1
|
Zheng D, Wei Z, Zhang C, Liu W, Gong C, Wu F, Guo W. ZNF692 promotes osteosarcoma cell proliferation, migration, and invasion through TNK2-mediated activation of the MEK/ERK pathway. Biol Direct 2024; 19:28. [PMID: 38650011 PMCID: PMC11034355 DOI: 10.1186/s13062-024-00472-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Osteosarcoma is a diverse and aggressive bone tumor. Driver genes regulating osteosarcoma initiation and progression remains incompletely defined. Zinc finger protein 692 (ZNF692), a kind of Krüppel C2H2 zinc finger transcription factor, exhibited abnormal expression in different types of malignancies and showed a correlation with the clinical prognosis of patients as well as the aggressive characteristics of cancer cells. Nevertheless, its specific role in osteosarcoma is still not well understood. METHODS We investigated the dysregulation and clinical significance of ZNF692 in osteosarcoma through bioinformatic method and experimental validation. A range of in vitro assays, including CCK-8, colony formation, EdU incorporation, wound healing, and transwell invasion tests, were conducted to assess the impact of ZNF692 on cell proliferation, migration, and invasion in osteosarcoma. A xenograft mouse model was established to evaluate the effect of ZNF692 on tumor growth in vivo. Western blot assay was used to measure the protein levels of MEK1/2, P-MEK1/2, ERK1/2, and P-ERK1/2 in cells that had been genetically modified to either reduce or increase the expression of ZNF692. The relationship between ZNF692 and tyrosine kinase non-receptor 2 (TNK2) were validated by qRT-PCR, chromatin immunoprecipitation and luciferase reporter assays. RESULTS Expression of ZNF692 was increased in both human osteosarcoma tissues and cell lines. Furthermore, the expression of ZNF692 served as an independent predictive biomarker in osteosarcoma. The results of the survival analysis indicated that increased expression of ZNF692 was associated with worse outcome. Downregulation of ZNF692 inhibits the proliferation, migration, and invasion of osteosarcoma cells, whereas upregulation of ZNF692 has the opposite impact. Western blot assay indicates that reducing ZNF692 decreases phosphorylation of MEK1/2 and ERK1/2, whereas increasing ZNF692 expression enhances their phosphorylation. U0126, a potent inhibitor specifically targeting the MEK/ERK signaling pathway, partially counteracts the impact of ZNF692 overexpression on the proliferation, migration, and invasion of osteosarcoma cells. In addition, ZNF692 specifically interacts with the promoter region of TNK2 and stimulates the transcription of TNK2 in osteosarcoma cells. Forcing the expression of TNK2 weakens the inhibitory impact of ZNF692 knockdown on P-MEK1/2 and P-ERK1/2. Similarly, partly inhibiting TNK2 counteracts the enhancing impact of ZNF692 overexpression on the phosphorylation of MEK1/2 and ERK1/2. Functional tests demonstrate that the suppressive effects of ZNF692 knockdown on cell proliferation, migration, and invasion are greatly reduced when TNK2 is overexpressed. In contrast, the reduction of TNK2 hinders the ability of ZNF692 overexpression to enhance cell proliferation, migration, and invasion. CONCLUSION ZNF692 promotes the proliferation, migration, and invasion of osteosarcoma cells via the TNK2-dependent stimulation of the MEK/ERK signaling pathway. The ZNF692-TNK2 axis might potentially function as a possible predictive biomarker and a promising target for novel therapeutics in osteosarcoma.
Collapse
Affiliation(s)
- Di Zheng
- Department of Orthopedics, Renmin Hospital of Wuhan University, 430060, Wuhan, China
| | - Zhun Wei
- Department of Orthopedics, Renmin Hospital of Wuhan University, 430060, Wuhan, China
| | - Chong Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, 430060, Wuhan, China
| | - Wenda Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, 430060, Wuhan, China
| | - Changtian Gong
- Department of Orthopedics, Renmin Hospital of Wuhan University, 430060, Wuhan, China
| | - Fei Wu
- Department of Orthopedics, Renmin Hospital of Wuhan University, 430060, Wuhan, China.
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, 430060, Wuhan, China.
| |
Collapse
|
2
|
Geckinli B, Turkyilmaz A, Alavanda C, Sager G, Arslan Ates E, Soylemez MA, Arman A. Novel, homozygous RAB3GAP1 c.2606 + 1G>A, p.Glu830ValfsTer9 variant and chromosome 3q29 duplication in a Turkish individual with Warburg micro syndrome. Clin Dysmorphol 2023; 32:55-61. [PMID: 36876345 DOI: 10.1097/mcd.0000000000000454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Warburg micro syndrome (WARBM) is a rare, autosomal recessive, neurodevelopmental disorder characterized by microcephaly, cortical dysplasia, corpus callosum hypoplasia, congenital hypotonia leading to subsequent spastic quadriplegia, severe developmental delay and hypogenitalism. Ophthalmologic findings that may affect any ocular segment including characteristic, small, atonic pupils. WARBM is known to be caused by biallelic, pathogenic variants in at least five genes although additional genetic loci may exist. The RAB3GAP1 c.748 + 1G>A, p.Asp250CysfsTer24 founder variant has been described in families of Turkish ancestry. We report the clinical and molecular findings in three, unrelated, Turkish families with WARBM. A novel c.974-2A>G variant causing WARBM in three siblings of Turkish descent was found. Functional studies of the novel, c.2606 + 1G>A variant in patients' mRNA revealed skipping of exon 22 which results in a premature stop codon in exon 23. However, the clinical consequences of this variant are blended given that the individual also had a maternally inherited chromosome 3q29 microduplication.
Collapse
Affiliation(s)
- Bilge Geckinli
- Department of Medical Genetics, Marmara University School of Medicine, Istanbul
| | - Ayberk Turkyilmaz
- Department of Medical Genetics, Marmara University School of Medicine, Istanbul
- Department of Medical Genetics, Karadeniz Technical University School of Medicine, Trabzon
| | - Ceren Alavanda
- Department of Medical Genetics, Marmara University School of Medicine, Istanbul
| | - Gunes Sager
- Department of Pediatric Neurology, Marmara University School of Medicine
- Pediatric Neurology, Kartal Dr. Lutfi Kirdar City Hospital
| | - Esra Arslan Ates
- Department of Medical Genetics, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Mehmet Ali Soylemez
- Department of Medical Genetics, Marmara University School of Medicine, Istanbul
| | - Ahmet Arman
- Department of Medical Genetics, Marmara University School of Medicine, Istanbul
| |
Collapse
|
3
|
Kan Y, Paung Y, Seeliger MA, Miller WT. Domain Architecture of the Nonreceptor Tyrosine Kinase Ack1. Cells 2023; 12:900. [PMID: 36980241 PMCID: PMC10047419 DOI: 10.3390/cells12060900] [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: 01/30/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
The nonreceptor tyrosine kinase (NRTK) Ack1 comprises a distinct arrangement of non-catalytic modules. Its SH3 domain has a C-terminal to the kinase domain (SH1), in contrast to the typical SH3-SH2-SH1 layout in NRTKs. The Ack1 is the only protein that shares a region of high homology to the tumor suppressor protein Mig6, a modulator of EGFR. The vertebrate Acks make up the only tyrosine kinase (TK) family known to carry a UBA domain. The GTPase binding and SAM domains are also uncommon in the NRTKs. In addition to being a downstream effector of receptor tyrosine kinases (RTKs) and integrins, Ack1 can act as an epigenetic regulator, modulate the degradation of the epidermal growth factor receptor (EGFR), confer drug resistance, and mediate the progression of hormone-sensitive tumors. In this review, we discuss the domain architecture of Ack1 in relation to other protein kinases that possess such defined regulatory domains.
Collapse
Affiliation(s)
- Yagmur Kan
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - YiTing Paung
- Department of Pharmacology, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - Markus A. Seeliger
- Department of Pharmacology, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
| | - W. Todd Miller
- Department of Physiology and Biophysics, School of Medicine, Stony Brook University, Stony Brook, NY 11794-8661, USA
- Department of Veterans Affairs Medical Center, Northport, NY 11768-2200, USA
| |
Collapse
|
4
|
Nourse JB, Russell SN, Moniz NA, Peter K, Seyfarth LM, Scott M, Park HA, Caldwell KA, Caldwell GA. Integrated regulation of dopaminergic and epigenetic effectors of neuroprotection in Parkinson's disease models. Proc Natl Acad Sci U S A 2023; 120:e2210712120. [PMID: 36745808 PMCID: PMC9963946 DOI: 10.1073/pnas.2210712120] [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: 06/22/2022] [Accepted: 01/05/2023] [Indexed: 02/08/2023] Open
Abstract
Whole-exome sequencing of Parkinson's disease (PD) patient DNA identified single-nucleotide polymorphisms (SNPs) in the tyrosine nonreceptor kinase-2 (TNK2) gene. Although this kinase had a previously demonstrated activity in preventing the endocytosis of the dopamine reuptake transporter (DAT), a causal role for TNK2-associated dysfunction in PD remains unresolved. We postulated the dopaminergic neurodegeneration resulting from patient-associated variants in TNK2 were a consequence of aberrant or prolonged TNK2 overactivity, the latter being a failure in TNK2 degradation by an E3 ubiquitin ligase, neuronal precursor cell-expressed developmentally down-regulated-4 (NEDD4). Interestingly, systemic RNA interference protein-3 (SID-3) is the sole TNK2 ortholog in the nematode Caenorhabditis elegans, where it is an established effector of epigenetic gene silencing mediated through the dsRNA-transporter, SID-1. We hypothesized that TNK2/SID-3 represents a node of integrated dopaminergic and epigenetic signaling essential to neuronal homeostasis. Use of a TNK2 inhibitor (AIM-100) or a NEDD4 activator [N-aryl benzimidazole 2 (NAB2)] in bioassays for either dopamine- or dsRNA-uptake into worm dopaminergic neurons revealed that sid-3 mutants displayed robust neuroprotection from 6-hydroxydopamine (6-OHDA) exposures, as did AIM-100 or NAB2-treated wild-type animals. Furthermore, NEDD4 activation by NAB2 in rat primary neurons correlated to a reduction in TNK2 levels and the attenuation of 6-OHDA neurotoxicity. CRISPR-edited nematodes engineered to endogenously express SID-3 variants analogous to TNK2 PD-associated SNPs exhibited enhanced susceptibility to dopaminergic neurodegeneration and circumvented the RNAi resistance characteristic of SID-3 dysfunction. This research exemplifies a molecular etiology for PD whereby dopaminergic and epigenetic signaling are coordinately regulated to confer susceptibility or resilience to neurodegeneration.
Collapse
Affiliation(s)
- J. Brucker Nourse
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
| | - Shannon N. Russell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
| | - Nathan A. Moniz
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
| | - Kylie Peter
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
| | - Lena M. Seyfarth
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
| | - Madison Scott
- Department of Human Nutrition and Hospitality Management, The University of Alabama, Tuscaloosa, AL35487
| | - Han-A Park
- Department of Human Nutrition and Hospitality Management, The University of Alabama, Tuscaloosa, AL35487
- Alabama Research Institute on Aging, The University of Alabama, Tuscaloosa, AL35487
- Center for Convergent Bioscience and Medicine, The University of Alabama, Tuscaloosa, AL35487
| | - Kim A. Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
- Alabama Research Institute on Aging, The University of Alabama, Tuscaloosa, AL35487
- Center for Convergent Bioscience and Medicine, The University of Alabama, Tuscaloosa, AL35487
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center of Excellence for Research in the Basic Biology of Aging, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL35294
| | - Guy A. Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL35487
- Center for Convergent Bioscience and Medicine, The University of Alabama, Tuscaloosa, AL35487
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center of Excellence for Research in the Basic Biology of Aging, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL35294
| |
Collapse
|
5
|
Long KLP, Muroy SE, Sorooshyari SK, Ko MJ, Jaques Y, Sudmant P, Kaufer D. Transcriptomic profiles of stress susceptibility and resilience in the amygdala and hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527777. [PMID: 36798395 PMCID: PMC9934702 DOI: 10.1101/2023.02.08.527777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
A single, severe episode of stress can bring about myriad responses amongst individuals, ranging from cognitive enhancement to debilitating and persistent anxiety; however, the biological mechanisms that contribute to resilience versus susceptibility to stress are poorly understood. The dentate gyrus (DG) of the hippocampus and the basolateral nucleus of the amygdala (BLA) are key limbic regions that are susceptible to the neural and hormonal effects of stress. Previous work has also shown that these regions contribute to individual variability in stress responses; however, the molecular mechanisms underlying the role of these regions in susceptibility and resilience are unknown. In this study, we profiled the transcriptomic signatures of the DG and BLA of rats with divergent behavioral outcomes after a single, severe stressor. We subjected rats to three hours of immobilization with exposure to fox urine and conducted a behavioral battery one week after stress to identify animals that showed persistent, high anxiety-like behavior. We then conducted bulk RNA sequencing of the DG and BLA from susceptible, resilient, and unexposed control rats. Differential gene expression analyses revealed that the molecular signatures separating each of the three groups were distinct and non-overlapping between the DG and BLA. In the amygdala, key genes associated with insulin and hormonal signaling corresponded with vulnerability. Specifically, Inhbb, Rab31 , and Ncoa3 were upregulated in the amygdala of stress-susceptible animals compared to resilient animals. In the hippocampus, increased expression of Cartpt - which encodes a key neuropeptide involved in reward, reinforcement, and stress responses - was strongly correlated with vulnerability to anxiety-like behavior. However, few other genes distinguished stress-susceptible animals from control animals, while a larger number of genes separated stress-resilient animals from control and stress-susceptible animals. Of these, Rnf112, Tbx19 , and UBALD1 distinguished resilient animals from both control and susceptible animals and were downregulated in resilience, suggesting that an active molecular response in the hippocampus facilitates protection from the long-term consequences of severe stress. These results provide novel insight into the mechanisms that bring about individual variability in the behavioral responses to stress and provide new targets for the advancement of therapies for stress-induced neuropsychiatric disorders.
Collapse
|
6
|
Wang A, Pei J, Shuai W, Lin C, Feng L, Wang Y, Lin F, Ouyang L, Wang G. Small Molecules Targeting Activated Cdc42-Associated Kinase 1 (ACK1/TNK2) for the Treatment of Cancers. J Med Chem 2021; 64:16328-16348. [PMID: 34735773 DOI: 10.1021/acs.jmedchem.1c01030] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Activated Cdc42-associated kinase 1 (ACK1/TNK2) is a nonreceptor tyrosine kinase with a unique structure. It not only can act as an activated transmembrane effector of receptor tyrosine kinases (RTKs) to transmit various RTK signals but also can play a corresponding role in epigenetic regulation. A number of studies have shown that ACK1 is a carcinogenic factor. Blockage of ACK1 has been proven to be able to inhibit cancer cell survival, proliferation, migration, and radiation resistance. Thus, ACK1 is a promising potential antitumor target. To date, despite many efforts to develop ACK1 inhibitors, no specific small molecule inhibitors have entered clinical trials. This Perspective provides an overview of the structural features, biological functions, and association with diseases of ACK1 and in vitro and in vivo activities, selectivity, and therapeutic potential of small molecule ACK1 inhibitors with different chemotypes.
Collapse
Affiliation(s)
- Aoxue Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Junping Pei
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Wen Shuai
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Lu Feng
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Feng Lin
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China.,Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
New avenues in molecular genetics for the diagnosis and application of therapeutics to the epilepsies. Epilepsy Behav 2021; 121:106428. [PMID: 31400936 DOI: 10.1016/j.yebeh.2019.07.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/14/2019] [Accepted: 07/06/2019] [Indexed: 11/22/2022]
Abstract
Genetic epidemiology studies have shown that most epilepsies involve some genetic cause. In addition, twin studies have helped strengthen the hypothesis that in most patients with epilepsy, a complex inheritance is involved. More recently, with the development of high-density single-nucleotide polymorphism (SNP) microarrays and next-generation sequencing (NGS) technologies, the discovery of genes related to the epilepsies has accelerated tremendously. Especially, the use of whole exome sequencing (WES) has had a considerable impact on the identification of rare genetic variants with large effect sizes, including inherited or de novo mutations in severe forms of childhood epilepsies. The identification of pathogenic variants in patients with these childhood epilepsies provides many benefits for patients and families, such as the confirmation of the genetic nature of the diseases. This process will allow for better genetic counseling, more accurate therapy decisions, and a significant positive emotional impact. However, to study the genetic component of the more common forms of epilepsy, the use of high-density SNP arrays in genome-wide association studies (GWAS) seems to be the strategy of choice. As such, researchers can identify loci containing genetic variants associated with the common forms of epilepsy. The knowledge generated over the past two decades about the effects of the mutations that cause the monogenic epilepsy is tremendous; however, the scientific community is just starting to apply this information in order to generate better target treatments.
Collapse
|
8
|
Brandao R, Kwa MQ, Yarden Y, Brakebusch C. ACK1 is dispensable for development, skin tumor formation, and breast cancer cell proliferation. FEBS Open Bio 2021; 11:1579-1592. [PMID: 33730447 PMCID: PMC8167857 DOI: 10.1002/2211-5463.13149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/16/2021] [Indexed: 11/29/2022] Open
Abstract
Activated Cdc42‐associated kinase 1 (ACK1), a widely expressed nonreceptor tyrosine kinase, is often amplified in cancer and has been shown to interact with Cell division cycle 42 (Cdc42), Epidermal growth factor receptor (EGFR), and several other cancer‐relevant molecules, suggesting a possible role for ACK1 in development and tumor formation. To directly address this scenario, we generated mice lacking a functional ACK1 gene (ACK1 ko) using CRISPR genome editing. ACK1 ko mice developed normally, displayed no obvious defect in tissue maintenance, and were fertile. Primary ACK1‐null keratinocytes showed normal phosphorylation of EGFR, but a tendency toward reduced activation of AKT serine/threonine kinase 1 (Akt) and Mitogen‐activated protein kinase 1 (Erk). DMBA/TPA‐induced skin tumor formation did not reveal significant differences between ACK1 ko and control mice. Deletion of the ACK1 gene in the breast cancer cell lines MDA‐MB‐231, 67NR, MCF7, 4T1, and T47D caused no differences in growth. Furthermore, EGF‐induced phosphorylation kinetics of Erk, Akt, and p130Cas were not detectably altered in T47D cells by the loss of ACK1. Finally, loss of ACK1 in MDA‐MB‐231 and T47D breast cancer cells had a very limited or no effect on directed cell migration. These data do not support a major role for ACK1 in Cdc42 and EGFR signaling, development, or tumor formation.
Collapse
Affiliation(s)
- Rafael Brandao
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
| | - Mei Qi Kwa
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
| | | | - Cord Brakebusch
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
| |
Collapse
|
9
|
Chirita Emandi A, Dobrescu AI, Doros G, Hyon C, Miclea D, Popoiu C, Puiu M, Arghirescu S. A Novel 3q29 Deletion in Association With Developmental Delay and Heart Malformation-Case Report With Literature Review. Front Pediatr 2019; 7:270. [PMID: 31338352 PMCID: PMC6628938 DOI: 10.3389/fped.2019.00270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/14/2019] [Indexed: 01/16/2023] Open
Abstract
3q29 deletion syndrome is a rare disorder, causing a complex phenotype. Clinical features are variable and relatively non-specific. Our report aims to present an atypical, de novo deletion in chromosome band 3q29 in a preschool boy, first child of healthy non-consanguineous parents, presenting a particular phenotype (microcephaly, "full moon" face, flattened facial profile, large ears, auricular polyp, and dental dystrophies), motor and cognitive delay, characteristics of autism spectrum disorder and aggressive behavior. He also presented intrauterine growth restriction (birth weight 2,400 g) and a ventricular septal defect. SNP Array revealed a 962 kb copy number loss, on the chromosome 3q29 band (195519857-196482211), consistent with 3q29 microdeletion syndrome. FISH analysis using a RP11-252K11 probe confirmed the deletion in the proband, which was not present in the parents. Although the patient's deletion is relatively small, it partly overlaps the canonical 3q29 deletion (defined between TFRC and DLG1 gene) and extends upstream, associating a different facial phenotype compared to the classic 3q29 deletion, nonetheless showing a similar psychiatric disorder. This deletion is different from the canonical region, as it does not include the PAK2 and DLG1 genes, considered as candidates for causing intellectual disability. Thus, narrowing the genotype-phenotype correlation for the 3q29 band, FBX045 is suggested as a candidate gene for the neuropsychiatric phenotype.
Collapse
Affiliation(s)
- Adela Chirita Emandi
- Discipline of Genetics, Victor Babeș University of Medicine and Pharmacy, Timișoara, Romania.,"Louis Turcanu" Clinical Emergency Hospital for Children, Timișoara, Romania
| | - Andreea Iulia Dobrescu
- Discipline of Genetics, Victor Babeș University of Medicine and Pharmacy, Timișoara, Romania.,"Louis Turcanu" Clinical Emergency Hospital for Children, Timișoara, Romania
| | - Gabriela Doros
- "Louis Turcanu" Clinical Emergency Hospital for Children, Timișoara, Romania.,IIIrd Pediatric Clinic, Pediatric Cardiology, Victor Babeș University of Medicine and Pharmacy, Timișoara, Romania
| | - Capucine Hyon
- Département de Génétique Médicale, AP-HP, GHUEP, Hôpital Armand Trousseau, Paris, France.,INSERM, UMRS 933, Hôpital Armand Trousseau, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | - Diana Miclea
- Genetics Department Cluj-Napoca, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Calin Popoiu
- "Louis Turcanu" Clinical Emergency Hospital for Children, Timișoara, Romania.,Discipline of Pediatric Surgery, Victor Babeș University of Medicine and Pharmacy, Timișoara, Romania
| | - Maria Puiu
- Discipline of Genetics, Victor Babeș University of Medicine and Pharmacy, Timișoara, Romania.,"Louis Turcanu" Clinical Emergency Hospital for Children, Timișoara, Romania
| | - Smaranda Arghirescu
- "Louis Turcanu" Clinical Emergency Hospital for Children, Timișoara, Romania.,IIIrd Pediatric Clinic, Victor Babeș University of Medicine and Pharmacy, Timișoara, Romania
| |
Collapse
|
10
|
Tassano E, Uccella S, Giacomini T, Severino M, Siri L, Gherzi M, Celle ME, Porta S, Gimelli G, Ronchetto P. 3q29 microduplication syndrome: Description of two new cases and delineation of the minimal critical region. Eur J Med Genet 2018; 61:428-433. [PMID: 29501613 DOI: 10.1016/j.ejmg.2018.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 01/31/2018] [Accepted: 02/26/2018] [Indexed: 12/28/2022]
Abstract
Heterogeneous clinical and neuropsychological features, such as intellectual disability, developmental and language delay, hypotonia, and, to a lesser extent, microcephaly that is present in about the half of the reported patients, characterize the 3q29 microduplication syndrome with usually a milder phenotype compared with the corresponding 3q29 microdeletion syndrome. The duplications described so far range from 2.3 Mb to 1.6 Mb, spanning from TFRC to BDH1 genes. Here we report on two patients with overlapping interstitial duplications of the 3q29 region differing in size. Patient 1 harboured a common-seized 3q29 microduplication spanning ∼1.6 Mb, while patient 2 carried a very small 3q29 microduplication of 448.8 Kb encompassing only two genes, DLG1 and BDH1. Both patients presented clinical characteristics similar to those reported in the literature in 3q29 microduplication syndrome. Interestingly, heterotopic gray matter nodules were found along the right lateral ventricle on brain MRI in patient 1, thus expanding the neuroradiological phenotype in 3q29 microduplication syndrome, while patient 2 allowed us to define with more precision the smallest region of overlap (SRO). Gene content analysis of the duplicated region suggests that gain-of-dosage of DLG1 and BDH1 may be a good candidate for the main clinical features of this syndrome.
Collapse
Affiliation(s)
- Elisa Tassano
- Laboratory of Cytogenetics, Istituto Giannina Gaslini, Genoa, Italy.
| | - Sara Uccella
- Unit of Child Neuropsychiatry, Istituto Giannina Gaslini, University of Genova, Genoa, Italy
| | - Thea Giacomini
- Unit of Child Neuropsychiatry, Istituto Giannina Gaslini, University of Genova, Genoa, Italy
| | | | - Laura Siri
- "La Nostra Famiglia" Association, Varazze (Sv) - Scientific Institute E. Medea, Lecco, Bosisio Parini, Italy
| | - Marcella Gherzi
- Unit of Child Neuropsychiatry, Istituto Giannina Gaslini, University of Genova, Genoa, Italy
| | - Maria Elena Celle
- Unit of Child Neuropsychiatry, Head Neck and Neuroscience Department, Istituto Giannina Gaslini, Genoa, Italy
| | - Simona Porta
- Laboratory of Cytogenetics, Istituto Giannina Gaslini, Genoa, Italy
| | - Giorgio Gimelli
- Laboratory of Cytogenetics, Istituto Giannina Gaslini, Genoa, Italy
| | | |
Collapse
|
11
|
Mao X, Qian S, Peng J, Cui W, Lu G, Zhan Y. Two patients with TNK2 mutations and late onset infantile spasm. Ann Neurol 2017; 81:161. [PMID: 27977884 DOI: 10.1002/ana.24838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Xiao Mao
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.,State Key Laboratory of Medical Genetics of China, Changsha
| | - Shuyi Qian
- Department of Nephrology, Hunan Provincial People's Hospital, Changsha
| | - Jinxin Peng
- Department of Neurology, Xiangya Hospital of Central South University, Changsha
| | - Weiren Cui
- WuXi NextCode Genomics Co, Shanghai, China
| | - Gui Lu
- WuXi NextCode Genomics Co, Shanghai, China
| | - Yajing Zhan
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
12
|
Depondt C, Heinzen EL, Goldstein DB. Reply. Ann Neurol 2016; 81:161-162. [PMID: 27977874 DOI: 10.1002/ana.24837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Chantal Depondt
- Department of Neurology, Erasmus Hospital, Free University of Brussels, Brussels, Belgium
| | - Erin L Heinzen
- Department of Pathology and Cell Biology, Columbia University, New York, NY
| | - David B Goldstein
- Department of Genetics, Institute for Genomic Medicine, Columbia University Medical Center, New York, NY
| |
Collapse
|
13
|
Zeng QS, Xie BH, Xie YK, Wang XN. Activated Cdc42 kinase 1 and hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2016; 24:3853-3859. [DOI: 10.11569/wcjd.v24.i27.3853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Primary liver cancer includes hepatocellular carcinoma (HCC) and cholangiocellular carcinoma. The incidence of HCC is different between countries and regions. As one of the common malignant tumors in China, HCC has high mortality and is the second most common cause of cancer-related death. Elucidating the molecular mechanism of HCC pathogenesis is important for the diagnosis and treatment of liver cancer in China. The expression of activated Cdc42 kinase 1 (ACK1) has been found in a variety of cancers, and ACK1 participates in the occurrence and development of cancers. However, there are currently few studies about the relationship between ACK1 protein and HCC. This paper reviews the structure characteristics and biological function of ACK1 as well as its relationship with invasion and metastasis of HCC.
Collapse
|
14
|
Farlow JL, Robak LA, Hetrick K, Bowling K, Boerwinkle E, Coban-Akdemir ZH, Gambin T, Gibbs RA, Gu S, Jain P, Jankovic J, Jhangiani S, Kaw K, Lai D, Lin H, Ling H, Liu Y, Lupski JR, Muzny D, Porter P, Pugh E, White J, Doheny K, Myers RM, Shulman JM, Foroud T. Whole-Exome Sequencing in Familial Parkinson Disease. JAMA Neurol 2016; 73:68-75. [PMID: 26595808 PMCID: PMC4946647 DOI: 10.1001/jamaneurol.2015.3266] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IMPORTANCE Parkinson disease (PD) is a progressive neurodegenerative disease for which susceptibility is linked to genetic and environmental risk factors. OBJECTIVE To identify genetic variants contributing to disease risk in familial PD. DESIGN, SETTING, AND PARTICIPANTS A 2-stage study design that included a discovery cohort of families with PD and a replication cohort of familial probands was used. In the discovery cohort, rare exonic variants that segregated in multiple affected individuals in a family and were predicted to be conserved or damaging were retained. Genes with retained variants were prioritized if expressed in the brain and located within PD-relevant pathways. Genes in which prioritized variants were observed in at least 4 families were selected as candidate genes for replication in the replication cohort. The setting was among individuals with familial PD enrolled from academic movement disorder specialty clinics across the United States. All participants had a family history of PD. MAIN OUTCOMES AND MEASURES Identification of genes containing rare, likely deleterious, genetic variants in individuals with familial PD using a 2-stage exome sequencing study design. RESULTS The 93 individuals from 32 families in the discovery cohort (49.5% [46 of 93] female) had a mean (SD) age at onset of 61.8 (10.0) years. The 49 individuals with familial PD in the replication cohort (32.6% [16 of 49] female) had a mean (SD) age at onset of 50.1 (15.7) years. Discovery cohort recruitment dates were 1999 to 2009, and replication cohort recruitment dates were 2003 to 2014. Data analysis dates were 2011 to 2015. Three genes containing a total of 13 rare and potentially damaging variants were prioritized in the discovery cohort. Two of these genes (TNK2 and TNR) also had rare variants that were predicted to be damaging in the replication cohort. All 9 variants identified in the 2 replicated genes in 12 families across the discovery and replication cohorts were confirmed via Sanger sequencing. CONCLUSIONS AND RELEVANCE TNK2 and TNR harbored rare, likely deleterious, variants in individuals having familial PD, with similar findings in an independent cohort. To our knowledge, these genes have not been previously associated with PD, although they have been linked to critical neuronal functions. Further studies are required to confirm a potential role for these genes in the pathogenesis of PD.
Collapse
Affiliation(s)
- Janice L Farlow
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis
| | - Laurie A Robak
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas3Department of Pediatrics, Baylor College of Medicine, Houston, Texas4Department of Pediatrics, Texas Children's Hospital, Houston5Jan and Dan Duncan Neurological Resear
| | - Kurt Hetrick
- Center for Inherited Disease Research, The Johns Hopkins University, Baltimore, Maryland
| | - Kevin Bowling
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | - Eric Boerwinkle
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas9Human Genetics Center, University of Texas Health Science Center, Houston
| | | | - Tomasz Gambin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas
| | - Shen Gu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Preti Jain
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama10Department of Pediatrics, Columbia University Medical Center, New York, New York
| | - Joseph Jankovic
- Department of Neurology, Baylor College of Medicine, Houston, Texas
| | - Shalini Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas
| | - Kaveeta Kaw
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas5Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston
| | - Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis
| | - Hai Lin
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indianapolis
| | - Hua Ling
- Center for Inherited Disease Research, The Johns Hopkins University, Baltimore, Maryland
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas3Department of Pediatrics, Baylor College of Medicine, Houston, Texas4Department of Pediatrics, Texas Children's Hospital, Houston8Human Genome Sequencing Center, Baylor
| | - Donna Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas
| | - Paula Porter
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas5Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston
| | - Elizabeth Pugh
- Center for Inherited Disease Research, The Johns Hopkins University, Baltimore, Maryland
| | - Janson White
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Kimberly Doheny
- Center for Inherited Disease Research, The Johns Hopkins University, Baltimore, Maryland
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama
| | - Joshua M Shulman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas5Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston11Department of Neurology, Baylor College of Medicine, Houston, Texas13Department
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis
| |
Collapse
|
15
|
Hardies K, Weckhuysen S, De Jonghe P, Suls A. Lessons learned from gene identification studies in Mendelian epilepsy disorders. Eur J Hum Genet 2015; 24:961-7. [PMID: 26603999 DOI: 10.1038/ejhg.2015.251] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 10/05/2015] [Accepted: 10/29/2015] [Indexed: 01/23/2023] Open
Abstract
Next-generation sequencing (NGS) technologies are now routinely used for gene identification in Mendelian disorders. Setting up cost-efficient NGS projects and managing the large amount of variants remains, however, a challenging job. Here we provide insights in the decision-making processes before and after the use of NGS in gene identification studies. Genetic factors are thought to have a role in ~70% of all epilepsies, and a variety of inheritance patterns have been described for seizure-associated gene defects. We therefore chose epilepsy as disease model and selected 35 NGS studies that focused on patients with a Mendelian epilepsy disorder. The strategies used for gene identification and their respective outcomes were reviewed. High-throughput NGS strategies have led to the identification of several new epilepsy-causing genes, enlarging our knowledge on both known and novel pathomechanisms. NGS findings have furthermore extended the awareness of phenotypical and genetic heterogeneity. By discussing recent studies we illustrate: (I) the power of NGS for gene identification in Mendelian disorders, (II) the accelerating pace in which this field evolves, and (III) the considerations that have to be made when performing NGS studies. Nonetheless, the enormous rise in gene discovery over the last decade, many patients and families included in gene identification studies still remain without a molecular diagnosis; hence, further genetic research is warranted. On the basis of successful NGS studies in epilepsy, we discuss general approaches to guide human geneticists and clinicians in setting up cost-efficient gene identification NGS studies.
Collapse
Affiliation(s)
- Katia Hardies
- VIB-Department of Molecular Genetics, Neurogenetics Group, University of Antwerp, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Sarah Weckhuysen
- VIB-Department of Molecular Genetics, Neurogenetics Group, University of Antwerp, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Peter De Jonghe
- VIB-Department of Molecular Genetics, Neurogenetics Group, University of Antwerp, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Division of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | - Arvid Suls
- VIB-Department of Molecular Genetics, Neurogenetics Group, University of Antwerp, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
16
|
Gonsales MC, Montenegro MA, Soler CV, Coan AC, Guerreiro MM, Lopes-Cendes I. Recent developments in the genetics of childhood epileptic encephalopathies: impact in clinical practice. ARQUIVOS DE NEURO-PSIQUIATRIA 2015; 73:946-58. [PMID: 26517219 DOI: 10.1590/0004-282x20150122] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/20/2015] [Indexed: 01/03/2023]
Abstract
Recent advances in molecular genetics led to the discovery of several genes for childhood epileptic encephalopathies (CEEs). As the knowledge about the genes associated with this group of disorders develops, it becomes evident that CEEs present a number of specific genetic characteristics, which will influence the use of molecular testing for clinical purposes. Among these, there are the presence of marked genetic heterogeneity and the high frequency of de novo mutations. Therefore, the main objectives of this review paper are to present and discuss current knowledge regarding i) new genetic findings in CEEs, ii) phenotype-genotype correlations in different forms of CEEs; and, most importantly, iii) the impact of these new findings in clinical practice. Accompanying this text we have included a comprehensive table, containing the list of genes currently known to be involved in the etiology of CEEs.
Collapse
Affiliation(s)
- Marina C Gonsales
- Instituto Brasileiro de Neurociências e Neurotecnologia, Faculdade de Ciências Médicas, Universidade de Campinas, Campinas, SP, Brazil
| | - Maria Augusta Montenegro
- Instituto Brasileiro de Neurociências e Neurotecnologia, Faculdade de Ciências Médicas, Universidade de Campinas, Campinas, SP, Brazil
| | - Camila V Soler
- Instituto Brasileiro de Neurociências e Neurotecnologia, Faculdade de Ciências Médicas, Universidade de Campinas, Campinas, SP, Brazil
| | - Ana Carolina Coan
- Instituto Brasileiro de Neurociências e Neurotecnologia, Faculdade de Ciências Médicas, Universidade de Campinas, Campinas, SP, Brazil
| | - Marilisa M Guerreiro
- Instituto Brasileiro de Neurociências e Neurotecnologia, Faculdade de Ciências Médicas, Universidade de Campinas, Campinas, SP, Brazil
| | - Iscia Lopes-Cendes
- Instituto Brasileiro de Neurociências e Neurotecnologia, Faculdade de Ciências Médicas, Universidade de Campinas, Campinas, SP, Brazil
| |
Collapse
|
17
|
Lesca G, Depienne C. Epilepsy genetics: the ongoing revolution. Rev Neurol (Paris) 2015; 171:539-57. [PMID: 26003806 DOI: 10.1016/j.neurol.2015.01.569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/24/2014] [Accepted: 01/20/2015] [Indexed: 01/04/2023]
Abstract
Epilepsies have long remained refractory to gene identification due to several obstacles, including a highly variable inter- and intrafamilial expressivity of the phenotypes, a high frequency of phenocopies, and a huge genetic heterogeneity. Recent technological breakthroughs, such as array comparative genomic hybridization and next generation sequencing, have been leading, in the past few years, to the identification of an increasing number of genomic regions and genes in which mutations or copy-number variations cause various epileptic disorders, revealing an enormous diversity of pathophysiological mechanisms. The field that has undergone the most striking revolution is that of epileptic encephalopathies, for which most of causing genes have been discovered since the year 2012. Some examples are the continuous spike-and-waves during slow-wave sleep and Landau-Kleffner syndromes for which the recent discovery of the role of GRIN2A mutations has finally confirmed the genetic bases. These new technologies begin to be used for diagnostic applications, and the main challenge now resides in the interpretation of the huge mass of variants detected by these methods. The identification of causative mutations in epilepsies provides definitive confirmation of the clinical diagnosis, allows accurate genetic counselling, and sometimes permits the development of new appropriate and specific antiepileptic therapies. Future challenges include the identification of the genetic or environmental factors that modify the epileptic phenotypes caused by mutations in a given gene and the understanding of the role of somatic mutations in sporadic epilepsies.
Collapse
Affiliation(s)
- G Lesca
- Service de génétique, groupement hospitalier Est, hospices civils de Lyon, 59, boulevard Pinel, 69677 Bron, France; Université Claude-Bernard Lyon 1, 43, boulevard du 11-Novembre-1918, 69100 Villeurbanne, France; CRNL, CNRS UMR 5292, Inserm U1028, bâtiment IMBL, 11, avenue Jean-Capelle, 69621 Villeurbanne cedex, France.
| | - C Depienne
- Département de génétique et cytogénétique, hôpital Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75651 Paris cedex 13, France; Sorbonne universités, UPMC université Paris 06, 4, place Jussieu, 75005 Paris, France; ICM, CNRS UMR 7225, Inserm U1127, 47, boulevard de l'Hôpital, 75651 Paris cedex 13, France
| |
Collapse
|
18
|
Boutry-Kryza N, Labalme A, Ville D, de Bellescize J, Touraine R, Prieur F, Dimassi S, Poulat AL, Till M, Rossi M, Bourel-Ponchel E, Delignières A, Le Moing AG, Rivier C, des Portes V, Edery P, Calender A, Sanlaville D, Lesca G. Molecular characterization of a cohort of 73 patients with infantile spasms syndrome. Eur J Med Genet 2014; 58:51-8. [PMID: 25497044 DOI: 10.1016/j.ejmg.2014.11.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 11/30/2014] [Indexed: 01/10/2023]
Abstract
Infantile Spasms syndrome (ISs) is a characterized by epileptic spasms occurring in clusters with an onset in the first year of life. West syndrome represents a subset of ISs that associates spasms in clusters, a hypsarrhythmia EEG pattern and a developmental arrest or regression. Aetiology of ISs is widely heterogeneous including many genetic causes. Many patients, however, remain without etiological diagnosis, which is critical for prognostic purpose and genetic counselling. In the present study, we performed genetic screening of 73 patients with different types of ISs by array-CGH and molecular analysis of 5 genes: CDKL5, STXBP1, KCNQ2, and GRIN2A, whose mutations cause different types of epileptic encephalopathies, including ISs, as well as MAGI2, which was suggested to be related to a subset of ISs. In total, we found a disease-causing mutation or CNV (Copy Number Variation) in 15% of the patients. These included 6 point mutations found in CDKL5 (n = 3) and STXBP1 (n = 3), 3 microdeletions (10 Mb in 2q24.3, 3.2 Mb in 5q14.3 including the region upstream to MEF2C, and 256 kb in 9q34 disrupting EHMT1), and 2 microduplications (671 kb in 2q24.3 encompassing SCN2A, and 11.93 Mb in Xq28). In addition, we discuss 3 CNVs as potential risk factors, including one 16p12.1 deletion, one intronic deletion of the NEDD4 gene, and one intronic deletion of CALN1 gene. The present findings highlight the efficacy of combined cytogenetic and targeted mutation screening to improve the diagnostic yield in patient with ISs.
Collapse
Affiliation(s)
- Nadia Boutry-Kryza
- Department of Molecular Genetics, Lyon University Hospital, Lyon, France; CRNL, CNRS UMR 5292, INSERM U1028, Lyon, France
| | - Audrey Labalme
- Department of Genetics, Lyon University Hospital, Lyon, France
| | - Dorothee Ville
- Reference Center for Tuberous Sclerosis and Rare Epileptic Syndromes, Lyon University Hospital, Lyon, France
| | - Julitta de Bellescize
- Epilepsy, Sleep and Pediatric Neurophysiology Department, Lyon University Hospital, Lyon, France
| | - Renaud Touraine
- Department of Genetics, Hospital Nord, Saint-Etienne University Hospital, France
| | - Fabienne Prieur
- Department of Genetics, Hospital Nord, Saint-Etienne University Hospital, France
| | - Sarra Dimassi
- CRNL, CNRS UMR 5292, INSERM U1028, Lyon, France; Department of Genetics, Lyon University Hospital, Lyon, France; Claude Bernard Lyon I University, Lyon, France
| | - Anne-Lise Poulat
- Reference Center for Tuberous Sclerosis and Rare Epileptic Syndromes, Lyon University Hospital, Lyon, France
| | - Marianne Till
- Department of Genetics, Lyon University Hospital, Lyon, France
| | - Massimiliano Rossi
- CRNL, CNRS UMR 5292, INSERM U1028, Lyon, France; Department of Genetics, Lyon University Hospital, Lyon, France
| | - Emilie Bourel-Ponchel
- Pediatric Functional Exploration of the Nervous System Service, Hospital Nord, Amiens University Hospital, Amiens, France
| | - Aline Delignières
- Department of Neurology, Hospital Nord, Amiens University Hospital, Amiens, France
| | - Anne-Gaelle Le Moing
- Department of Neurology, Hospital Nord, Amiens University Hospital, Amiens, France
| | - Clotilde Rivier
- Department of Pediatrics, Hospital Nord-Ouest, Villefranche sur Saone, France
| | - Vincent des Portes
- Reference Center for Tuberous Sclerosis and Rare Epileptic Syndromes, Lyon University Hospital, Lyon, France; Claude Bernard Lyon I University, Lyon, France; CNRS UMR 5403, Institut des Sciences Cognitives, L2C2, Bron, France
| | - Patrick Edery
- CRNL, CNRS UMR 5292, INSERM U1028, Lyon, France; Department of Genetics, Lyon University Hospital, Lyon, France; Claude Bernard Lyon I University, Lyon, France
| | - Alain Calender
- Department of Molecular Genetics, Lyon University Hospital, Lyon, France; Claude Bernard Lyon I University, Lyon, France; INSERM U1052, Lyon, France
| | - Damien Sanlaville
- CRNL, CNRS UMR 5292, INSERM U1028, Lyon, France; Department of Genetics, Lyon University Hospital, Lyon, France; Claude Bernard Lyon I University, Lyon, France
| | - Gaetan Lesca
- CRNL, CNRS UMR 5292, INSERM U1028, Lyon, France; Department of Genetics, Lyon University Hospital, Lyon, France; Claude Bernard Lyon I University, Lyon, France.
| |
Collapse
|
19
|
Mahajan K, Mahajan NP. ACK1/TNK2 tyrosine kinase: molecular signaling and evolving role in cancers. Oncogene 2014; 34:4162-7. [PMID: 25347744 PMCID: PMC4411206 DOI: 10.1038/onc.2014.350] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 09/17/2014] [Accepted: 09/22/2014] [Indexed: 01/11/2023]
Abstract
Deregulated tyrosine kinase signaling alters cellular homeostasis to drive cancer progression. The emergence of a non-receptor tyrosine kinase, ACK1 as an oncogenic kinase, has uncovered novel mechanisms by which tyrosine kinase signaling promotes cancer progression. While early studies focused on ACK1 (also known as activated Cdc42-associated kinase 1 or TNK2) as a cytosolic effecter of activated transmembrane receptor tyrosine kinases (RTKs), wherein it shuttles between the cytosol and the nucleus to rapidly transduce extracellular signals from the RTKs to the intracellular effectors, recent data unfold a new aspect of its functionality as an epigenetic regulator. ACK1 interacts with the Estrogen Receptor (ER)/histone demethylase KDM3A (JHDM2a) complex, modifies KDM3A by tyrosine phosphorylation to regulate transcriptional outcome at HOXA1 locus to promote the growth of tamoxifen-resistant breast cancer. It is also well established that ACK1 regulates the activity of Androgen Receptor (AR) by tyrosine phosphorylation to fuel the growth of hormone-refractory prostate cancers. Further, recent explosion in genomic sequencing has revealed recurrent ACK1 gene amplification and somatic mutations in a variety of human malignancies, providing a molecular basis for its role in neoplastic transformation. In this review, we will discuss the various facets of ACK1 signaling, including its newly uncovered epigenetic regulator function, which enables cells to bypass the blockade to major survival pathways to promote resistance to standard cancer treatments. Not surprisingly, cancer cells appear to acquire an `addiction’ to ACK1 mediated survival, particularly under stress conditions, such as growth factor deprivation or genotoxic insults or hormone deprivation. With the accelerated development of potent and selective ACK1 inhibitors, targeted treatment for cancers harboring aberrant ACK1 activity may soon become a clinical reality.
Collapse
Affiliation(s)
- K Mahajan
- 1] Moffitt Cancer Center, Drug Discovery Department, Tampa, FL, USA [2] Department of Oncologic Sciences, University of South Florida, Tampa, FL, USA
| | - N P Mahajan
- 1] Moffitt Cancer Center, Drug Discovery Department, Tampa, FL, USA [2] Department of Oncologic Sciences, University of South Florida, Tampa, FL, USA
| |
Collapse
|
20
|
Millichap JG. SCN1A and Susceptibility to MTL Epilepsy, Hippocampal Sclerosis and Febrile Seizures. Pediatr Neurol Briefs 2013. [DOI: 10.15844/pedneurbriefs-27-12-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
21
|
Abstract
In the September 12, 2013 issue of Nature, the Epi4K Consortium (Allen et al., 2013) reported sequencing 264 patient trios with epileptic encephalopathies. The Consortium focused on genes exceptionally intolerant to sequence variations and found substantial interconnections with autism and intellectual disability gene networks.
Collapse
Affiliation(s)
- Gaia Novarino
- Howard Hughes Medical Institute, Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
22
|
Allen AS, Berkovic SF, Cossette P, Delanty N, Dlugos D, Eichler EE, Epstein MP, Glauser T, Goldstein DB, Han Y, Heinzen EL, Hitomi Y, Howell KB, Johnson MR, Kuzniecky R, Lowenstein DH, Lu YF, Madou MRZ, Marson AG, Mefford HC, Esmaeeli Nieh S, O'Brien TJ, Ottman R, Petrovski S, Poduri A, Ruzzo EK, Scheffer IE, Sherr EH, Yuskaitis CJ, Abou-Khalil B, Alldredge BK, Bautista JF, Berkovic SF, Boro A, Cascino GD, Consalvo D, Crumrine P, Devinsky O, Dlugos D, Epstein MP, Fiol M, Fountain NB, French J, Friedman D, Geller EB, Glauser T, Glynn S, Haut SR, Hayward J, Helmers SL, Joshi S, Kanner A, Kirsch HE, Knowlton RC, Kossoff EH, Kuperman R, Kuzniecky R, Lowenstein DH, McGuire SM, Motika PV, Novotny EJ, Ottman R, Paolicchi JM, Parent JM, Park K, Poduri A, Scheffer IE, Shellhaas RA, Sherr EH, Shih JJ, Singh R, Sirven J, Smith MC, Sullivan J, Lin Thio L, Venkat A, Vining EPG, Von Allmen GK, Weisenberg JL, Widdess-Walsh P, Winawer MR. De novo mutations in epileptic encephalopathies. Nature 2013; 501:217-21. [PMID: 23934111 PMCID: PMC3773011 DOI: 10.1038/nature12439] [Citation(s) in RCA: 1136] [Impact Index Per Article: 103.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 07/09/2013] [Indexed: 12/21/2022]
Abstract
Epileptic encephalopathies are a devastating group of severe childhood epilepsy disorders for which the cause is often unknown. Here we report a screen for de novo mutations in patients with two classical epileptic encephalopathies: infantile spasms (n = 149) and Lennox-Gastaut syndrome (n = 115). We sequenced the exomes of 264 probands, and their parents, and confirmed 329 de novo mutations. A likelihood analysis showed a significant excess of de novo mutations in the ∼4,000 genes that are the most intolerant to functional genetic variation in the human population (P = 2.9 × 10(-3)). Among these are GABRB3, with de novo mutations in four patients, and ALG13, with the same de novo mutation in two patients; both genes show clear statistical evidence of association with epileptic encephalopathy. Given the relevant site-specific mutation rates, the probabilities of these outcomes occurring by chance are P = 4.1 × 10(-10) and P = 7.8 × 10(-12), respectively. Other genes with de novo mutations in this cohort include CACNA1A, CHD2, FLNA, GABRA1, GRIN1, GRIN2B, HNRNPU, IQSEC2, MTOR and NEDD4L. Finally, we show that the de novo mutations observed are enriched in specific gene sets including genes regulated by the fragile X protein (P < 10(-8)), as has been reported previously for autism spectrum disorders.
Collapse
|