1
|
Yu Z, Vromman A, Nguyen NQH, Schuermans A, Rentz T, Vellarikkal SK, Uddin MM, Niroula A, Griffin G, Honigberg MC, Lin AE, Gibson CJ, Katz DH, Tahir U, Fang S, Haidermota S, Ganesh S, Antoine T, Weinstock J, Austin TR, Ramachandran VS, Peloso GM, Hornsby W, Ganz P, Manson JE, Haring B, Kooperberg CL, Reiner AP, Bis JC, Psaty BM, Min YI, Correa A, Lange LA, Post WS, Rotter JI, Rich SS, Wilson JG, Ebert BL, Yu B, Ballantyne CM, Coresh J, Sankaran VG, Bick AG, Jaiswal S, Gerszten RE, Libby P, Gupta RM, Natarajan P. Human Plasma Proteomic Profile of Clonal Hematopoiesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.25.550557. [PMID: 39554199 PMCID: PMC11565774 DOI: 10.1101/2023.07.25.550557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Plasma proteomic profiles associated with subclinical somatic mutations in blood cells may offer novel insights into downstream clinical consequences. Here, we explore such patterns in clonal hematopoiesis of indeterminate potential (CHIP), which is linked to several cancer and non-cancer outcomes, including coronary artery disease (CAD). Among 61,833 ancestrally diverse participants (3,881 with CHIP) from NHLBI TOPMed and UK Biobank with blood-based DNA sequencing and proteomic measurements (1,148 proteins by SomaScan in TOPMed and 2,917 proteins by Olink in UK Biobank), we identified 32 and 345 unique proteins from TOPMed and UK Biobank, respectively, associated with the most prevalent driver genes ( DNMT3A , TET2 , and ASXL1 ). These associations showed substantial heterogeneity by driver genes, sex, and race, and were enriched for immune response and inflammation pathways. Mendelian randomization in humans, coupled with ELISA in hematopoietic Tet2 -/- vs wild-type mice validation, disentangled causal proteomic perturbations from TET2 CHIP. Lastly, we identified plasma proteins shared between CHIP and CAD.
Collapse
|
2
|
Liu F, Schrack JA, Walston J, Mathias RA, Windham BG, Grams ME, Coresh J, Walker KA. Mid-life plasma proteins associated with late-life prefrailty and frailty: a proteomic analysis. GeroScience 2024; 46:5247-5265. [PMID: 38856871 PMCID: PMC11336072 DOI: 10.1007/s11357-024-01219-8] [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: 03/25/2024] [Accepted: 05/21/2024] [Indexed: 06/11/2024] Open
Abstract
Physical frailty is a syndrome that typically manifests in later life, although the pathogenic process causing physical frailty likely begins decades earlier. To date, few studies have examined the biological signatures in mid-life associated with physical frailty later in life. Among 4,189 middle-aged participants (57.8 ± 5.0 years, 55.8% women) from the Atherosclerosis Risk in Community (ARIC) study, we evaluated the associations of 4,955 plasma proteins (log 2-transformed and standardized) measured using the SomaScan platform with their frailty status approximately 20 years later. Using multinomial logistic regression models adjusting for demographics, health behaviors, kidney function, total cholesterol, and comorbidities, 12 and 221 proteins were associated with prefrailty and frailty in later life, respectively (FDR p < 0.05). Top frailty-associated proteins included neurocan core protein (NCAN, OR = 0.66), fatty acid-binding protein heart (FABP3, OR = 1.62) and adipocyte (FABP4, OR = 1.65), as well proteins involved in the contactin-1 (CNTN1), toll-like receptor 5 (TLR5), and neurogenic locus notch homolog protein 1 (NOTCH1) signaling pathway relevant to skeletal muscle regeneration, myelination, and inflammation. Pathway analyses suggest midlife dysregulation of inflammation, metabolism, extracellular matrix, angiogenesis, and lysosomal autophagy among those at risk for late-life frailty. After further adjusting for midlife body mass index (BMI) - an established frailty risk factor - only CNTN1 (OR = 0.75) remained significantly associated with frailty. Post-hoc analyses demonstrated that the top 41 midlife frailty-associated proteins mediate 32% of the association between mid-life BMI and late-life frailty. Our findings provide new insights into frailty etiology earlier in the life course, enhancing the potential for prevention.
Collapse
Affiliation(s)
- Fangyu Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Jennifer A Schrack
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Center On Aging and Health, Johns Hopkins University, Baltimore, MD, USA
| | - Jeremy Walston
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Rasika A Mathias
- Genomics and Precision Health Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infection Disease, Bethesda, MD, USA
| | - B Gwen Windham
- Department of Medicine, MIND Center, University of Mississippi Medical Center, Jackson, MS, USA
| | - Morgan E Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Population Health and Medicine, Optimal Aging Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute On Aging, Baltimore, MD, USA
| |
Collapse
|
3
|
Qin L, Qiu M, Lin Q, Jiang B, Zhan S, Wei X, Wei J, Liu Y, Wen Q, Chen P, Jiang Y, Zhou Z, Liang X, Cao J, Gong Y, Wei Y, Wei X, Yu H. Association between novel genetic variants of Notch signaling pathway genes and survival of hepatitis B virus-related hepatocellular carcinoma. Cancer Med 2024; 13:e7040. [PMID: 38562021 PMCID: PMC10985410 DOI: 10.1002/cam4.7040] [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: 09/16/2023] [Revised: 10/22/2023] [Accepted: 02/08/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Although the Notch pathway plays an important role in formation and progression of hepatocellular carcinoma (HCC), few studies have reported the associations between functional genetic variants and the survival of hepatitis B virus (HBV)-related HCC. METHODS In the present study, we performed multivariable Cox proportional hazard regression analysis to evaluate associations between 36,101 SNPs in 264 Notch pathway-related genes and overall survival (OS) of 866 patients with HBV-related HCC. RESULTS It was found that three independent SNPs (NEURL1B rs4868192, CNTN1 rs444927 and FCER2 rs1990975) were significantly associated with the HBV-related HCC OS. The number of protective genotypes (NPGs) were significantly associated with better survival in a dose-response manner (ptrend <0.001). Compared with the model with sole clinical factors, the addition of protective genotypes to the predict models significantly increased the AUC, i.e., from 72.72% to 75.13% (p = 0.002) and from 72.04% to 74.76 (p = 0.004) for 3-year and 5-year OS, respectively. The expression quantitative trait loci (eQTL) analysis further revealed that the rs4868192 C allele was associated with lower mRNA expression levels of NEURL1B in the whole blood (p = 1.71 × 10-3), while the rs1990975 T allele was correlated with higher mRNA expression levels of FCER2 in the whole blood and normal liver tissues (p = 3.51 × 10-5 and 0.033, respectively). CONCLUSIONS Three potentially functional SNPs of NEURL1B, CNTN1 and FCER2 may serve as potential prognostic biomarkers for HBV-related HCC.
Collapse
Affiliation(s)
- Liming Qin
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Moqin Qiu
- Department of Respiratory OncologyGuangxi Medical University Cancer HospitalNanningChina
| | - Qiuling Lin
- Department of Clinical ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Binbin Jiang
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Shicheng Zhan
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Xueyan Wei
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Junjie Wei
- Department of Epidemiology and Health Statistics, School of Public HealthGuangxi Medical UniversityNanningChina
| | - Yingchun Liu
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Qiuping Wen
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Peiqin Chen
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Yanji Jiang
- Department of Scientific ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Zihan Zhou
- Department of Cancer Prevention and ControlGuangxi Medical University Cancer HospitalNanningChina
| | - Xiumei Liang
- Department of Disease Process ManagementGuangxi Medical University Cancer HospitalNanningChina
| | - Ji Cao
- Department of Cancer Prevention and ControlGuangxi Medical University Cancer HospitalNanningChina
| | - Yizhen Gong
- Department of Clinical ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Yuying Wei
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Xiaoxia Wei
- Department of Clinical ResearchGuangxi Medical University Cancer HospitalNanningChina
| | - Hongping Yu
- Department of Experimental ResearchGuangxi Medical University Cancer HospitalNanningChina
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor(Guangxi Medical University), Ministry of EducationNanningChina
- Key Cultivated Laboratory of Cancer Molecular Medicine of Guangxi Health CommissionGuangxi Medical University Cancer HospitalNanningChina
| |
Collapse
|
4
|
Dunn PJ, Lea RA, Maksemous N, Smith RA, Sutherland HG, Haupt LM, Griffiths LR. Exonic mutations in cell-cell adhesion may contribute to CADASIL-related CSVD pathology. Hum Genet 2023; 142:1361-1373. [PMID: 37422595 PMCID: PMC10449969 DOI: 10.1007/s00439-023-02584-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/27/2023] [Indexed: 07/10/2023]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a condition caused by mutations in NOTCH3 and results in a phenotype characterised by recurrent strokes, vascular dementia and migraines. Whilst a genetic basis for the disease is known, the molecular mechanisms underpinning the pathology of CADASIL are still yet to be determined. Studies conducted at the Genomics Research Centre (GRC) have also identified that only 15-23% of individuals clinically suspected of CADASIL have mutations in NOTCH3. Based on this, whole exome sequencing was used to identify novel genetic variants for CADASIL-like cerebral small-vessel disease (CSVD). Analysis of functionally important variants in 50 individuals was investigated using overrepresentation tests in Gene ontology software to identify biological processes that are potentially affected in this group of patients. Further investigation of the genes in these processes was completed using the TRAPD software to identify if there is an increased number (burden) of mutations that are associated with CADASIL-like pathology. Results from this study identified that cell-cell adhesion genes were positively overrepresented in the PANTHER GO-slim database. TRAPD burden testing identified n = 15 genes that had a higher number of rare (MAF < 0.001) and predicted functionally relevant (SIFT < 0.05, PolyPhen > 0.8) mutations compared to the gnomAD v2.1.1 exome control dataset. Furthermore, these results identified ARVCF, GPR17, PTPRS, and CELSR1 as novel candidate genes in CADASIL-related pathology. This study identified a novel process that may be playing a role in the vascular damage related to CADASIL-related CSVD and implicated n = 15 genes in playing a role in the disease.
Collapse
Affiliation(s)
- Paul J Dunn
- Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
- Faculty of Health Sciences and Medicine, Bond University, 15 University Drive, Robina, Gold Coast, QLD, 4226, Australia
| | - Rodney A Lea
- Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Neven Maksemous
- Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Robert A Smith
- Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Heidi G Sutherland
- Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Larisa M Haupt
- Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, Australia
- Max Planck Queensland Centre for the Materials Sciences of Extracellular Matrices, Brisbane, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia.
| |
Collapse
|
5
|
Zhang L, Zeng Z, Lu X, Li M, Yao J, Zou G, Chen Z, Li Q, Li C, Li F. CNTN1 in the Nucleus Accumbens is Involved in Methamphetamine-Induced Conditioned Place Preference in Mice. Neurotox Res 2023; 41:324-337. [PMID: 37014368 DOI: 10.1007/s12640-023-00640-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/02/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023]
Abstract
Methamphetamine (Meth), a commonly used central nervous system stimulant, is highly addictive. Currently, there is no effective treatment for Meth dependence and abuse, although cell adhesion molecules (CAMs) have been shown to play an important role in the formation and remodeling of synapses in the nervous system while also being involved in addictive behavior. Contactin 1 (CNTN1) is a CAM that is widely expressed in the brain; nevertheless, its role in Meth addiction remains unclear. Therefore, in the present study, we established mouse models of single and repeated Meth exposure and subsequently determined that CNTN1 expression in the nucleus accumbens (NAc) was upregulated in mice following single or repeated Meth exposure, whereas CNTN1 expression in the hippocampus was not significantly altered. Intraperitoneal injection of the dopamine receptor 2 antagonist haloperidol reversed Meth-induced hyperlocomotion and upregulation of CNTN1 expression in the NAc. Additionally, repeated Meth exposure also induced conditioned place preference (CPP) in mice and upregulated the expression levels of CNTN1, NR2A, NR2B, and PSD95 in the NAc. Using an AAV-shRNA-based approach to specifically silence CNTN1 expression in the NAc via brain stereotaxis reversed Meth-induced CPP and decreased the expression levels of NR2A, NR2B, and PSD95 in the NAc. These findings suggest that CNTN1 expression in the NAc plays an important role in Meth-induced addiction, and the underlying mechanism may be related to the expression of synapse-associated proteins in the NAc. The results of this study improved our understanding of the role of cell adhesion molecules in Meth addiction.
Collapse
Affiliation(s)
- Linxuan Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan Province, China
| | - Zehao Zeng
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan Province, China
| | - Xiaoyu Lu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan Province, China
| | - Mengqing Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan Province, China
| | - Jiayu Yao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan Province, China
| | - Guangjing Zou
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan Province, China
| | - Zhaorong Chen
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan Province, China
| | - Qian Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan Province, China
| | - Changqi Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan Province, China
| | - Fang Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, Hunan Province, China.
| |
Collapse
|
6
|
Bizzoca A, Jirillo E, Flace P, Gennarini G. Overall Role of Contactins Expression in Neurodevelopmental Events and Contribution to Neurological Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:1176-1193. [PMID: 36515028 DOI: 10.2174/1871527322666221212160048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/21/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Neurodegenerative disorders may depend upon a misregulation of the pathways which sustain neurodevelopmental control. In this context, this review article focuses on Friedreich ataxia (FA), a neurodegenerative disorder resulting from mutations within the gene encoding the Frataxin protein, which is involved in the control of mitochondrial function and oxidative metabolism. OBJECTIVE The specific aim of the present study concerns the FA molecular and cellular substrates, for which available transgenic mice models are proposed, including mutants undergoing misexpression of adhesive/morphoregulatory proteins, in particular belonging to the Contactin subset of the immunoglobulin supergene family. METHODS In both mutant and control mice, neurogenesis was explored by morphological/morphometric analysis through the expression of cell type-specific markers, including b-tubulin, the Contactin-1 axonal adhesive glycoprotein, as well as the Glial Fibrillary Acidic Protein (GFAP). RESULTS Specific consequences were found to arise from the chosen misexpression approach, consisting of a neuronal developmental delay associated with glial upregulation. Protective effects against the arising phenotype resulted from antioxidants (essentially epigallocatechin gallate (EGCG)) administration, which was demonstrated through the profiles of neuronal (b-tubulin and Contactin 1) as well as glial (GFAP) markers, in turn indicating the concomitant activation of neurodegeneration and neuro repair processes. The latter also implied activation of the Notch-1 signaling. CONCLUSION Overall, this study supports the significance of changes in morphoregulatory proteins expression in the FA pathogenesis and of antioxidant administration in counteracting it, which, in turn, allows to devise potential therapeutic approaches.
Collapse
Affiliation(s)
- Antonella Bizzoca
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| | - Paolo Flace
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| | - Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| |
Collapse
|
7
|
Xu S, Han L, Wei Y, Zhang B, Wang Q, Liu J, Liu M, Chen Z, Wang Z, Chen H, Zhu Q. MicroRNA-200c-targeted contactin 1 facilitates the replication of influenza A virus by accelerating the degradation of MAVS. PLoS Pathog 2022; 18:e1010299. [PMID: 35171955 PMCID: PMC8849533 DOI: 10.1371/journal.ppat.1010299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/21/2022] [Indexed: 01/06/2023] Open
Abstract
Influenza A viruses (IAVs) continuously challenge the poultry industry and human health. Elucidation of the host factors that modulate the IAV lifecycle is vital for developing antiviral drugs and vaccines. In this study, we infected A549 cells with IAVs and found that host protein contactin-1 (CNTN1), a member of the immunoglobulin superfamily, enhanced viral replication. Bioinformatic prediction and experimental validation indicated that the expression of CNTN1 was reduced by microRNA-200c (miR-200c) through directly targeting. We further showed that CNTN1-modulated viral replication in A549 cells is dependent on type I interferon signaling. Co-immunoprecipitation experiments revealed that CNTN1 specifically interacts with MAVS and promotes its proteasomal degradation by removing its K63-linked ubiquitination. Moreover, we discovered that the deubiquitinase USP25 is recruited by CNTN1 to catalyze the deubiquitination of K63-linked MAVS. Consequently, the CNTN1-induced degradation cascade of MAVS blocked RIG-I-MAVS-mediated interferon signaling, leading to enhanced viral replication. Taken together, our data reveal novel roles of CNTN1 in the type I interferon pathway and regulatory mechanism of IAV replication.
Collapse
Affiliation(s)
- Shuai Xu
- State Key Laboratory of Veterinary Etiological Biology, College of Animal Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, PR China
| | - Lu Han
- State Key Laboratory of Veterinary Etiological Biology, College of Animal Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, PR China
| | - Yanli Wei
- State Key Laboratory of Veterinary Etiological Biology, College of Animal Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, PR China
| | - Bo Zhang
- State Key Laboratory of Veterinary Etiological Biology, College of Animal Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, PR China
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Qian Wang
- State Key Laboratory of Veterinary Etiological Biology, College of Animal Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, PR China
| | - Junwen Liu
- State Key Laboratory of Veterinary Etiological Biology, College of Animal Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, PR China
| | - Minxuan Liu
- State Key Laboratory of Veterinary Etiological Biology, College of Animal Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, PR China
| | - Zhaoshan Chen
- State Key Laboratory of Veterinary Etiological Biology, College of Animal Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, PR China
| | - Zhengxiang Wang
- State Key Laboratory of Veterinary Etiological Biology, College of Animal Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, PR China
| | - Hualan Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Qiyun Zhu
- State Key Laboratory of Veterinary Etiological Biology, College of Animal Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, PR China
| |
Collapse
|
8
|
E3 Ubiquitin Ligase Regulators of Notch Receptor Endocytosis: From Flies to Humans. Biomolecules 2022; 12:biom12020224. [PMID: 35204725 PMCID: PMC8961608 DOI: 10.3390/biom12020224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
Notch is a developmental receptor, conserved in the evolution of the metazoa, which regulates cell fate proliferation and survival in numerous developmental contexts, and also regulates tissue renewal and repair in adult organisms. Notch is activated by proteolytic removal of its extracellular domain and the subsequent release of its intracellular domain, which then acts in the nucleus as part of a transcription factor complex. Numerous regulatory mechanisms exist to tune the amplitude, duration and spatial patterning of this core signalling mechanism. In Drosophila, Deltex (Dx) and Suppressor of dx (Su(dx)) are E3 ubiquitin ligases which interact with the Notch intracellular domain to regulate its endocytic trafficking, with impacts on both ligand-dependent and ligand-independent signal activation. Homologues of Dx and Su(dx) have been shown to also interact with one or more of the four mammalian Notch proteins and other target substrates. Studies have shown similarities, specialisations and diversifications of the roles of these Notch regulators. This review collates together current research on vertebrate Dx and Su(dx)-related proteins, provides an overview of their various roles, and discusses their contributions to cell fate regulation and disease.
Collapse
|
9
|
Eve M, Gandawijaya J, Yang L, Oguro-Ando A. Neuronal Cell Adhesion Molecules May Mediate Neuroinflammation in Autism Spectrum Disorder. Front Psychiatry 2022; 13:842755. [PMID: 35492721 PMCID: PMC9051034 DOI: 10.3389/fpsyt.2022.842755] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by restrictive and repetitive behaviors, alongside deficits in social interaction and communication. The etiology of ASD is largely unknown but is strongly linked to genetic variants in neuronal cell adhesion molecules (CAMs), cell-surface proteins that have important roles in neurodevelopment. A combination of environmental and genetic factors are believed to contribute to ASD pathogenesis. Inflammation in ASD has been identified as one of these factors, demonstrated through the presence of proinflammatory cytokines, maternal immune activation, and activation of glial cells in ASD brains. Glial cells are the main source of cytokines within the brain and, therefore, their activity is vital in mediating inflammation in the central nervous system. However, it is unclear whether the aforementioned neuronal CAMs are involved in modulating neuroimmune signaling or glial behavior. This review aims to address the largely unexplored role that neuronal CAMs may play in mediating inflammatory cascades that underpin neuroinflammation in ASD, primarily focusing on the Notch, nuclear factor-κB (NF-κB), and mitogen-activated protein kinase (MAPK) cascades. We will also evaluate the available evidence on how neuronal CAMs may influence glial activity associated with inflammation. This is important when considering the impact of environmental factors and inflammatory responses on ASD development. In particular, neural CAM1 (NCAM1) can regulate NF-κB transcription in neurons, directly altering proinflammatory signaling. Additionally, NCAM1 and contactin-1 appear to mediate astrocyte and oligodendrocyte precursor proliferation which can alter the neuroimmune response. Importantly, although this review highlights the limited information available, there is evidence of a neuronal CAM regulatory role in inflammatory signaling. This warrants further investigation into the role other neuronal CAM family members may have in mediating inflammatory cascades and would advance our understanding of how neuroinflammation can contribute to ASD pathology.
Collapse
Affiliation(s)
- Madeline Eve
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Josan Gandawijaya
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Liming Yang
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Asami Oguro-Ando
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
10
|
Epithelial exosomal contactin-1 promotes monocyte-derived dendritic cell-dominant T-cell responses in asthma. J Allergy Clin Immunol 2021; 148:1545-1558. [PMID: 33957164 DOI: 10.1016/j.jaci.2021.04.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Exosomes have emerged as a vital player in cell-cell communication; however, whether airway epithelial cell (AEC)-generated exosomes participate in asthma development remains unknown. OBJECTIVE Our aims were to characterize the AEC-secreted exosomes and the potentially functional protein(s) that may contribute to the proinflammatory effects of AEC exosomes in the dendritic cell (DC)-dominant airway allergic models and to confirm their clinical significance in patients with asthma. METHODS Mice were treated with exosomes derived from house dust mite (HDM)-stimulated AECs (HDM-AEC-EXOs) or monocyte-derived DCs primed by HDM and/or contactin-1 (CNTN1). The numbers of DCs in the lung were determined by flow cytometry. Proteomic analysis of purified HDM-AEC-EXOs was performed. CNTN1 small interfering RNA was designed to probe its role in airway allergy, and γ-secretase inhibitor was used to determine involvement of the Notch pathway. RESULTS HDM-AEC-EXOs facilitate the recruitment, proliferation, migration, and activation of monocyte-derived DCs in cell culture and in mice. CNTN1 in exosomes is a critical player in asthma pathology. RNA interference-mediated silencing and pharmaceutical inhibitors characterize Notch2 receptor as necessary for relaying the CNTN1 signal to activate TH2 cell/TH17 cell immune response. Studies of patients with asthma also support existence of the CNTN1-Notch2 axis that has been observed in cell and mouse models. CONCLUSION This study's findings reveal a novel role for CNTN1 in asthma pathogenesis mediated through exosome secretion, indicating a potential strategy for the treatment of allergic airway inflammation.
Collapse
|
11
|
Lecluze E, Rolland AD, Filis P, Evrard B, Leverrier-Penna S, Maamar MB, Coiffec I, Lavoué V, Fowler PA, Mazaud-Guittot S, Jégou B, Chalmel F. Dynamics of the transcriptional landscape during human fetal testis and ovary development. Hum Reprod 2021; 35:1099-1119. [PMID: 32412604 DOI: 10.1093/humrep/deaa041] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/10/2020] [Indexed: 12/17/2022] Open
Abstract
STUDY QUESTION Which transcriptional program triggers sex differentiation in bipotential gonads and downstream cellular events governing fetal testis and ovary development in humans? SUMMARY ANSWER The characterization of a dynamically regulated protein-coding and non-coding transcriptional landscape in developing human gonads of both sexes highlights a large number of potential key regulators that show an early sexually dimorphic expression pattern. WHAT IS KNOWN ALREADY Gonadal sex differentiation is orchestrated by a sexually dimorphic gene expression program in XX and XY developing fetal gonads. A comprehensive characterization of its non-coding counterpart offers promising perspectives for deciphering the molecular events underpinning gonad development and for a complete understanding of the etiology of disorders of sex development in humans. STUDY DESIGN, SIZE, DURATION To further investigate the protein-coding and non-coding transcriptional landscape during gonad differentiation, we used RNA-sequencing (RNA-seq) and characterized the RNA content of human fetal testis (N = 24) and ovaries (N = 24) from 6 to 17 postconceptional week (PCW), a key period in sex determination and gonad development. PARTICIPANTS/MATERIALS, SETTING, METHODS First trimester fetuses (6-12 PCW) and second trimester fetuses (13-14 and 17 PCW) were obtained from legally induced normally progressing terminations of pregnancy. Total RNA was extracted from whole human fetal gonads and sequenced as paired-end 2 × 50 base reads. Resulting sequences were mapped to the human genome, allowing for the assembly and quantification of corresponding transcripts. MAIN RESULTS AND THE ROLE OF CHANCE This RNA-seq analysis of human fetal testes and ovaries at seven key developmental stages led to the reconstruction of 22 080 transcripts differentially expressed during testicular and/or ovarian development. In addition to 8935 transcripts displaying sex-independent differential expression during gonad development, the comparison of testes and ovaries enabled the discrimination of 13 145 transcripts that show a sexually dimorphic expression profile. The latter include 1479 transcripts differentially expressed as early as 6 PCW, including 39 transcription factors, 40 long non-coding RNAs and 20 novel genes. Despite the use of stringent filtration criteria (expression cut-off of at least 1 fragment per kilobase of exon model per million reads mapped, fold change of at least 2 and false discovery rate adjusted P values of less than <1%), the possibility of assembly artifacts and of false-positive differentially expressed transcripts cannot be fully ruled out. LARGE-SCALE DATA Raw data files (fastq) and a searchable table (.xlss) containing information on genomic features and expression data for all refined transcripts have been submitted to the NCBI GEO under accession number GSE116278. LIMITATIONS, REASONS FOR CAUTION The intrinsic nature of this bulk analysis, i.e. the sequencing of transcripts from whole gonads, does not allow direct identification of the cellular origin(s) of the transcripts characterized. Potential cellular dilution effects (e.g. as a result of distinct proliferation rates in XX and XY gonads) may account for a few of the expression profiles identified as being sexually dimorphic. Finally, transcriptome alterations that would result from exposure to pre-abortive drugs cannot be completely excluded. Although we demonstrated the high quality of the sorted cell populations used for experimental validations using quantitative RT-PCR, it cannot be totally excluded that some germline expression may correspond to cell contamination by, for example, macrophages. WIDER IMPLICATIONS OF THE FINDINGS For the first time, this study has led to the identification of 1000 protein-coding and non-coding candidate genes showing an early, sexually dimorphic, expression pattern that have not previously been associated with sex differentiation. Collectively, these results increase our understanding of gonad development in humans, and contribute significantly to the identification of new candidate genes involved in fetal gonad differentiation. The results also provide a unique resource that may improve our understanding of the fetal origin of testicular and ovarian dysgenesis syndromes, including cryptorchidism and testicular cancers. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the French National Institute of Health and Medical Research (Inserm), the University of Rennes 1, the French School of Public Health (EHESP), the Swiss National Science Foundation [SNF n° CRS115_171007 to B.J.], the French National Research Agency [ANR n° 16-CE14-0017-02 and n° 18-CE14-0038-02 to F.C.], the Medical Research Council [MR/L010011/1 to P.A.F.] and the European Community's Seventh Framework Programme (FP7/2007-2013) [under grant agreement no 212885 to P.A.F.] and from the European Union's Horizon 2020 Research and Innovation Programme [under grant agreement no 825100 to P.A.F. and S.M.G.]. There are no competing interests related to this study.
Collapse
Affiliation(s)
- Estelle Lecluze
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Antoine D Rolland
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Panagiotis Filis
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Bertrand Evrard
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Sabrina Leverrier-Penna
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France.,Univ Poitiers, STIM, CNRS ERL7003, Poitiers Cedex 9, CNRS ERL7003, France
| | - Millissia Ben Maamar
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Isabelle Coiffec
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Vincent Lavoué
- Service Gynécologie et Obstétrique, CHU Rennes, F-35000 Rennes, France
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Séverine Mazaud-Guittot
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Bernard Jégou
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| | - Frédéric Chalmel
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, F-35000 Rennes, France
| |
Collapse
|
12
|
Kalafatakis I, Savvaki M, Velona T, Karagogeos D. Implication of Contactins in Demyelinating Pathologies. Life (Basel) 2021; 11:life11010051. [PMID: 33451101 PMCID: PMC7828632 DOI: 10.3390/life11010051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Demyelinating pathologies comprise of a variety of conditions where either central or peripheral myelin is attacked, resulting in white matter lesions and neurodegeneration. Myelinated axons are organized into molecularly distinct domains, and this segregation is crucial for their proper function. These defined domains are differentially affected at the different stages of demyelination as well as at the lesion and perilesion sites. Among the main players in myelinated axon organization are proteins of the contactin (CNTN) group of the immunoglobulin superfamily (IgSF) of cell adhesion molecules, namely Contactin-1 and Contactin-2 (CNTN1, CNTN2). The two contactins perform their functions through intermolecular interactions, which are crucial for myelinated axon integrity and functionality. In this review, we focus on the implication of these two molecules as well as their interactors in demyelinating pathologies in humans. At first, we describe the organization and function of myelinated axons in the central (CNS) and the peripheral (PNS) nervous system, further analyzing the role of CNTN1 and CNTN2 as well as their interactors in myelination. In the last section, studies showing the correlation of the two contactins with demyelinating pathologies are reviewed, highlighting the importance of these recognition molecules in shaping the function of the nervous system in multiple ways.
Collapse
|
13
|
Liang Y, Ma C, Li F, Nie G, Zhang H. The Role of Contactin 1 in Cancers: What We Know So Far. Front Oncol 2020; 10:574208. [PMID: 33194679 PMCID: PMC7658624 DOI: 10.3389/fonc.2020.574208] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022] Open
Abstract
Cancers are among the difficult-to-treat diseases despite advances in diagnosis and treatment. Although newer effective targets remain to be discovered, targeted therapy has emerged as a promising field. In the last decade, contactin 1 (CNTN1) has surfaced as an important cancer-related molecule. CNTN1 is a neuronal membrane glycoprotein, which, if overexpressed, is found in different cancer cell lines, cancer tissues, and transgenic mice. It is positively associated with lymphatic invasion, metastasis, late TNM stage, and a short overall survival time. However, the role of CNTN1 in cancer cell proliferation remains unclear. In addition, CNTN1 is involved in cancer cell invasion, migration, metastasis, and chemoresistance by promoting epithelial-mesenchymal transition and mediating several signal transduction pathways. Several studies suggest CNTN1 as a new therapeutic target for cancers. This review aims to summarize the research developments on CNTN1 in various cancers, to establish its role in epithelial-mesenchymal transition and signal transduction pathways, and to identify promising areas for further investigation.
Collapse
Affiliation(s)
- Yumei Liang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Cui Ma
- Department of Pediatric Hematology, The First Hospital of Jilin University, Changchun, China
| | - Fengjuan Li
- Oncology Department of Tumor Center, The First Hospital of Jilin University, Changchun, China
| | - Guanhua Nie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Haining Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Bizzoca A, Caracciolo M, Corsi P, Magrone T, Jirillo E, Gennarini G. Molecular and Cellular Substrates for the Friedreich Ataxia. Significance of Contactin Expression and of Antioxidant Administration. Molecules 2020; 25:E4085. [PMID: 32906751 PMCID: PMC7570916 DOI: 10.3390/molecules25184085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 11/16/2022] Open
Abstract
In this study, the neural phenotype is explored in rodent models of the spinocerebellar disorder known as the Friedreich Ataxia (FA), which results from mutations within the gene encoding the Frataxin mitochondrial protein. For this, the M12 line, bearing a targeted mutation, which disrupts the Frataxin gene exon 4 was used, together with the M02 line, which, in addition, is hemizygous for the human Frataxin gene mutation (Pook transgene), implying the occurrence of 82-190 GAA repeats within its first intron. The mutant mice phenotype was compared to the one of wild type littermates in regions undergoing differential profiles of neurogenesis, including the cerebellar cortex and the spinal cord by using neuronal (β-tubulin) and glial (Glial Fibrillary Acidic Protein) markers as well as the Contactin 1 axonal glycoprotein, involved in neurite growth control. Morphological/morphometric analyses revealed that while in Frataxin mutant mice the neuronal phenotype was significantly counteracted, a glial upregulation occurred at the same time. Furthermore, Contactin 1 downregulation suggested that changes in the underlying gene contributed to the disorder pathogenesis. Therefore, the FA phenotype implies an alteration of the developmental profile of neuronal and glial precursors. Finally, epigallocatechin gallate polyphenol administration counteracted the disorder, indicating protective effects of antioxidant administration.
Collapse
Affiliation(s)
| | | | | | | | | | - Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. I-70124 Bari, Italy; (A.B.); (M.C.); (P.C.); (T.M.); (E.J.)
| |
Collapse
|
15
|
Gu Y, Li T, Kapoor A, Major P, Tang D. Contactin 1: An Important and Emerging Oncogenic Protein Promoting Cancer Progression and Metastasis. Genes (Basel) 2020; 11:E874. [PMID: 32752094 PMCID: PMC7465769 DOI: 10.3390/genes11080874] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Even with recent progress, cancer remains the second leading cause of death, outlining a need to widen the current understanding on oncogenic factors. Accumulating evidence from recent years suggest Contactin 1 (CNTN1)'s possession of multiple oncogenic activities in a variety of cancer types. CNTN1 is a cell adhesion molecule that is dysregulated in many human carcinomas and plays important roles in cancer progression and metastases. Abnormalities in CNTN1 expression associate with cancer progression and poor prognosis. Mechanistically, CNTN1 functions in various signaling pathways frequently altered in cancer, such as the vascular endothelial growth factor C (VEGFC)-VEGF receptor 3 (VEFGR3)/fms-related tyrosine kinase 4 (Flt4) axis, phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT), Notch signaling pathway and epithelial-mesenchymal transition (EMT) process. These oncogenic events are resulted via interactions between tumor and stroma, which can be contributed by CNTN1, an adhesion protein. CNTN1 expression in breast cancer correlates with the expression of genes functioning in cancer-stroma interactions and skeletal system development. Evidence supports that CNTN1 promotes cancer-stromal interaction, resulting in activation of a complex network required for cancer progression and metastasis (bone metastasis for breast cancer). CNTN1 inhibitions has been proven to be effective in experimental models to reduce oncogenesis. In this paper, we will review CNTN1's alterations in cancer, its main biochemical mechanisms and interactions with its relevant cancer pathways.
Collapse
Affiliation(s)
- Yan Gu
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
- The Research Institute of St Joe's Hamilton, St. Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St. Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| | - Taosha Li
- Life-Tech Industry Alliance, Shenzhen 518000, China
| | - Anil Kapoor
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
- The Research Institute of St Joe's Hamilton, St. Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St. Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- Department of Surgery, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Pierre Major
- Department of Oncology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Damu Tang
- Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
- The Research Institute of St Joe's Hamilton, St. Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
- Urological Cancer Center for Research and Innovation (UCCRI), St. Joseph's Hospital, Hamilton, ON L8N 4A6, Canada
| |
Collapse
|
16
|
The Interaction Between Contactin and Amyloid Precursor Protein and Its Role in Alzheimer’s Disease. Neuroscience 2020; 424:184-202. [DOI: 10.1016/j.neuroscience.2019.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 01/06/2023]
|
17
|
Dieterich K, Kimber E, Hall JG. Central nervous system involvement in arthrogryposis multiplex congenita: Overview of causes, diagnosis, and care. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2019; 181:345-353. [PMID: 31410997 DOI: 10.1002/ajmg.c.31732] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/13/2019] [Accepted: 07/17/2019] [Indexed: 12/14/2022]
Abstract
Arthrogryposis or AMC, arthrogryposis multiplex congenita, is defined as multiple congenital joint contractures in more than two joints and in different body areas. The common cause of all AMC is lack of movement in utero, which in turn can have different causes, one of which is CNS involvement. Intellectual disability/CNS involvement is found in approximately 25% of all AMC. AMC with CNS involvement includes a large number of genetic syndromes. So far, more than 400 genes have been identified as linked to AMC, with and without CNS involvement. A number of neonatally lethal syndromes and syndromes resulting in severe disability due to CNS malfunction belong to this group of syndromes. There are several X-linked disorders with AMC, which are primarily related to intellectual disability. A number of neuromuscular disorders may include AMC and CNS/brain involvement. Careful clinical evaluation by a geneticist and a pediatrician/pediatric neurologist is the first step in making a specific diagnosis. Further investigations may include MRI of the brain and spinal cord, electroencephalogram, blood chemistry for muscle enzymes, other organ investigations (ophtalmology, cardiology, gastrointestinal, and genitourinary systems). Nerve conduction studies, electromyogram, and muscle pathology may be of help when there is associated peripheral nervous system involvement. But most importantly, genetic investigations with targeted or rather whole exome or genome sequencing should be performed. A correct diagnosis is important in planning adequate treatment, in genetic counselling and also for future understanding of pathogenic mechanisms and possible new treatments. A multidiciplinary team is needed both in investigation and treatment.
Collapse
Affiliation(s)
- Klaus Dieterich
- Univ. Grenoble Alpes, Inserm, U1216, GIN, Grenoble, France.,CHU Grenoble Alpes, Génétique Médicale, Grenoble, France
| | - Eva Kimber
- Department of Women's and Children's Health, Uppsala University Children's Hospital, Uppsala, Sweden.,Department of Paediatrics, Institute of Clinical Sciences, University of Gothenburg, The Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Judith G Hall
- Professor Emerita, Department of Pediatrics and Medical Genetics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
18
|
Picocci S, Bizzoca A, Corsi P, Magrone T, Jirillo E, Gennarini G. Modulation of Nerve Cell Differentiation: Role of Polyphenols and of Contactin Family Components. Front Cell Dev Biol 2019; 7:119. [PMID: 31380366 PMCID: PMC6656924 DOI: 10.3389/fcell.2019.00119] [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: 01/31/2019] [Accepted: 06/12/2019] [Indexed: 12/18/2022] Open
Abstract
In this study the mechanisms are explored, which modulate expression and function of cell surface adhesive glycoproteins of the Immunoglobulin Supergene Family (IgSF), and in particular of its Contactin subset, during neuronal precursor developmental events. In this context, a specific topic concerns the significance of the expression profile of such molecules and their ability to modulate signaling pathways activated through nutraceuticals, in particular polyphenols, administration. Both in vitro and in vivo approaches are chosen. As for the former, by using as a model the human SH-SY5Y neuroblastoma line, the effects of grape seed polyphenols are evaluated on proliferation and commitment/differentiation events along the neuronal lineage. In SH-SY5Y cell cultures, polyphenols were found to counteract precursor proliferation while promoting their differentiation, as deduced by studying their developmental parameters through the expression of cell cycle and neuronal commitment/differentiation markers as well as by measuring neurite growth. In such cultures, Cyclin E expression and BrdU incorporation were downregulated, indicating reduced precursor proliferation while increased neuronal differentiation was inferred from upregulation of cell cycle exit (p27–Kip) and neuronal commitment (NeuN) markers as well as by measuring neurite length through morphometric analysis. The polyphenol effects on developmental parameters were also explored in vivo, in cerebellar cortex, by using as a model the TAG/F3 transgenic line, which undergoes delayed neural development as a consequence of Contactin1 adhesive glycoprotein upregulation and premature expression under control of the Contactin2 gene (Cntn-2) promoter. In this transgenic line, a Notch pathway activation is known to occur and polyphenol treatment was found to counteract such an effect, demonstrated through downregulation of the Hes-1 transcription factor. Polyphenols also downregulated the expression of adhesive glycoproteins of the Contactin family themselves, demonstrated for both Contactin1 and Contactin2, indicating the involvement of changes in the expression of the underlying genes in the observed phenotype. These data support the hypothesis that the complex control exerted by polyphenols on neural development involves modulation of expression and function of the genes encoding cell adhesion molecules of the Contactin family and of the associated signaling pathways, indicating potential mechanisms whereby such compounds may control neurogenesis.
Collapse
Affiliation(s)
- Sabrina Picocci
- Laboratories of Developmental Neurobiology, Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Aldo Moro, Bari, Italy
| | - Antonella Bizzoca
- Laboratories of Developmental Neurobiology, Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Aldo Moro, Bari, Italy
| | - Patrizia Corsi
- Laboratories of Developmental Neurobiology, Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Aldo Moro, Bari, Italy
| | - Thea Magrone
- Laboratories of Immunology, Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Aldo Moro, Bari, Italy
| | - Emilio Jirillo
- Laboratories of Immunology, Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Aldo Moro, Bari, Italy
| | - Gianfranco Gennarini
- Laboratories of Developmental Neurobiology, Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
19
|
Qian Y, Zhang L, Cai M, Li H, Xu H, Yang H, Zhao Z, Rhie SK, Farnham PJ, Shi J, Lu W. The prostate cancer risk variant rs55958994 regulates multiple gene expression through extreme long-range chromatin interaction to control tumor progression. SCIENCE ADVANCES 2019; 5:eaaw6710. [PMID: 31328168 PMCID: PMC6636982 DOI: 10.1126/sciadv.aaw6710] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/11/2019] [Indexed: 05/15/2023]
Abstract
Genome-wide association studies identified single-nucleotide polymorphism (SNP) rs55958994 as a significant variant associated with increased susceptibility to prostate cancer. However, the mechanisms by which this SNP mediates increased risk to cancer are still unknown. In this study, we show that this variant is located in an enhancer active in prostate cancer cells. Deletion of this enhancer from prostate tumor cells resulted in decreased tumor initiation, tumor growth, and invasive migration, as well as a loss of stem-like cells. Using a combination of capture chromosome conformation capture (Capture-C) and RNA sequencing, we identified genes on the same and different chromosomes as targets regulated by the enhancer. Furthermore, we show that expression of individual candidate target genes in an enhancer-deleted cell line rescued different aspects of tumorigenesis. Our data suggest that the rs55958994-associated enhancer affects prostate cancer progression by influencing expression of multiple genes via long-range chromatin interactions.
Collapse
Affiliation(s)
- Yuyang Qian
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, 300071 Tianjin, China
| | - Lei Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, 300071 Tianjin, China
| | - Mingyang Cai
- Department of Stem Cell Biology and Regenerative Medicine, Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Hongxia Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, 300071 Tianjin, China
| | - Heming Xu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, 300071 Tianjin, China
| | - Hongzhen Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, 300071 Tianjin, China
| | - Zhongfang Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, 300071 Tianjin, China
| | - Suhn Kyong Rhie
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Peggy J. Farnham
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jiandang Shi
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, 300071 Tianjin, China
| | - Wange Lu
- Department of Stem Cell Biology and Regenerative Medicine, Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
20
|
Polyphenol Effects on Splenic Cytokine Response in Post-Weaning Contactin 1-Overexpressing Transgenic Mice. Molecules 2019; 24:molecules24122205. [PMID: 31212848 PMCID: PMC6631041 DOI: 10.3390/molecules24122205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/03/2019] [Accepted: 06/07/2019] [Indexed: 12/18/2022] Open
Abstract
Background: In mice, postnatal immune development has previously been investigated, and evidence of a delayed maturation of the adaptive immune response has been detected. Methods: In this study, the effects of red grape polyphenol oral administration on the murine immune response were explored using pregnant mice (TAG/F3 transgenic and wild type (wt) mice) as the animal model. The study was performed during pregnancy as well as during lactation until postnatal day 8. Suckling pups from polyphenol-administered dams as well as day 30 post-weaning pups (dietary-administered with polyphenols) were used. Polyphenol effects were evaluated, measuring splenic cytokine secretion. Results: Phorbol myristate acetate-activated splenocytes underwent the highest cytokine production at day 30 in both wt and TAG/F3 mice. In the latter, release of interferon (IFN)-γ and tumor necrosis factor (TNF)-α was found to be higher than in the wt counterpart. In this context, polyphenols exerted modulating activities on day 30 TAG/F3 mice, inducing release of interleukin (IL)-10 in hetero mice while abrogating release of IL-2, IFN-γ, TNF-α, IL-6, and IL-4 in homo and hetero mice. Conclusion: Polyphenols are able to prevent the development of an inflammatory/allergic profile in postnatal TAG/F3 mice.
Collapse
|
21
|
Rippe C, Albinsson S, Guron G, Nilsson H, Swärd K. Targeting transcriptional control of soluble guanylyl cyclase via NOTCH for prevention of cardiovascular disease. Acta Physiol (Oxf) 2019; 225:e13094. [PMID: 29754438 DOI: 10.1111/apha.13094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 12/18/2022]
Abstract
Soluble guanylyl cyclase (sGC) is an effector enzyme of nitric oxide (NO). Recent work has unravelled how levels of this enzyme are controlled, and highlighted a role in vascular disease. We provide a timely summary of available knowledge on transcriptional regulation of sGC, including influences from the NOTCH signalling pathway and genetic variants. It is speculated that hypertension-induced repression of sGC starts a vicious circle that can be initiated by periods of stress, diet or genetic factors, and a key tenet is that reduction in sGC further raises blood pressure. The idea that dysregulation of sGC contributes to syndromes caused by defective NOTCH signalling is advanced, and we discuss drug repositioning for vascular disease prevention. The advantage of targeting sGC expression rather than activity is also considered. It is argued that transcriptional inputs on sGC arise from interactions with other cells, the extracellular matrix and microRNAs (miRNAs), and concluded that the promise of sGC as a target for prevention of cardiovascular disease has increased in recent time.
Collapse
Affiliation(s)
- C. Rippe
- Department of Experimental Medical Science; Lund University; Lund Sweden
| | - S. Albinsson
- Department of Experimental Medical Science; Lund University; Lund Sweden
| | - G. Guron
- Department of Physiology; University of Gothenburg; Gothenburg Sweden
| | - H. Nilsson
- Department of Physiology; University of Gothenburg; Gothenburg Sweden
| | - K. Swärd
- Department of Experimental Medical Science; Lund University; Lund Sweden
| |
Collapse
|
22
|
Chen YA, Lu IL, Tsai JW. Contactin-1/F3 Regulates Neuronal Migration and Morphogenesis Through Modulating RhoA Activity. Front Mol Neurosci 2018; 11:422. [PMID: 30515076 PMCID: PMC6255823 DOI: 10.3389/fnmol.2018.00422] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/30/2018] [Indexed: 01/06/2023] Open
Abstract
During neocortical development, newborn neurons migrate along radial fibers from the germinal ventricular zone (VZ) toward the cortical plate (CP) to populate the cerebral cortex. This radial migration requires adhesion activities between neurons and radial fibers; however, past research has identified only a limited number of adhesion molecules involved in this process. Contactin-1/F3 (Cntn1), a cell adhesion molecule expressed in the developing nervous system is essential for many key developmental events including neural cell adhesion, neurite outgrowth, axon guidance and myelination. However, the potential role of Cntn1 in neuronal migration during cortical development has not been investigated. Here we used in utero electroporation to introduce short hairpin RNA (shRNA) to knock down (KD) Cntn1 in neural stem cells in vivo. We found that Cntn1 KD led to a delay in neuronal migration. The arrested cells presented abnormal morphology in their leading process and more multipolar cells were observed in the deep layers of the brain, suggestive of dysregulation in process formation. Intriguingly, Cntn1 KD also resulted in upregulation of RhoA, a negative regulator for neuronal migration. Interference of RhoA by expression of the dominant-negative RhoAN19 partially rescued the neuronal migration defects caused by Cntn1 KD. Our results showed that Cntn1 is a novel adhesion protein that is essential for neuronal migration and regulates process formation of newborn cortical neurons through modulating RhoA signaling pathway.
Collapse
Affiliation(s)
- Yi-An Chen
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - I-Ling Lu
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Jin-Wu Tsai
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan.,Brain Research Center, National Yang-Ming University, Taipei, Taiwan.,Biophotonics and Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
23
|
PLP1 and CNTN1 gene variation modulates the microstructure of human white matter in the corpus callosum. Brain Struct Funct 2018; 223:3875-3887. [DOI: 10.1007/s00429-018-1729-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 08/03/2018] [Indexed: 12/27/2022]
|
24
|
Lu IL, Chen C, Tung CY, Chen HH, Pan JP, Chang CH, Cheng JS, Chen YA, Wang CH, Huang CW, Kang YN, Chang HY, Li LL, Chang KP, Shih YH, Lin CH, Kwan SY, Tsai JW. Identification of genes associated with cortical malformation using a transposon-mediated somatic mutagenesis screen in mice. Nat Commun 2018; 9:2498. [PMID: 29950674 PMCID: PMC6021418 DOI: 10.1038/s41467-018-04880-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022] Open
Abstract
Mutations in genes involved in the production, migration, or differentiation of cortical neurons often lead to malformations of cortical development (MCDs). However, many genetic mutations involved in MCD pathogenesis remain unidentified. Here we developed a genetic screening paradigm based on transposon-mediated somatic mutagenesis by in utero electroporation and the inability of mutant neuronal precursors to migrate to the cortex and identified 33 candidate MCD genes. Consistent with the screen, several genes have already been implicated in neural development and disorders. Functional disruption of the candidate genes by RNAi or CRISPR/Cas9 causes altered neuronal distributions that resemble human cortical dysplasia. To verify potential clinical relevance of these candidate genes, we analyzed somatic mutations in brain tissue from patients with focal cortical dysplasia and found that mutations are enriched in these candidate genes. These results demonstrate that this approach is able to identify potential mouse genes involved in cortical development and MCD pathogenesis. Cortical malformations have a variety of causes. Here the authors use transposon mutagenesis to insert mutations into neural stem cells in the developing mouse cortex to screen for new candidate genes for cortical malformation, and validate some targets in human brain tissue.
Collapse
Affiliation(s)
- I-Ling Lu
- Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan
| | - Chien Chen
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,National Yang-Ming University School of Medicine, Taipei, 112, Taiwan
| | - Chien-Yi Tung
- VYM Genome Research Center of National Yang-Ming University, Taipei, 112, Taiwan.,Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Hsin-Hung Chen
- National Yang-Ming University School of Medicine, Taipei, 112, Taiwan.,Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Jia-Ping Pan
- VYM Genome Research Center of National Yang-Ming University, Taipei, 112, Taiwan
| | - Chia-Hsiang Chang
- Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan.,Taiwan International Graduate Program (TIGP) in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, 112, Taiwan
| | - Jia-Shing Cheng
- Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan
| | - Yi-An Chen
- Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan
| | - Chun-Hung Wang
- Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan
| | - Chia-Wei Huang
- Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan
| | - Yi-Ning Kang
- Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan
| | - Hsin-Yun Chang
- Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan
| | - Lei-Li Li
- Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan
| | - Kai-Ping Chang
- National Yang-Ming University School of Medicine, Taipei, 112, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Yang-Hsin Shih
- National Yang-Ming University School of Medicine, Taipei, 112, Taiwan.,Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Chi-Hung Lin
- VYM Genome Research Center of National Yang-Ming University, Taipei, 112, Taiwan.,Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, 112, Taiwan.,Institute of Biophotonics, National Yang-Ming University, Taipei, 112, Taiwan
| | - Shang-Yeong Kwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,National Yang-Ming University School of Medicine, Taipei, 112, Taiwan
| | - Jin-Wu Tsai
- Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan. .,Brain Research Center, National Yang-Ming University, Taipei, 112, Taiwan. .,Biophotonics and Molecular Imaging Research Center, National Yang-Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
25
|
Chen N, He S, Geng J, Song ZJ, Han PH, Qin J, Zhao Z, Song YC, Wang HX, Dang CX. Overexpression of Contactin 1 promotes growth, migration and invasion in Hs578T breast cancer cells. BMC Cell Biol 2018; 19:5. [PMID: 29673312 PMCID: PMC5907708 DOI: 10.1186/s12860-018-0154-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 02/28/2018] [Indexed: 01/06/2023] Open
Abstract
Background Contactin1 (CNTN1) has been shown to play an important role in the invasion and metastasis of several tumors; however, the role of CNTN1 in breast cancer has not been fully studied. The purpose of this study is to investigate the role of CNTN1 in regulating tumor growth, migration and invasion in breast cancer. Results To investigate its function, CNTN1 was expressed in Hs578T cells. CNTN1 expression was confirmed by western blot, immunohistochemistry and real-time RT-PCR. The effect of CNTN1 overexpression on proliferation, migration and invasion of Hs578T breast cancer cells was assessed in vitro and in vivo. Our results showed that CNTN1 overexpression promoted Hs578T cell proliferation, cell cycle progression, colony formation, invasion and migration. Notably, overexpression of CNTN1 in Hs578T cells enhanced the growth of mouse xenograft tumors. Conclusions CNTN1 promotes growth, metastasis and invasion of Hs578T breast cancer cell line. Thus, therapies targeting CNTN1 may prove efficacious for breast cancer. However, further investigation is required to understand the mechanism by which CNTN1 influences proliferation, metastasis and invasion in breast cancer. Electronic supplementary material The online version of this article (10.1186/s12860-018-0154-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nan Chen
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Sai He
- Department of Breast Cancer, Shaanxi Provincial Tumor Hospital, Xi'an, Shaanxi, China
| | - Jie Geng
- Department of Peripheral Vascular Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhang-Jun Song
- Department of Breast Cancer, Shaanxi Provincial Tumor Hospital, Xi'an, Shaanxi, China
| | - Pi-Hua Han
- Department of Breast Cancer, Shaanxi Provincial Tumor Hospital, Xi'an, Shaanxi, China
| | - Juan Qin
- Department of Breast Cancer, Shaanxi Provincial Tumor Hospital, Xi'an, Shaanxi, China
| | - Zheng Zhao
- Department of Medical Oncology, Shaanxi Provincial Tumor Hospital, Xi'an, Shaanxi, China
| | - Yong-Chun Song
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hu-Xia Wang
- Department of Breast Cancer, Shaanxi Provincial Tumor Hospital, Xi'an, Shaanxi, China
| | - Cheng-Xue Dang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
26
|
Velásquez E, Nogueira FCS, Velásquez I, Schmitt A, Falkai P, Domont GB, Martins-de-Souza D. Synaptosomal Proteome of the Orbitofrontal Cortex from Schizophrenia Patients Using Quantitative Label-Free and iTRAQ-Based Shotgun Proteomics. J Proteome Res 2017; 16:4481-4494. [PMID: 28949146 DOI: 10.1021/acs.jproteome.7b00422] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Schizophrenia is a chronic and incurable neuropsychiatric disorder that affects about one percent of the world population. The proteomic characterization of the synaptosome fraction of the orbitofrontal cortex is useful for providing valuable information about the molecular mechanisms of synaptic functions in these patients. Quantitative analyses of synaptic proteins were made with eight paranoid schizophrenia patients and a pool of eight healthy controls free of mental diseases. Label-free and iTRAQ labeling identified a total of 2018 protein groups. Statistical analyses revealed 12 and 55 significantly dysregulated proteins by iTRAQ and label-free, respectively. Quantitative proteome analyses showed an imbalance in the calcium signaling pathway and proteins such as reticulon-1 and cytochrome c, related to endoplasmic reticulum stress and programmed cell death. Also, it was found that there is a significant increase in limbic-system-associated membrane protein and α-calcium/calmodulin-dependent protein kinase II, associated with the regulation of human behavior. Our data contribute to a better understanding about apoptosis as a possible pathophysiological mechanism of this disease as well as neural systems supporting social behavior in schizophrenia. This study also is a joint effort of the Chr 15 C-HPP team and the Human Brain Proteome Project of B/D-HPP. All MS proteomics data are deposited in the ProteomeXchange Repository under PXD006798.
Collapse
Affiliation(s)
- Erika Velásquez
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-909 Rio de Janeiro, Brazil
| | - Fabio C S Nogueira
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-909 Rio de Janeiro, Brazil.,Laboratory of Proteomics, LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-598 Rio de Janeiro, Brazil
| | | | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University of Munich (LMU) , 80336 Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University of Munich (LMU) , 80336 Munich, Germany
| | - Gilberto B Domont
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro , Rio de Janeiro, 21941-909 Rio de Janeiro, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry, Institute of Biology, University of Campinas (UNICAMP) , Campinas, 13083-862 São Paulo, Brazil.,UNICAMP's Neurobiology Center , Campinas, 13083-888 São Paulo, Brazil.,Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico , São Paulo, 01060-970 São Paulo, Brazil
| |
Collapse
|
27
|
Tilley SK, Joseph RM, Kuban KCK, Dammann OU, O’Shea TM, Fry RC. Genomic biomarkers of prenatal intrauterine inflammation in umbilical cord tissue predict later life neurological outcomes. PLoS One 2017; 12:e0176953. [PMID: 28493900 PMCID: PMC5426658 DOI: 10.1371/journal.pone.0176953] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/19/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Preterm birth is a major risk factor for neurodevelopmental delays and disorders. This study aimed to identify genomic biomarkers of intrauterine inflammation in umbilical cord tissue in preterm neonates that predict cognitive impairment at 10 years of age. STUDY DESIGN Genome-wide messenger RNA (mRNA) levels from umbilical cord tissue were obtained from 43 neonates born before 28 weeks of gestation. Genes that were differentially expressed across four indicators of intrauterine inflammation were identified and their functions examined. Exact logistic regression was used to test whether expression levels in umbilical cord tissue predicted neurocognitive function at 10 years of age. RESULTS Placental indicators of inflammation were associated with changes in the mRNA expression of 445 genes in umbilical cord tissue. Transcripts with decreased expression showed significant enrichment for biological signaling processes related to neuronal development and growth. The altered expression of six genes was found to predict neurocognitive impairment when children were 10 years old These genes include two that encode for proteins involved in neuronal development. CONCLUSION Prenatal intrauterine inflammation is associated with altered gene expression in umbilical cord tissue. A set of six of the differentially expressed genes predict cognitive impairment later in life, suggesting that the fetal environment is associated with significant adverse effects on neurodevelopment that persist into later childhood.
Collapse
Affiliation(s)
- Sloane K. Tilley
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Robert M. Joseph
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Karl C. K. Kuban
- Department of Pediatrics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Olaf U. Dammann
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - T. Michael O’Shea
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
28
|
Wu ZQ, Li D, Huang Y, Chen XP, Huang W, Liu CF, Zhao HQ, Xu RX, Cheng M, Schachner M, Ma QH. Caspr Controls the Temporal Specification of Neural Progenitor Cells through Notch Signaling in the Developing Mouse Cerebral Cortex. Cereb Cortex 2017; 27:1369-1385. [PMID: 26740489 DOI: 10.1093/cercor/bhv318] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The generation of layer-specific neurons and astrocytes by radial glial cells during development of the cerebral cortex follows a precise temporal sequence, which is regulated by intrinsic and extrinsic factors. The molecular mechanisms controlling the timely generation of layer-specific neurons and astrocytes remain not fully understood. In this study, we show that the adhesion molecule contactin-associated protein (Caspr), which is involved in the maintenance of the polarized domains of myelinated axons, is essential for the timing of generation of neurons and astrocytes in the developing mouse cerebral cortex. Caspr is expressed by radial glial cells, which are neural progenitor cells that generate both neurons and astrocytes. Absence of Caspr in neural progenitor cells delays the production cortical neurons and induces precocious formation of cortical astrocytes, without affecting the numbers of progenitor cells. At the molecular level, Caspr cooperates with the intracellular domain of Notch to repress transcription of the Notch effector Hes1. Suppression of Notch signaling via a Hes1 shRNA rescues the abnormal neurogenesis and astrogenesis in Caspr-deficient mice. These findings establish Caspr as a novel key regulator that controls the temporal specification of cell fate in radial glial cells of the developing cerebral cortex through Notch signaling.
Collapse
Affiliation(s)
- Zhi-Qiang Wu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Di Li
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Ya Huang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Xi-Ping Chen
- Department of Forensic Medicine, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Wenhui Huang
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg D-66421, Germany
| | - Chun-Feng Liu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - He-Qing Zhao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Ru-Xiang Xu
- Affiliated Bayi Brain Hospital, Beijing Military Hospital, Southern Medical University, Beijing 100070, China
| | - Mei Cheng
- Binzhou Medical University, Yantai, Shandong Province 264000, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province 515041, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| |
Collapse
|
29
|
Gulisano W, Bizzoca A, Gennarini G, Palmeri A, Puzzo D. Role of the adhesion molecule F3/Contactin in synaptic plasticity and memory. Mol Cell Neurosci 2016; 81:64-71. [PMID: 28038945 DOI: 10.1016/j.mcn.2016.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 12/07/2016] [Accepted: 12/22/2016] [Indexed: 12/14/2022] Open
Abstract
Cell adhesion molecules (CAMs) have a pivotal role in building and maintaining synaptic structures during brain development participating in axonal elongation and pathfinding, glial guidance of neuronal migration, as well as myelination. CAMs expression persists in the adult brain particularly in structures undergoing postnatal neurogenesis and involved in synaptic plasticity and memory as the hippocampus. Among the neural CAMs, we have recently focused on F3/Contactin, a glycosylphosphatidyl inositol-anchored glycoprotein belonging to the immunoglobulin superfamily, involved in neuronal development, synaptic maintenance and organization of neuronal networks. Here, we discuss our recent data suggesting that F3/Contactin exerts a role in hippocampal synaptic plasticity and memory in adult and aged mice. In particular, we have studied long-term potentiation (LTP), spatial and object recognition memory, and phosphorylation of the transcription factor cAMP-Responsive-Element Binding protein (CREB) in a transgenic mouse model of F3/Contactin overexpression. We also investigated whether F3/Contactin might influence neuronal apoptosis and the production of amyloid-beta peptide (Aβ), known to be one of the main pathogenetic hallmarks of Alzheimer's disease (AD). In conclusion, a further understanding of F3/Contactin role in synaptic plasticity and memory might have interesting clinical outcomes in cognitive disorders, such as aging and AD, offering innovative therapeutic opportunities.
Collapse
Affiliation(s)
- Walter Gulisano
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonella Bizzoca
- Section of Physiology, Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Gianfranco Gennarini
- Section of Physiology, Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Agostino Palmeri
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| | - Daniela Puzzo
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
30
|
Ocklenburg S, Gerding WM, Arning L, Genç E, Epplen JT, Güntürkün O, Beste C. Myelin Genes and the Corpus Callosum: Proteolipid Protein 1 (PLP1) and Contactin 1 (CNTN1) Gene Variation Modulates Interhemispheric Integration. Mol Neurobiol 2016; 54:7908-7916. [DOI: 10.1007/s12035-016-0285-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/02/2016] [Indexed: 01/06/2023]
|
31
|
Gennarini G, Bizzoca A, Picocci S, Puzzo D, Corsi P, Furley AJW. The role of Gpi-anchored axonal glycoproteins in neural development and neurological disorders. Mol Cell Neurosci 2016; 81:49-63. [PMID: 27871938 DOI: 10.1016/j.mcn.2016.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/10/2016] [Accepted: 11/14/2016] [Indexed: 01/06/2023] Open
Abstract
This review article focuses on the Contactin (CNTN) subset of the Immunoglobulin supergene family (IgC2/FNIII molecules), whose components share structural properties (the association of Immunoglobulin type C2 with Fibronectin type III domains), as well as a general role in cell contact formation and axonal growth control. IgC2/FNIII molecules include 6 highly related components (CNTN 1-6), associated with the cell membrane via a Glycosyl Phosphatidyl Inositol (GPI)-containing lipid tail. Contactin 1 and Contactin 2 share ~50 (49.38)% identity at the aminoacid level. They are components of the cell surface, from which they may be released in soluble forms. They bind heterophilically to multiple partners in cis and in trans, including members of the related L1CAM family and of the Neurexin family Contactin-associated proteins (CNTNAPs or Casprs). Such interactions are important for organising the neuronal membrane, as well as for modulating the growth and pathfinding of axon tracts. In addition, they also mediate the functional maturation of axons by promoting their interactions with myelinating cells at the nodal, paranodal and juxtaparanodal regions. Such interactions also mediate differential ionic channels (both Na+ and K+) distribution, which is of critical relevance in the generation of the peak-shaped action potential. Indeed, thanks to their interactions with Ankyrin G, Na+ channels map within the nodal regions, where they drive axonal depolarization. However, no ionic channels are found in the flanking Contactin1-containing paranodal regions, where CNTN1 interactions with Caspr1 and with the Ig superfamily component Neurofascin 155 in cis and in trans, respectively, build a molecular barrier between the node and the juxtaparanode. In this region K+ channels are clustered, depending upon molecular interactions with Contactin 2 and with Caspr2. In addition to these functions, the Contactins appear to have also a role in degenerative and inflammatory disorders: indeed Contactin 2 is involved in neurodegenerative disorders with a special reference to the Alzheimer disease, given its ability to work as a ligand of the Alzheimer Precursor Protein (APP), which results in increased Alzheimer Intracellular Domain (AICD) release in a γ-secretase-dependent manner. On the other hand Contactin 1 drives Notch signalling activation via the Hes pathway, which could be consistent with its ability to modulate neuroinflammation events, and with the possibility that Contactin 1-dependent interactions may participate to the pathogenesis of the Multiple Sclerosis and of other inflammatory disorders.
Collapse
Affiliation(s)
- Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy.
| | - Antonella Bizzoca
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Sabrina Picocci
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - Patrizia Corsi
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Andrew J W Furley
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2NT, UK
| |
Collapse
|
32
|
Frei JA, Stoeckli ET. SynCAMs - From axon guidance to neurodevelopmental disorders. Mol Cell Neurosci 2016; 81:41-48. [PMID: 27594578 DOI: 10.1016/j.mcn.2016.08.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 08/28/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022] Open
Abstract
Many cell adhesion molecules are located at synapses but only few of them can be considered synaptic cell adhesion molecules in the strict sense. Besides the Neurexins and Neuroligins, the LRRTMs (leucine rich repeat transmembrane proteins) and the SynCAMs/CADMs can induce synapse formation when expressed in non-neuronal cells and therefore are true synaptic cell adhesion molecules. SynCAMs (synaptic cell adhesion molecules) are a subfamily of the immunoglobulin superfamily of cell adhesion molecules. As suggested by their name, they were first identified as cell adhesion molecules at the synapse which were sufficient to trigger synapse formation. They also contribute to myelination by mediating axon-glia cell contacts. More recently, their role in earlier stages of neural circuit formation was demonstrated, as they also guide axons both in the peripheral and in the central nervous system. Mutations in SynCAM genes were found in patients diagnosed with autism spectrum disorders. The diverse functions of SynCAMs during development suggest that neurodevelopmental disorders are not only due to defects in synaptic plasticity. Rather, early steps of neural circuit formation are likely to contribute.
Collapse
Affiliation(s)
- Jeannine A Frei
- Hussman Institute for Autism, 801 W Baltimore Street, Baltimore, MD 20201, United States
| | - Esther T Stoeckli
- Dept of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
33
|
Wang J, Huang Y, Zhang J, Wei Y, Mahoud S, Bakheet AMH, Wang L, Zhou S, Tang J. Pathway-related molecules of VEGFC/D-VEGFR3/NRP2 axis in tumor lymphangiogenesis and lymphatic metastasis. Clin Chim Acta 2016; 461:165-71. [PMID: 27527412 DOI: 10.1016/j.cca.2016.08.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/07/2016] [Accepted: 08/11/2016] [Indexed: 12/12/2022]
Abstract
Precondition for tumor lymphatic metastasis is that tumor cells induce formation of original and newborn lymphatic vessels and invade surrounding lymphatic vessels in tumor stroma, while some pathway-related molecules play an important role in mechanisms associated with proliferation and migration of lymphatic endothelial cells (LECs) and tumor cells. In lymphangiogenesis and lymphatic metastasis, the pathway-related molecules of VEGFC/D-VEGFR3/NRP2 axis, such as Furin-like enzyme, CNTN1, Prox1, LYVE-1, Podoplanin, SOX18, SDF1 and CXCR4, are direct constitutors as a portion of VEGFC/D-VEGFR3/NRP2 axis, and their biological activities rely on this ligand-receptor system. These axis-related signal molecules could gradually produce waterfall-like cascading effects, mediate differentiation and maturation of LECs, remodel original and neonatal lymphatic vessels, as well as ultimately promote tumor cell chemotaxis, migration, invasion and metastasis to lymphoid tracts. This review summarizes the structure and function features of pathway-related molecules of VEGFC/D-VEGFR3/NRP2 axis, the expression changes of these molecules in different anatomic organs or histopathologic types or development stages of various tumors, the characteristics of transduction, implementation, integration of signal networks, the interactive effects on biological behaviors between tumor cells and lymphatic endothelial cells, and their molecular mechanisms and significances in tumor lymphangiogenesis and lymphatic metastasis.
Collapse
Affiliation(s)
- Jingwen Wang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, Liaoning 116044, China
| | - Yuhong Huang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, Liaoning 116044, China
| | - Jun Zhang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, Liaoning 116044, China
| | - Yuanyi Wei
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, Liaoning 116044, China
| | - Salma Mahoud
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, Liaoning 116044, China
| | - Ahmed Musa Hago Bakheet
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, Liaoning 116044, China
| | - Li Wang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, Liaoning 116044, China
| | - Shuting Zhou
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, Liaoning 116044, China
| | - Jianwu Tang
- Department of Pathology, Dalian Medical University, Key Laboratory for Tumor Metastasis and Intervention of Liaoning Province, 9 West, Lvshun Southern Road, Dalian, Liaoning 116044, China.
| |
Collapse
|
34
|
Costa D, Castelo R. Umbilical cord gene expression reveals the molecular architecture of the fetal inflammatory response in extremely preterm newborns. Pediatr Res 2016; 79:473-81. [PMID: 26539667 PMCID: PMC4823644 DOI: 10.1038/pr.2015.233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 08/17/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND The fetal inflammatory response (FIR) in placental membranes to an intrauterine infection often precedes premature birth raising neonatal mortality and morbidity. However, the precise molecular events behind FIR still remain largely unknown, and little has been investigated at gene expression level. METHODS We collected publicly available microarray expression data profiling umbilical cord (UC) tissue derived from the cohort of extremely low gestational age newborns (ELGANs) and interrogate them for differentially expressed (DE) genes between FIR and non-FIR-affected ELGANs. RESULTS We found a broad and complex FIR UC gene expression signature, changing up to 19% (3,896/20,155) of all human genes at 1% false discovery rate. Significant changes of a minimum 50% magnitude (1,097/3,896) affect the upregulation of many inflammatory pathways and molecules, such as cytokines, toll-like receptors, and calgranulins. Remarkably, they also include the downregulation of neurodevelopmental pathways and genes, such as Fragile-X mental retardation 1 (FMR1), contactin 1 (CNTN1), and adenomatous polyposis coli (APC). CONCLUSION The FIR expression signature in UC tissue contains molecular clues about signaling pathways that trigger FIR, and it is consistent with an acute inflammatory response by fetal innate and adaptive immune systems, which participate in the pathogenesis of neonatal brain damage.
Collapse
Affiliation(s)
- Daniel Costa
- Department of Pediatrics, Hospital de Figueres, Figueres, Spain
| | - Robert Castelo
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain,Research Program on Biomedical Informatics, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain,()
| |
Collapse
|
35
|
Yan J, Ojo D, Kapoor A, Lin X, Pinthus JH, Aziz T, Bismar TA, Wei F, Wong N, De Melo J, Cutz JC, Major P, Wood G, Peng H, Tang D. Neural Cell Adhesion Protein CNTN1 Promotes the Metastatic Progression of Prostate Cancer. Cancer Res 2016; 76:1603-14. [PMID: 26795349 DOI: 10.1158/0008-5472.can-15-1898] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 12/28/2015] [Indexed: 11/16/2022]
Abstract
Prostate cancer metastasis is the main cause of disease-related mortality. Elucidating the mechanisms underlying prostate cancer metastasis is critical for effective therapeutic intervention. In this study, we performed gene-expression profiling of prostate cancer stem-like cells (PCSC) derived from DU145 human prostate cancer cells to identify factors involved in metastatic progression. Our studies revealed contactin 1 (CNTN1), a neural cell adhesion protein, to be a prostate cancer-promoting factor. CNTN1 knockdown reduced PCSC-mediated tumor initiation, whereas CNTN1 overexpression enhanced prostate cancer cell invasion in vitro and promoted xenograft tumor formation and lung metastasis in vivo. In addition, CNTN1 overexpression in DU145 cells and corresponding xenograft tumors resulted in elevated AKT activation and reduced E-cadherin (CDH1) expression. CNTN1 expression was not readily detected in normal prostate glands, but was clearly evident on prostate cancer cells in primary tumors and lymph node and bone metastases. Tumors from 637 patients expressing CNTN1 were associated with prostate cancer progression and worse biochemical recurrence-free survival following radical prostatectomy (P < 0.05). Collectively, our findings demonstrate that CNTN1 promotes prostate cancer progression and metastasis, prompting further investigation into the mechanisms that enable neural proteins to become aberrantly expressed in non-neural malignancies.
Collapse
Affiliation(s)
- Judy Yan
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Diane Ojo
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Anil Kapoor
- Department of Surgery, McMaster University, Hamilton, Canada
| | - Xiaozeng Lin
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | | | - Tariq Aziz
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Tarek A Bismar
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Canada
| | - Fengxiang Wei
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada. The Genetics Laboratory, Institute of Women and Children's Health, Longgang District, Shenzhen, China
| | - Nicholas Wong
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Jason De Melo
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Jean-Claude Cutz
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Pierre Major
- Department of Oncology, McMaster University, Hamilton, Canada
| | - Geoffrey Wood
- Department of Veterinary Pathology, University of Guelph, Guelph, Canada
| | - Hao Peng
- Department of Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, Canada
| | - Damu Tang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada.
| |
Collapse
|
36
|
Chen DH, Yu JW, Jiang BJ. Roles of contactin-1 in solid tumors. Shijie Huaren Xiaohua Zazhi 2015; 23:4785-4791. [DOI: 10.11569/wcjd.v23.i30.4785] [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
The neural cell adhesion molecule contactin-1 (CNTN1), first identified as a member of the contactin subpopulation of the immunoglobulin superfamily, is associated with many other cell surface proteins expressed on a variety of neurocytes, contributing to their functions and maturation. It has been recently found that the abnormal expression of CNTN1 has a close correlation with tumor initiation, development, invasiveness, metastasis and prognosis. The acquired metastatic ability of malignant tumors is caused by a population of cancer cells with the capacities of invasiveness, metastasis, adherence and proliferation, in which abnormal gene expression may play an important role. This review focuses on the current advances in research of CNTN1 in the nerve system, and mainly in the malignant tumors, with an aim to provide new clues to clinical prevention, diagnosis and treatment of these malignancies.
Collapse
|
37
|
Chen DH, Yu JW, Jiang BJ. Contactin 1: A potential therapeutic target and biomarker in gastric cancer. World J Gastroenterol 2015; 21:9707-9716. [PMID: 26361417 PMCID: PMC4562954 DOI: 10.3748/wjg.v21.i33.9707] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/02/2015] [Accepted: 07/15/2015] [Indexed: 02/06/2023] Open
Abstract
Despite advances in diagnosis and treatment, gastric cancer remains one of the most common malignant tumors worldwide, and early diagnosis remains a challenge. The lack of effective methods to detect these tumors early is a major factor contributing to the high mortality in patients with gastric cancer, who are typically diagnosed at an advanced stage. Additionally, the early detection of metastases and the curative treatment of gastric cancer are difficult to achieve, and the detailed mechanisms remain to be fully elucidated. Thus, the identification of valuable predictive biomarkers and therapeutic targets to improve the prognosis of patients with gastric cancer is becoming increasingly important. Contactin 1 (CNTN1), a cell adhesion molecule, is a glycosylphosphatidylinositol-anchored neuronal membrane protein that plays an important role in cancer progression. The expression of CNTN1 is upregulated in primary lesions, and its expression level correlates with tumor metastasis in cancer patients. The current evidence reveals that the functions of CNTN1 in the development and progression of cancer likely promote the invasion and metastasis of cancer cells via the VEGFC/FLT4 axis, the RHOA-dependent pathway, the Notch signaling pathway and the epithelial-mesenchymal transition progression. Therefore, CNTN1 may be a novel biomarker and a possible therapeutic target in cancer treatment in the near future.
Collapse
|
38
|
Bizzoca A, Picocci S, Corsi P, Arbia S, Croci L, Consalez GG, Gennarini G. The gene encoding the mouse contactin-1 axonal glycoprotein is regulated by the collier/Olf1/EBF family early B-Cell factor 2 transcription factor. Dev Neurobiol 2015; 75:1420-40. [DOI: 10.1002/dneu.22293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/17/2015] [Accepted: 03/22/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Antonella Bizzoca
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs; Medical School, University of Bari; Policlinico Bari I-70124 Italy
| | - Sabrina Picocci
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs; Medical School, University of Bari; Policlinico Bari I-70124 Italy
| | - Patrizia Corsi
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs; Medical School, University of Bari; Policlinico Bari I-70124 Italy
| | - Stefania Arbia
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs; Medical School, University of Bari; Policlinico Bari I-70124 Italy
| | - Laura Croci
- Division of Neuroscience; San Raffaele Scientific Institute; Milano I-20132 Italy
| | - G. Giacomo Consalez
- Division of Neuroscience; San Raffaele Scientific Institute; Milano I-20132 Italy
- Università Vita-Salute San Raffaele; Milano I-20132 Italy
| | - Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs; Medical School, University of Bari; Policlinico Bari I-70124 Italy
| |
Collapse
|
39
|
Puzzo D, Bizzoca A, Loreto C, Guida CA, Gulisano W, Frasca G, Bellomo M, Castorina S, Gennarini G, Palmeri A. Role of F3/contactin expression profile in synaptic plasticity and memory in aged mice. Neurobiol Aging 2015; 36:1702-1715. [PMID: 25659859 DOI: 10.1016/j.neurobiolaging.2015.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 12/30/2014] [Accepted: 01/03/2015] [Indexed: 12/14/2022]
Abstract
We have recently shown that overexpression of the F3/contactin adhesive glycoprotein (also known as Contactin-1) promotes neurogenesis in adult hippocampus, which correlates with improved synaptic plasticity and memory. Because F3/contactin levels physiologically decrease with age, here, we aim at investigating whether its overexpression might counteract the cognitive decline in aged animals. For this we use 20- to 24-month-old TAG/F3 transgenic mice in which F3/contactin overexpression is driven by regulatory sequences from the gene encoding the transient axonal glycoprotein TAG-1 throughout development. We show that aged TAG/F3 mice display improved hippocampal long-term potentiation and memory compared with wild-type littermates. The same mice undergo a decrease of neuronal apoptosis at the hippocampal level, which correlated to a decrease of active caspase-3; by contrast, procaspase-3 and Bax as well as the anti-apoptotic and plasticity-related pathway BDNF/CREB/Bcl-2 were rather increased. Interestingly, amyloid-precursor protein processing was shifted toward sAPPα generation, with a decrease of sAPPβ and amyloid-beta levels. Our data confirm that F3/contactin plays a role in hippocampal synaptic plasticity and memory also in aged mice, suggesting that it acts on molecular pathways related to apoptosis and amyloid-beta production.
Collapse
Affiliation(s)
- Daniela Puzzo
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonella Bizzoca
- Section of Physiology, Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Bari, Italy
| | - Carla Loreto
- Section of Anatomy, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Chiara A Guida
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Walter Gulisano
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppina Frasca
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Maria Bellomo
- Faculty of Psychology and Educational Sciences, University "Kore", Enna, Italy
| | - Sergio Castorina
- Section of Anatomy, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Gianfranco Gennarini
- Section of Physiology, Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Bari, Italy.
| | - Agostino Palmeri
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
40
|
Butts T, Green MJ, Wingate RJT. Development of the cerebellum: simple steps to make a 'little brain'. Development 2014; 141:4031-41. [PMID: 25336734 DOI: 10.1242/dev.106559] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cerebellum is a pre-eminent model for the study of neurogenesis and circuit assembly. Increasing interest in the cerebellum as a participant in higher cognitive processes and as a locus for a range of disorders and diseases make this simple yet elusive structure an important model in a number of fields. In recent years, our understanding of some of the more familiar aspects of cerebellar growth, such as its territorial allocation and the origin of its various cell types, has undergone major recalibration. Furthermore, owing to its stereotyped circuitry across a range of species, insights from a variety of species have contributed to an increasingly rich picture of how this system develops. Here, we review these recent advances and explore three distinct aspects of cerebellar development - allocation of the cerebellar anlage, the significance of transit amplification and the generation of neuronal diversity - each defined by distinct regulatory mechanisms and each with special significance for health and disease.
Collapse
Affiliation(s)
- Thomas Butts
- MRC Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, UK
| | - Mary J Green
- National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Richard J T Wingate
- MRC Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK
| |
Collapse
|
41
|
Rolfe RA, Nowlan NC, Kenny EM, Cormican P, Morris DW, Prendergast PJ, Kelly D, Murphy P. Identification of mechanosensitive genes during skeletal development: alteration of genes associated with cytoskeletal rearrangement and cell signalling pathways. BMC Genomics 2014; 15:48. [PMID: 24443808 PMCID: PMC3905281 DOI: 10.1186/1471-2164-15-48] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 12/18/2013] [Indexed: 12/15/2022] Open
Abstract
Background Mechanical stimulation is necessary for regulating correct formation of the skeleton. Here we test the hypothesis that mechanical stimulation of the embryonic skeletal system impacts expression levels of genes implicated in developmentally important signalling pathways in a genome wide approach. We use a mutant mouse model with altered mechanical stimulation due to the absence of limb skeletal muscle (Splotch-delayed) where muscle-less embryos show specific defects in skeletal elements including delayed ossification, changes in the size and shape of cartilage rudiments and joint fusion. We used Microarray and RNA sequencing analysis tools to identify differentially expressed genes between muscle-less and control embryonic (TS23) humerus tissue. Results We found that 680 independent genes were down-regulated and 452 genes up-regulated in humeri from muscle-less Spd embryos compared to littermate controls (at least 2-fold; corrected p-value ≤0.05). We analysed the resulting differentially expressed gene sets using Gene Ontology annotations to identify significant enrichment of genes associated with particular biological processes, showing that removal of mechanical stimuli from muscle contractions affected genes associated with development and differentiation, cytoskeletal architecture and cell signalling. Among cell signalling pathways, the most strongly disturbed was Wnt signalling, with 34 genes including 19 pathway target genes affected. Spatial gene expression analysis showed that both a Wnt ligand encoding gene (Wnt4) and a pathway antagonist (Sfrp2) are up-regulated specifically in the developing joint line, while the expression of a Wnt target gene, Cd44, is no longer detectable in muscle-less embryos. The identification of 84 genes associated with the cytoskeleton that are down-regulated in the absence of muscle indicates a number of candidate genes that are both mechanoresponsive and potentially involved in mechanotransduction, converting a mechanical stimulus into a transcriptional response. Conclusions This work identifies key developmental regulatory genes impacted by altered mechanical stimulation, sheds light on the molecular mechanisms that interpret mechanical stimulation during skeletal development and provides valuable resources for further investigation of the mechanistic basis of mechanoregulation. In particular it highlights the Wnt signalling pathway as a potential point of integration of mechanical and molecular signalling and cytoskeletal components as mediators of the response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Paula Murphy
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
42
|
Okamoto M, Namba T, Shinoda T, Kondo T, Watanabe T, Inoue Y, Takeuchi K, Enomoto Y, Ota K, Oda K, Wada Y, Sagou K, Saito K, Sakakibara A, Kawaguchi A, Nakajima K, Adachi T, Fujimori T, Ueda M, Hayashi S, Kaibuchi K, Miyata T. TAG-1-assisted progenitor elongation streamlines nuclear migration to optimize subapical crowding. Nat Neurosci 2013. [PMID: 24056697 DOI: 10.1038/nn.3525.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neural progenitors exhibit cell cycle-dependent interkinetic nuclear migration (INM) along the apicobasal axis. Despite recent advances in understanding its underlying molecular mechanisms, the processes to which INM contributes mechanically and the regulation of INM by the apicobasally elongated morphology of progenitors remain unclear. We found that knockdown of the cell-surface molecule TAG-1 resulted in retraction of neocortical progenitors' basal processes. Highly shortened stem-like progenitors failed to undergo basalward INM and became overcrowded in the periventricular (subapical) space. Surprisingly, the overcrowded progenitors left the apical surface and migrated into basal neuronal territories. These observations, together with the results of in toto imaging and physical tests, suggest that progenitors may sense and respond to excessive mechanical stress. Although, unexpectedly, the heterotopic progenitors remained stem-like and continued to sequentially produce neurons until the late embryonic period, histogenesis was severely disrupted. Thus, INM is essential for preventing overcrowding of nuclei and their somata, thereby ensuring normal brain histogenesis.
Collapse
Affiliation(s)
- Mayumi Okamoto
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Okamoto M, Namba T, Shinoda T, Kondo T, Watanabe T, Inoue Y, Takeuchi K, Enomoto Y, Ota K, Oda K, Wada Y, Sagou K, Saito K, Sakakibara A, Kawaguchi A, Nakajima K, Adachi T, Fujimori T, Ueda M, Hayashi S, Kaibuchi K, Miyata T. TAG-1-assisted progenitor elongation streamlines nuclear migration to optimize subapical crowding. Nat Neurosci 2013; 16:1556-66. [PMID: 24056697 DOI: 10.1038/nn.3525] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 08/22/2013] [Indexed: 12/15/2022]
Abstract
Neural progenitors exhibit cell cycle-dependent interkinetic nuclear migration (INM) along the apicobasal axis. Despite recent advances in understanding its underlying molecular mechanisms, the processes to which INM contributes mechanically and the regulation of INM by the apicobasally elongated morphology of progenitors remain unclear. We found that knockdown of the cell-surface molecule TAG-1 resulted in retraction of neocortical progenitors' basal processes. Highly shortened stem-like progenitors failed to undergo basalward INM and became overcrowded in the periventricular (subapical) space. Surprisingly, the overcrowded progenitors left the apical surface and migrated into basal neuronal territories. These observations, together with the results of in toto imaging and physical tests, suggest that progenitors may sense and respond to excessive mechanical stress. Although, unexpectedly, the heterotopic progenitors remained stem-like and continued to sequentially produce neurons until the late embryonic period, histogenesis was severely disrupted. Thus, INM is essential for preventing overcrowding of nuclei and their somata, thereby ensuring normal brain histogenesis.
Collapse
Affiliation(s)
- Mayumi Okamoto
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Puzzo D, Bizzoca A, Privitera L, Furnari D, Giunta S, Girolamo F, Pinto M, Gennarini G, Palmeri A. F3/Contactin promotes hippocampal neurogenesis, synaptic plasticity, and memory in adult mice. Hippocampus 2013; 23:1367-82. [PMID: 23939883 DOI: 10.1002/hipo.22186] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 07/23/2013] [Accepted: 08/02/2013] [Indexed: 12/18/2022]
Abstract
F3/contactin, a cell-adhesion molecule belonging to the immunoglobulin supergene family, is involved in several aspects of neural development including synapse building, maintenance and functioning. Here, we examine F3/contactin function in adult hippocampal neurogenesis, synaptic plasticity, and memory, using as a model TAG/F3 transgenic mice, where F3/contactin overexpression was induced under control of regulatory sequences from the human TAG-1 (TAX-1) gene. Transgenic mice aged 5 (M5) and 12 (M12) months exhibited an increase in hippocampal size, which correlated with positive effects on precursor proliferation and NeuN expression, these data suggesting a possible role for F3/contactin in promoting adult hippocampal neurogenesis. On the functional level, TAG/F3 mice exhibited increased CA1 long-term potentiation and improved spatial and object recognition memory, notably at 12 months of age. Interestingly, these mice showed an increased expression of the phosphorylated transcription factor CREB, which may represent the main molecular correlate of the observed morphological and functional effects. Altogether, these findings indicate for the first time that F3/contactin plays a role in promoting adult hippocampal neurogenesis and that this effect correlates with improved synaptic function and memory.
Collapse
Affiliation(s)
- Daniela Puzzo
- Section of Physiology, Department of Bio-Medical Sciences, University of Catania, Catania, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hadas Y, Nitzan N, Furley AJW, Kozlov SV, Klar A. Distinct cis regulatory elements govern the expression of TAG1 in embryonic sensory ganglia and spinal cord. PLoS One 2013; 8:e57960. [PMID: 23469119 PMCID: PMC3582508 DOI: 10.1371/journal.pone.0057960] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 01/29/2013] [Indexed: 01/06/2023] Open
Abstract
Cell fate commitment of spinal progenitor neurons is initiated by long-range, midline-derived, morphogens that regulate an array of transcription factors that, in turn, act sequentially or in parallel to control neuronal differentiation. Included among these are transcription factors that regulate the expression of receptors for guidance cues, thereby determining axonal trajectories. The Ig/FNIII superfamily molecules TAG1/Axonin1/CNTN2 (TAG1) and Neurofascin (Nfasc) are co-expressed in numerous neuronal cell types in the CNS and PNS – for example motor, DRG and interneurons - both promote neurite outgrowth and both are required for the architecture and function of nodes of Ranvier. The genes encoding TAG1 and Nfasc are adjacent in the genome, an arrangement which is evolutionarily conserved. To study the transcriptional network that governs TAG1 and Nfasc expression in spinal motor and commissural neurons, we set out to identify cis elements that regulate their expression. Two evolutionarily conserved DNA modules, one located between the Nfasc and TAG1 genes and the second directly 5′ to the first exon and encompassing the first intron of TAG1, were identified that direct complementary expression to the CNS and PNS, respectively, of the embryonic hindbrain and spinal cord. Sequential deletions and point mutations of the CNS enhancer element revealed a 130bp element containing three conserved E-boxes required for motor neuron expression. In combination, these two elements appear to recapitulate a major part of the pattern of TAG1 expression in the embryonic nervous system.
Collapse
Affiliation(s)
- Yoav Hadas
- Dept. of medical neurobiology, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Noa Nitzan
- Dept. of medical neurobiology, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Andrew J. W. Furley
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, United Kingdom
- * E-mail: (AV); (SVK); (AJWF)
| | - Serguei V. Kozlov
- Center for Advanced Preclinical Research, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research (FNLCR), Frederick, Maryland, United States of America
- * E-mail: (AV); (SVK); (AJWF)
| | - Avihu Klar
- Dept. of medical neurobiology, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
- * E-mail: (AV); (SVK); (AJWF)
| |
Collapse
|
46
|
Massaro A, Bizzoca A, Corsi P, Pinto MF, Carratù MR, Gennarini G. Significance of F3/Contactin gene expression in cerebral cortex and nigrostriatal development. Mol Cell Neurosci 2012; 50:221-37. [PMID: 22579730 DOI: 10.1016/j.mcn.2012.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 03/19/2012] [Accepted: 05/02/2012] [Indexed: 12/31/2022] Open
Abstract
F3/Contactin is a neuronal surface glycoprotein, which plays a general role in neural development and, in particular, in neuronal and oligodendrocyte differentiation. In a previous study using the F3/EGFP transgenic mice, which express an EGFP reporter under control of the regulatory region from the mouse F3/Contactin gene, the activation of the F3/Contactin promoter was found to correlate with granule and Purkinje neuron differentiation in developing cerebellar cortex. Here we report that in developing cerebral cortex and basal ganglia the F3/Contactin gene is mostly activated during early commitment of neuronal precursors, thus indicating a region-specific profile of its developmental activation. We also report that, in the same structures of F3/EGFP mice, a downregulation of the endogenous F3/Contactin gene occurs, which correlates with upregulation of the dopaminergic phenotype and with locomotor pattern abnormalities. Therefore, F3/EGFP transgenic mice exhibit morphological and functional phenotypes recapitulating those arising from imbalance of the striatal dopaminergic pathway. As for the underlying mechanisms, we postulate that in F3/EGFP mice F3/Contactin downregulation results from the ability of transgene promoter sequences to interfere with the activation of the endogenous gene, thus realizing an F3/Contactin knockdown model, while dopaminergic upregulation is consistent with a general F3/Contactin inhibitory effect on the neuronal phenotype.
Collapse
Affiliation(s)
- Antonio Massaro
- Department of Basic Medical Sciences, Medical School, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | | | | | | | | | | |
Collapse
|