishment of "risk" by genetic testing has become a classic model of molecular medicine being integrated into patient care and offering rearranged during transfection directed prophylactic surgical management. In addition, novel approaches to treatment based on this genetic knowledge have already shown early promise in randomized clinical trials.
Collapse
Affiliation(s)
- Sam W Moore
- Division of Pediatric Surgery, Faculty of Medicine, University of Stellenbosch, Tygerberg, South Africa.
| | | |
Collapse
|
33
|
Ferro M, Savino MT, Ortensi B, Finetti F, Genovese L, Masi G, Ulivieri C, Benati D, Pelicci G, Baldari CT. The Shc family protein adaptor, Rai, negatively regulates T cell antigen receptor signaling by inhibiting ZAP-70 recruitment and activation. PLoS One 2011; 6:e29899. [PMID: 22242145 PMCID: PMC3248456 DOI: 10.1371/journal.pone.0029899] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 12/08/2011] [Indexed: 12/16/2022] Open
Abstract
Rai/ShcC is a member of the Shc family of protein adaptors expressed with the highest abundance in the central nervous system, where it exerts a protective function by coupling neurotrophic receptors to the PI3K/Akt survival pathway. Rai is also expressed, albeit at lower levels, in other cell types, including T and B lymphocytes. We have previously reported that in these cells Rai attenuates antigen receptor signaling, thereby impairing not only cell proliferation but also, opposite to neurons, cell survival. Here we have addressed the mechanism underlying the inhibitory activity of Rai on TCR signaling. We show that Rai interferes with the TCR signaling cascade one of the earliest steps –recruitment of the initiating kinase ZAP-70 to the phosphorylated subunit of the TCR/CD3 complex, which results in a generalized dampening of the downstream signaling events. The inhibitory activity of Rai is associated to its inducible recruitment to phosphorylated CD3, which occurs in the physiological signaling context of the immune synapse. Rai is moreover found as a pre-assembled complex with ZAP-70 and also constitutively interacts with the regulatory p85 subunit of PI3K, similar to neuronal cells, notwithstanding the opposite biological outcome, i.e. impairment of PI-3K/Akt activation. The data highlight the ability of Rai to establish interactions with the TCR and key signaling mediators which, either directly (e.g. by inhibiting ZAP-70 recruitment to the TCR or sequestering ZAP-70/PI3K in the cytosol) or indirectly (e.g. by promoting the recruitment of effectors responsible for signal extinction) prevent full triggering of the TCR signaling cascade.
Collapse
Affiliation(s)
- Micol Ferro
- Department of Evolutionary Biology European Institute of Oncology, Milan, Italy
| | - Maria Teresa Savino
- Department of Evolutionary Biology European Institute of Oncology, Milan, Italy
| | - Barbara Ortensi
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Francesca Finetti
- Department of Evolutionary Biology European Institute of Oncology, Milan, Italy
| | - Luca Genovese
- Department of Evolutionary Biology European Institute of Oncology, Milan, Italy
| | - Giulia Masi
- Department of Evolutionary Biology European Institute of Oncology, Milan, Italy
| | - Cristina Ulivieri
- Department of Evolutionary Biology European Institute of Oncology, Milan, Italy
| | - Daniela Benati
- Department of Evolutionary Biology European Institute of Oncology, Milan, Italy
| | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Cosima T. Baldari
- Department of Evolutionary Biology European Institute of Oncology, Milan, Italy
- Istituto Toscano Tumori, University of Siena, Siena, Italy
- * E-mail:
| |
Collapse
|
34
|
Bassotti G, Villanacci V. Can "functional" constipation be considered as a form of enteric neuro-gliopathy? Glia 2011; 59:345-350. [PMID: 21264943 DOI: 10.1002/glia.21115] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 11/03/2010] [Indexed: 12/12/2022]
Abstract
Constipation has been traditionally viewed and classified as a functional or idiopathic disorder. However, evidence has been accumulating that suggests how constipation might be considered as due to abnormalities of the enteric nervous system, since alterations of this system, not evident in conventional histological examination, may be present in these patients. These abnormalities often consist in decrease or loss of the enteric glial cells, a pathological finding present in most types of constipation so far investigated. In this article we will discuss these evidences, and will try to consider constipation no more as a simple functional or idiopathic disorder but as a form of enteric neuro-gliopathy.
Collapse
Affiliation(s)
- Gabrio Bassotti
- Department of Clinical and Experimental Medicine, Gastroenterology and Hepatology Section, University of Perugia, Italy.
| | | |
Collapse
|
35
|
Schettini G, Govoni S, Racchi M, Rodriguez G. Phosphorylation of APP-CTF-AICD domains and interaction with adaptor proteins: signal transduction and/or transcriptional role--relevance for Alzheimer pathology. J Neurochem 2010; 115:1299-308. [PMID: 21039524 DOI: 10.1111/j.1471-4159.2010.07044.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In recent decades, the study of the amyloid precursor protein (APP) and of its proteolytic products carboxy terminal fragment (CTF), APP intracellular C-terminal domain (AICD) and amyloid beta has been mostly focussed on the role of APP as a producer of the toxic amyloid beta peptide. Here, we reconsider the role of APP suggesting, in a provocative way, the protein as a central player in a putative signalling pathway. We highlight the presence in the cytosolic tail of APP of the YENPTY motif which is typical of tyrosine kinase receptors, the phosphorylation of the tyrosine, serine and threonine residues, the kinases involved and the interaction with intracellular adaptor proteins. In particular, we examine the interaction with Shc and Grb2 regulators, which through the activation of Ras proteins elicit downstream signalling events such as the MAPK pathway. The review also addresses the interaction of APP, CTFs and AICD with other adaptor proteins and in particular with Fe65 for nuclear transcriptional activity and the importance of phosphorylation for sorting the secretases involved in the amyloidogenic or non-amyloidogenic pathways. We provide a novel perspective on Alzheimer's disease pathogenesis, focussing on the perturbation of the physiological activities of APP-CTFs and AICD as an alternative perspective from that which normally focuses on the accumulation of neurotoxic proteolytic fragments.
Collapse
Affiliation(s)
- Gennaro Schettini
- Department of Oncology, Biology and Genetics, University of Genova, Genova, Italy.
| | | | | | | |
Collapse
|
36
|
Santarpia L, Myers JN, Sherman SI, Trimarchi F, Clayman GL, El-Naggar AK. Genetic alterations in the RAS/RAF/mitogen-activated protein kinase and phosphatidylinositol 3-kinase/Akt signaling pathways in the follicular variant of papillary thyroid carcinoma. Cancer 2010; 116:2974-83. [PMID: 20564403 DOI: 10.1002/cncr.25061] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND The follicular variant of papillary thyroid carcinoma (FVPTC) is the second most common histotype among papillary thyroid cancers (PTCs). Although the prognosis of FVPTC is similar to the conventional phenotype, differential diagnostic difficulties may not be uncommon with other follicular thyroid neoplasms, and little is known about their genetic alterations. Defining these alterations may lead to the identification of diagnostic and biologic markers. METHODS In this study, the authors evaluated genetic alterations and downstream-activated signals of the Ras/Raf-mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)/v-akt murine thymoma viral oncogene (Akt) (PI3K/Akt) signaling pathways in 30 FVPTC tissue specimens. Tumors and matched normal thyroid samples were tested for RAS, for the v-raf murine sarcoma viral oncogene (BRAF) substitution of valine (V) for glutamate (E) at codon 600 (the V600E mutation), for phosphatase and tensin homolog (PTEN), for catalytic PI3k p110 subunit alpha (PIK3CA), for AKT, and for the presence of rearranged during transfection (ret) proto-oncogene/PTC (RET-PTC) and paired box-8 (PAX8)/peroxisome proliferator-activated receptor gamma (PPARgamma) fusion protein (PAX8-PPARgamma) rearrangements by direct sequencing and reverse transcriptase-polymerases chain reaction analyses, respectively. Western blot analysis was used to assess the effects of these gene abnormalities on the activation of the 2 pathways. RESULTS Genetic alterations were identified in 70% of FVPTCs. Activation of the MAPK and PI3K pathways was observed in 74% and 22% of tumors, respectively. The alterations that were identified in the genes of the 2 pathways were mutually exclusive. Chromosomal RET-PTC and PAX8-PPARgamma rearrangements were observed in 20% and 17% of tumors, respectively. It was noteworthy that some FVPTCs with RET-PTC had the coactivation of both pathways. CONCLUSIONS RET-PTC and PAX8-PPARgamma rearrangements and mutations of the neuroblastoma RAS viral oncogene homolog N-RAS at codon 61 were the most common genetic alterations in FVPTCs. Activation of the MAPK pathway was a frequent event in FVPTCs, and the PI3K signaling pathway could be coactivated in RET-PTC tumors. These findings may have important therapeutic implication in patients with FVPTC.
Collapse
Affiliation(s)
- Libero Santarpia
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | |
Collapse
|
37
|
Finetti F, Savino MT, Baldari CT. Positive and negative regulation of antigen receptor signaling by the Shc family of protein adapters. Immunol Rev 2010; 232:115-34. [PMID: 19909360 DOI: 10.1111/j.1600-065x.2009.00826.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The Shc adapter family includes four members that are expressed as multiple isoforms and participate in signaling by a variety of cell-surface receptors. The biological relevance of Shc proteins as well as their variegated function, which relies on their highly conserved modular structure, is underscored by the distinct and dramatic phenotypic alterations resulting from deletion of individual Shc isoforms both in the mouse and in two model organisms, Drosophila melanogaster and Caenorhabditis elegans. The p52 isoform of ShcA couples antigen and cytokine receptors to Ras activation in both lymphoid and myeloid cells. However, the recognition of the spectrum of activities of p52ShcA in the immune system has been steadily expanding in recent years to other fundamental processes both at the cell and organism levels. Two other Shc family members, p66ShcA and p52ShcC/Rai, have been identified recently in T and B lymphocytes, where they antagonize survival and attenuate antigen receptor signaling. These developments reveal an unexpected and complex interplay of multiple Shc proteins in lymphocytes.
Collapse
Affiliation(s)
- Francesca Finetti
- Department of Evolutionary Biology, University of Siena, Siena, Italy
| | | | | |
Collapse
|
38
|
Magrassi L, Marziliano N, Inzani F, Cassini P, Chiaranda I, Skrap M, Pizzolito S, Arienta C, Arbustini E. EDG3 and SHC3 on chromosome 9q22 are co-amplified in human ependymomas. Cancer Lett 2009; 290:36-42. [PMID: 19748727 DOI: 10.1016/j.canlet.2009.08.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 08/17/2009] [Accepted: 08/18/2009] [Indexed: 11/16/2022]
Abstract
By qPCR we found that EDG3 and SHC3 were amplified in 60% of ependymomas but none in choroid plexus papillomas. In ependymomas EDG3 and SHC3 amplification increased Shc3 protein levels while EDG3 was less affected. Both proteins were co-immunoprecipitated from ependymoma and Shc3 was tyrosine phosphorylated thus presumably active. We showed by digestion with N-glycosidase-F that EDG3 was glycosylated indicating that EDG3 protein was not retained in the endoplasmic reticulum. The co-immunoprecipitation of Shc3 and EDG3 proteins from ependymomas with amplification of SHC3 and EDG3 genes suggests that the two proteins co-operate and are important for ependymomas in vivo.
Collapse
Affiliation(s)
- Lorenzo Magrassi
- Neurochirurgia Dipartimento di Scienze Chirurgiche Università di Pavia Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Santarpia L, Ye L, Gagel RF. Beyond RET: potential therapeutic approaches for advanced and metastatic medullary thyroid carcinoma. J Intern Med 2009; 266:99-113. [PMID: 19522829 DOI: 10.1111/j.1365-2796.2009.02112.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Medullary thyroid carcinoma (MTC) is a rare calcitonin-producing neuroendocrine tumour that originates from the parafollicular C-cells of the thyroid gland. The RET proto-oncogene encodes the RET receptor tyrosine kinase, which has essential roles in cell survival, differentiation and proliferation. Activating mutations of RET are associated with the pathogenesis of MTC and have been demonstrated in nearly all hereditary and in 30-50% of sporadic MTC cases, making this receptor an excellent target for small-molecule inhibitors for this tumour. Clinical trials of small organic inhibitors of tyrosine kinase receptors (TKIs) targeting the RET receptor have shown efficacy for treatment of metastatic MTC with 30-50% of patients responding to these agents. Despite the importance of the RET receptor in MTC, it is clear that other signal transduction pathways, tyrosine kinase receptors, and tumour suppressor genes are involved in MTC tumourigenesis and progression. A better understanding of molecular cross-talk between these signal pathways and the RET receptor may lead to combinatorial therapy that will improve outcomes beyond what is currently possible with RET-directed TKIs. Finally, there is evidence that immunological-based therapy using dendritic cell vaccination strategies have been effective for reducing tumour mass in a small number of patients. The identification of additional MTC-specific tumour antigens and a better understanding of specific epitopes in these tumour antigens may lead to improvement of response rates.
Collapse
Affiliation(s)
- L Santarpia
- The Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
40
|
Savino MT, Ortensi B, Ferro M, Ulivieri C, Fanigliulo D, Paccagnini E, Lazzi S, Osti D, Pelicci G, Baldari CT. Rai acts as a negative regulator of autoimmunity by inhibiting antigen receptor signaling and lymphocyte activation. THE JOURNAL OF IMMUNOLOGY 2009; 182:301-8. [PMID: 19109161 DOI: 10.4049/jimmunol.182.1.301] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Rai (ShcC) belongs to the family of Shc adaptor proteins and is expressed in neuronal cells, where it acts as a survival factor activating the PI3K/Akt survival pathway. In vivo, Rai protects the brain from ischemic damage. In this study, we show that Rai is expressed in T and B lymphocytes. Based on the finding that Rai(-/-) mice consistently develop splenomegaly, the role of Rai in lymphocyte homeostasis and proliferation was addressed. Surprisingly, as opposed to neurons, Rai was found to impair lymphocyte survival. Furthermore, Rai deficiency results in a reduction in the frequency of peripheral T cells with a concomitant increase in the frequency of B cells. Rai(-/-) lymphocytes display enhanced proliferative responses to Ag receptor engagement in vitro, which correlates with enhanced signaling by the TCR and BCR, and more robust responses to allergen sensitization in vivo. A high proportion of Rai(-/-) mice develop a lupus-like autoimmune syndrome characterized by splenomegaly, spontaneous peripheral T and B cell activation, autoantibody production, and deposition of immune complexes in the kidney glomeruli, resulting in autoimmune glomerulonephritis. The data identify Rai as a negative regulator of lymphocyte survival and activation and show that loss of this protein results in breaking of immunological tolerance and development of systemic autoimmunity.
Collapse
|
41
|
Pasini L, Turco MY, Luzi L, Aladowicz E, Fagiani E, Lanfrancone L. Melanoma: targeting signaling pathways and RaLP. Expert Opin Ther Targets 2008; 13:93-104. [DOI: 10.1517/14728220802607363] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
42
|
Lodyga M, De Falco V, Bai XH, Kapus A, Melillo RM, Santoro M, Liu M. XB130, a tissue-specific adaptor protein that couples the RET/PTC oncogenic kinase to PI 3-kinase pathway. Oncogene 2008; 28:937-49. [PMID: 19060924 DOI: 10.1038/onc.2008.447] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
XB130 is a recently cloned 130 kDa-adaptor protein and Src kinase substrate, structurally similar to actin-filament-associated protein. Here we show that XB130 is predominantly expressed in the thyroid. Given that XB130 is a thyroid-specific tyrosine kinase substrate, we asked whether it is targeted by RET/PTC, a genetically rearranged, constitutively active, thyroid-specific tyrosine kinase that plays a pathogenic role in papillary thyroid cancer. RET/PTC induced robust tyrosine phosphorylation of XB130, which promoted its subsequent association with the p85alpha subunit of phosphatidylinositol 3-kinase (PI 3-kinase). We identified tyrosine 54 of XB130 as the major target of RET/PTC-mediated phosphorylation and a critical binding site for the SH2 domains of p85alpha. Importantly, downregulation of XB130 in TPC1 papillary thyroid cancer cells, harboring the RET/PTC1 kinase, strongly reduced Akt activity without altering ERK1/2 phosphorylation, and concomitantly inhibited cell-cycle progression and survival in suspension. In conclusion, XB130 is a novel substrate of the RET/PTC kinase that links RET/PTC signaling to PI 3-kinase activation, and thereby plays an important role in sustaining proliferation and survival of thyroid tumor cells.
Collapse
Affiliation(s)
- M Lodyga
- Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
Moore SW, Zaahl MG. Multiple endocrine neoplasia syndromes, children, Hirschsprung's disease and RET. Pediatr Surg Int 2008; 24:521-30. [PMID: 18365214 DOI: 10.1007/s00383-008-2137-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2008] [Indexed: 01/20/2023]
Abstract
Multiple endocrine neoplasia (MEN) type 2 syndromes are autosomal dominant clinical associations characterized by a common clinical feature, medullary thyroid carcinoma (MTC). The ability to accurately predict the risk by genetic RET proto-oncogene analysis has resulted in the active follow-up of children at risk for developing early metastatic tumours and which can be prevented by prophylactic thyroidectomy. The C634 and M918T mutations (associated with MEN2A and MEN2B, respectively) are particularly associated with early aggressive behavior and distant metastatic spread requiring early intervention. RET is known to be involved in cellular signalling processes during development and controls the survival, proliferation, differentiation and migration of the enteric nervous system (ENS) progenitor cells, as well as the survival and regeneration of sympathetic neural and kidney cells. The centrality of RET in the etiology of both MEN2 and HSCR is now well established with fairly consistent associations existing between RET genotype and phenotype in MEN2. The relationship between Hirschsprung's disease (HSCR) MEN2 syndromes appears to be a highly significant one, sharing a common etiological factor in the RET proto-oncogene. It is now well accepted that most HSCR arises from loss of function, RET mutations, RET haploinsufficiency or RET polymorphisms and haplotypes of the RET promotor region. MEN2 syndromes result from gene up regulation due to germline activating mutations in the RET proto-oncogene (1:500,000). MTC is mostly associated with variations in the 5 cysteine RET radicals and codon-risk management protocols are of considerable value but not infallible. Oncogenic RET mutations may, however, vary between specific population groups. RET analysis in MEN has revolutionized the management of children of MEN2 and allowed surgical prediction and prophylaxis to take place. We discuss the role of genetic testing and possible guidelines for the management of patients from MTC families. The future appears full of promise and the current evaluation of RET-targeting tyrosine kinase and other inhibitors are of considerable interest in the management of these conditions.
Collapse
Affiliation(s)
- S W Moore
- Faculty of Health Sciences, University of Stellenbosch, 7505, Tygerberg, South Africa.
| | | |
Collapse
|
44
|
Villanacci V, Bassotti G, Ortensi B, Fisogni S, Cathomas G, Maurer CA, Galletti A, Salerni B, Pelicci G. Expression of the Rai (Shc C) adaptor protein in the human enteric nervous system. Neurogastroenterol Motil 2008; 20:206-212. [PMID: 17919311 DOI: 10.1111/j.1365-2982.2007.01017.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The adaptor protein Rai (ShcC/N-Shc) is almost exclusively present in the nervous system, although little is documented about its expression in the gut and the enteric nervous system (ENS). As Rai is a physiological substrate of Ret, an important factor for the development of ENS, we have evaluated the expression of Rai in the ENS in various segments of the human gastrointestinal tract. The expression of Rai was assessed by immunohistochemistry in disease-free human gut samples (oesophagus, stomach, small bowel and colon) obtained from subjects undergoing surgical procedures. Rai was not expressed in the epithelia or lymphoid tissue, whereas a moderate level of expression was observed in the endothelial cells of blood vessels and on the outer membrane of smooth muscle cells in both the muscularis mucosae and the muscularis propria. In the ENS, strong positivity was observed only in enteric glial cells, overlapping with GFAP and S100. In conclusion, Rai is expressed in the human gut, especially in the enteric glial cells. We conclude that Rai may provide an additional marker for this cell type.
Collapse
Affiliation(s)
- V Villanacci
- II Department of Pathology, Spedali Civili and University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Venezia V, Nizzari M, Carlo P, Corsaro A, Florio T, Russo C. Amyloid precursor protein and presenilin involvement in cell signaling. NEURODEGENER DIS 2007; 4:101-11. [PMID: 17596704 DOI: 10.1159/000101834] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
To date the most relevant role for the amyloid precursor protein (APP) and for the presenilins (PSs) on Alzheimer's disease (AD) genesis is linked to the 'amyloid hypothesis', which considers an aberrant formation of amyloid-beta peptides the cause of neurodegeneration. In this view, APP is merely a substrate, cleaved by the gamma-secretase complex to form toxic amyloid peptides, PSs are key players in gamma-secretase complex, and corollary or secondary events are Tau-linked pathology and gliosis. A second theory, complementary to the amyloid hypothesis, proposes that APP and PSs may modulate a yet unclear cell signal, the disruption of which may induce cell-cycle abnormalities, neuronal death, eventually amyloid formation and finally dementia. This hypothesis is supported by the presence of a complex network of proteins, with a clear relevance for signal transduction mechanisms, which interact with APP or PSs. In this scenario, the C-terminal domain of APP has a pivotal role due to the presence of the 682YENPTY687 motif that represents the docking site for multiple interacting proteins involved in cell signaling. In this review we discuss the significance of novel findings related to cell signaling events modulated by APP and PSs for AD development.
Collapse
Affiliation(s)
- Valentina Venezia
- Department of Oncology, Biology and Genetics, University of Genova, Genova, Italy
| | | | | | | | | | | |
Collapse
|
46
|
Gustin JA, Yang M, Johnson EM, Milbrandt J. Deciphering adaptor specificity in GFL-dependent RET-mediated proliferation and neurite outgrowth. J Neurochem 2007; 102:1184-94. [PMID: 17663753 DOI: 10.1111/j.1471-4159.2007.04624.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Glial cell derived neurotrophic factor (GDNF)-dependent receptor tyrosine kinase RET activity is required for proper development of the nervous system and genitourinary tract. Loss-of-function mutations in RET are associated with enteric nervous system abnormalities (Hirschsprung disease) and renal deficits (Potter's syndrome), whereas activating mutations lead to hereditary cancer syndromes (multiple endocrine neoplasia type 2A and type 2B). RET activation is crucial for the proper regulation of a variety of cellular processes including cell migration, proliferation and neurite outgrowth. By analyzing a series of RET mutants we found that Y1062 was critical for stimulating GDNF-mediated proliferation as well as proliferation stimulated by GDNF-independent oncogenic forms of RET. Studies using small interfering RNA driven by lentivirus to knock-down expression of particular adaptor proteins that interact with RET phospho-Y1062, demonstrated that only Src-homology 2 and growth factor receptor binding protein 2 were necessary for RET-mediated proliferation by wild type and oncogenic forms of RET. Interestingly, we discovered that Y1062 was also required for GDNF-stimulated neurite outgrowth. However, small interfering RNAs to either Src-homology 2 or growth factor receptor binding protein 2 or a panel of other adaptor proteins known to interact with RET Y1062 were incapable of blocking GDNF-stimulated neurite formation, indicating that differential use of intracellular adaptors is responsible for regulating alternative RET-stimulated cellular events such as proliferation versus a differentiation response like neurite outgrowth.
Collapse
Affiliation(s)
- Jason A Gustin
- Department of Pathology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
47
|
Jones N, Hardy WR, Friese MB, Jorgensen C, Smith MJ, Woody NM, Burden SJ, Pawson T. Analysis of a Shc family adaptor protein, ShcD/Shc4, that associates with muscle-specific kinase. Mol Cell Biol 2007; 27:4759-73. [PMID: 17452444 PMCID: PMC1951494 DOI: 10.1128/mcb.00184-07] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 03/12/2007] [Accepted: 04/13/2007] [Indexed: 12/31/2022] Open
Abstract
Shc family proteins serve as phosphotyrosine adaptor molecules in various receptor-mediated signaling pathways. In mammals, three distinct Shc genes have been described that encode proteins characterized by two phosphotyrosine-interaction modules, an amino-terminal phosphotyrosine binding (PTB) domain and a carboxy-terminal Src homology 2 domain. Here, we report the analysis of an uncharacterized fourth Shc family protein, ShcD/Shc4, that is expressed in adult brain and skeletal muscle. Consistent with this expression pattern, we find that ShcD can associate via its PTB domain with the phosphorylated muscle-specific kinase (MuSK) receptor tyrosine kinase and undergo tyrosine phosphorylation downstream of activated MuSK. Interestingly, additional sites of tyrosine phosphorylation, including a novel Grb2 binding site, are present on ShcD that are not found in other Shc family proteins. Activation of MuSK upon agrin binding at the neuromuscular junction (NMJ) induces clustering and tyrosine phosphorylation of acetylcholine receptors (AChRs) required for synaptic transmission. ShcD is coexpressed with MuSK in the postsynaptic region of the NMJ, and in cultured myotubes stimulated with agrin, expression of ShcD appears to be important for early tyrosine phosphorylation of the AChR. Thus, we have characterized a new member of the Shc family of docking proteins, which may mediate a specific aspect of signaling downstream of the MuSK receptor.
Collapse
Affiliation(s)
- Nina Jones
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Li MD, Sun D, Lou XY, Beuten J, Payne TJ, Ma JZ. Linkage and association studies in African- and Caucasian-American populations demonstrate that SHC3 is a novel susceptibility locus for nicotine dependence. Mol Psychiatry 2007; 12:462-73. [PMID: 17179996 DOI: 10.1038/sj.mp.4001933] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Our previous linkage study demonstrated that the 9q22-q23 chromosome region showed a 'suggestive' linkage to nicotine dependence (ND) in the Framingham Heart Study population. In this study, we provide further evidence for the linkage of this region to ND in an independent sample. Within this region, the gene encoding Src homology 2 domain-containing transforming protein C3 (SHC3) represents a plausible candidate for association with ND, assessed by smoking quantity (SQ), the Heaviness of Smoking Index (HSI) and the Fagerström Test for ND (FTND). We utilized 11 single-nucleotide polymorphisms within SHC3 to examine the association with ND in 602 nuclear families of either African-American (AA) or European-American (EA) origin. Individual SNP-based analysis indicated three SNPs for AAs and one for EAs were significantly associated with at least one ND measure. Haplotype analysis revealed that the haplotypes A-C-T-A-T-A of rs12519-rs3750399-rs4877042-rs2297313-rs1547696-rs1331188, with a frequency of 27.8 and 17.6%, and C-T-A-G-T of rs3750399-rs4877042-rs2297313-rs3818668-rs1547696, at a frequency of 44.7 and 30.6% in the AA and Combined samples, respectively, were significantly inversely associated with the ND measures. In the EA sample, another haplotype with a frequency of 10.6%, A-G-T-G of rs1331188-rs1556384-rs4534195-rs1411836, showed a significant inverse association with ND measures. These associations remained significant after Bonferroni correction. We further demonstrated the SHC3 contributed 40.1-59.2% (depending on the ND measures) of the linkage signals detected on chromosome 9. As further support, we found that nicotine administered through infusion increased the Shc3 mRNA level by 60% in the rat striatum, and decreased it by 22% in the nucleus accumbens (NA). At the protein level, Shc3 was decreased by 38.0% in the NA and showed no change in the striatum. Together, these findings strongly implicate SHC3 in the etiology of ND, which represents an important biological candidate for further investigation.
Collapse
Affiliation(s)
- M D Li
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22911, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Fagiani E, Giardina G, Luzi L, Cesaroni M, Quarto M, Capra M, Germano G, Bono M, Capillo M, Pelicci P, Lanfrancone L. RaLP, a New Member of the Src Homology and Collagen Family, Regulates Cell Migration and Tumor Growth of Metastatic Melanomas. Cancer Res 2007; 67:3064-73. [PMID: 17409413 DOI: 10.1158/0008-5472.can-06-2301] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Src homology and collagen (Src) family of adaptor proteins comprises six Shc-like proteins encoded by three loci in mammals (Shc, Rai, and Sli). Shc-like proteins are tyrosine kinase substrates, which regulate diverse signaling pathways and cellular functions, including Ras and proliferation (p52/p46Shc), phosphatidylinositol 3-kinase and survival (p54Rai), and mitochondrial permeability transition and apoptosis (p66Shc). Here, we report the identification, cloning, and sequence characterization of a new member of the Shc family that we termed RaLP. RaLP encodes a 69-kDa protein characterized by the CH2-PTB-CH1-SH2 modularity, typical of the Shc protein family, and expressed, among adult tissues, only in melanomas. Analysis of RaLP expression during the melanoma progression revealed low expression in normal melanocytes and benign nevi, whereas high levels of RaLP protein were found at the transition from radial growth phase to vertical growth phase and metastatic melanomas, when tumor cells acquire migratory competence and invasive potential. Notably, silencing of RaLP expression in metastatic melanomas by RNA interference reduced tumorigenesis in vivo. Analysis of RaLP in melanoma signal transduction pathways revealed that (a) when ectopically expressed in RaLP-negative melanocytes and nonmetastatic melanoma cells, it functions as a substrate of activated insulin-like growth factor-1 and epidermal growth factor receptors and increases Ras/mitogen-activated protein kinase (MAPK) signaling and cell migration, whereas (b) its silencing in RaLP-positive melanoma cells abrogates cell migration in vitro, without affecting MAPK signaling, suggesting that RaLP activates both Ras-dependent and Ras-independent migratory pathways in melanomas. These findings indicate that RaLP is a specific marker of metastatic melanomas, a critical determinant in the acquisition of the migratory phenotype by melanoma cells, and a potential target for novel anti-melanoma therapeutic strategies.
Collapse
Affiliation(s)
- Ernesta Fagiani
- Department of Experimental Oncology, European Institute of Oncology, Universita' degli Studi di Milano, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Peviani M, Cheroni C, Troglio F, Quarto M, Pelicci G, Bendotti C. Lack of changes in the PI3K/AKT survival pathway in the spinal cord motor neurons of a mouse model of familial amyotrophic lateral sclerosis. Mol Cell Neurosci 2007; 34:592-602. [PMID: 17303436 DOI: 10.1016/j.mcn.2007.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2006] [Revised: 12/29/2006] [Accepted: 01/03/2007] [Indexed: 10/23/2022] Open
Abstract
The vulnerability of motor neurons in transgenic SOD1G93A mice, a model of familial amyotrophic lateral sclerosis (ALS), may depend on the failure of these cells to activate survival mechanisms in response to the toxic mutant SOD1. To test this we investigated whether defects in the PI3K/Akt pathway, a survival signal, and of its neuron-specific activator, Rai, were important for motor neuron degeneration in these mice. No substantial changes were found in the levels of Rai, PI3K(p85) or phosphorylated Akt (P-Akt) in the ventral horn of spinal cord of SOD1G93A mice during disease progression. P-Akt immunoreactivity was the same in degenerating and healthy motor neurons. Rai ablation in SOD1G93A mice slightly accelerated the motor dysfunction without affecting their life span. Thus, motor neurons in SOD1G93A mice do not lose the pro-survival PI3K/Akt signal nor increase it in order to suppress the cell death mechanisms.
Collapse
Affiliation(s)
- M Peviani
- Lab. Molecular Neurobiology, Dept. Neuroscience, Istituto di Ricerche Farmacologiche "Mario Negri", Via Eritrea 62, 20157 Milano, Italy
| | | | | | | | | | | |
Collapse
|