1
|
Xie B, Liang J, Jiang J, Zeng T, Liu L, Xie D, Zhu G, Xiong L, Zhang K, Liu D, Gong J, Chen X, Lai R, Xie H. Zebrafish myo7aa affects congenital hearing by regulating Rho-GTPase signaling. Front Mol Neurosci 2024; 17:1405109. [PMID: 39081296 PMCID: PMC11287254 DOI: 10.3389/fnmol.2024.1405109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/31/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction myo7aa, the homolog of the human Usher 1B syndrome pathogenic gene, myo7A, plays an important role in stereociliary development and maintenance, therefore, is critical for hearing and balance. However, the molecular mechanisms that myo7aa regulate hearing and balance still need to be studied. Methods In this study, we generated two independent zebrafish myo7aa knockout lines using CRISPR/Cas9 technology. To investigate the effects of myo7aa on hearing, YO-PRO-1 staining and startle response assay were used. To gain insight into the specific molecular mechanisms by which myo7aa affects hearing, transcriptome sequencing and bioinformatics analysis were employed. Results Our study showed that hair cells of myo7aa-/- zebrafish can not take up YO-PRO-1 fluorescent dye and are insensitive to acoustic stimulation in myo7aa-/- zebrafish compared to wild type. Genes related to the Rho GTPase signaling pathway, such as arhgap33, dab2ip, and arghef40, are significantly down-regulated in myo7aa-/- zebrafish embryos at 3 dpf. GTP and ATP compensation can partially rescue the hair cell defects in myo7aa knockout zebrafish. Discussion Our findings suggest that zebrafish myo7aa affects congenital hearing by regulating Rho GTPase signaling, and loss of myo7aa leads to abnormal Rho GTPase signaling and impairs hair cell function. myo7aa, myo7A, arhgap33, dab2ip, arghef40 and myo7aa-/- fonts in the abstract are italicized. -/- is a superscript format.
Collapse
Affiliation(s)
- Binling Xie
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jiaxin Liang
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jifan Jiang
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Ting Zeng
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Ling Liu
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Dinghua Xie
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ganghua Zhu
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lei Xiong
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, Hunan, China
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Kanjia Zhang
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dong Liu
- Nantong Laboratory of Development and Diseases, Key Laboratory of Neuroregeneration of Jiangsu and MOE, School of Life Sciences, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Jie Gong
- Nantong Laboratory of Development and Diseases, Key Laboratory of Neuroregeneration of Jiangsu and MOE, School of Life Sciences, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Xiangding Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ruosha Lai
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Huaping Xie
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
2
|
Seregin AA, Smirnova LP, Dmitrieva EM, Zavialova MG, Simutkin GG, Ivanova SA. Differential Expression of Proteins Associated with Bipolar Disorder as Identified Using the PeptideShaker Software. Int J Mol Sci 2023; 24:15250. [PMID: 37894929 PMCID: PMC10607299 DOI: 10.3390/ijms242015250] [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: 07/28/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
The prevalence of bipolar disorder (BD) in modern society is growing rapidly, but due to the lack of paraclinical criteria, its differential diagnosis with other mental disorders is somewhat challenging. In this regard, the relevance of proteomic studies is increasing due to the development of methods for processing large data arrays; this contributes to the discovery of protein patterns of pathological processes and the creation of new methods of diagnosis and treatment. It seems promising to search for proteins involved in the pathogenesis of BD in an easily accessible material-blood serum. Sera from BD patients and healthy individuals were purified via affinity chromatography to isolate 14 major proteins and separated using 1D SDS-PAGE. After trypsinolysis, the proteins in the samples were identified via HPLC/mass spectrometry. Mass spectrometric data were processed using the OMSSA and X!Tandem search algorithms using the UniProtKB database, and the results were analyzed using PeptideShaker. Differences in proteomes were assessed via an unlabeled NSAF-based analysis using a two-tailed Bonferroni-adjusted t-test. When comparing the blood serum proteomes of BD patients and healthy individuals, 10 proteins showed significant differences in NSAF values. Of these, four proteins were predominantly present in BD patients with the maximum NSAF value: 14-3-3 protein zeta/delta; ectonucleoside triphosphate diphosphohydrolase 7; transforming growth factor-beta-induced protein ig-h3; and B-cell CLL/lymphoma 9 protein. Further exploration of the role of these proteins in BD is warranted; conducting such studies will help develop new paraclinical criteria and discover new targets for BD drug therapy.
Collapse
Affiliation(s)
- Alexander A. Seregin
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634014, Russia; (A.A.S.)
| | - Liudmila P. Smirnova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634014, Russia; (A.A.S.)
| | - Elena M. Dmitrieva
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634014, Russia; (A.A.S.)
| | | | - German G. Simutkin
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634014, Russia; (A.A.S.)
| | - Svetlana A. Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634014, Russia; (A.A.S.)
| |
Collapse
|
3
|
Zolboot N, Xiao Y, Du JX, Ghanem MM, Choi SY, Junn MJ, Zampa F, Huang Z, MacRae IJ, Lippi G. MicroRNAs are necessary for the emergence of Purkinje cell identity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.560023. [PMID: 37808721 PMCID: PMC10557743 DOI: 10.1101/2023.09.28.560023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Brain computations are dictated by the unique morphology and connectivity of neuronal subtypes, features established by closely timed developmental events. MicroRNAs (miRNAs) are critical for brain development, but current technologies lack the spatiotemporal resolution to determine how miRNAs instruct the steps leading to subtype identity. Here, we developed new tools to tackle this major gap. Fast and reversible miRNA loss-of-function revealed that miRNAs are necessary for cerebellar Purkinje cell (PC) differentiation, which previously appeared miRNA-independent, and resolved distinct miRNA critical windows in PC dendritogenesis and climbing fiber synaptogenesis, key determinants of PC identity. To identify underlying mechanisms, we generated a mouse model, which enables precise mapping of miRNAs and their targets in rare cell types. With PC-specific maps, we found that the PC-enriched miR-206 drives exuberant dendritogenesis and modulates synaptogenesis. Our results showcase vastly improved approaches for dissecting miRNA function and reveal that many critical miRNA mechanisms remain largely unexplored. Highlights Fast miRNA loss-of-function with T6B impairs postnatal Purkinje cell developmentReversible T6B reveals critical miRNA windows for dendritogenesis and synaptogenesisConditional Spy3-Ago2 mouse line enables miRNA-target network mapping in rare cellsPurkinje cell-enriched miR-206 regulates its unique dendritic and synaptic morphology.
Collapse
|
4
|
Yun EJ, Kim S, Hsieh JT, Baek ST. Wnt/β-catenin signaling pathway induces autophagy-mediated temozolomide-resistance in human glioblastoma. Cell Death Dis 2020; 11:771. [PMID: 32943609 PMCID: PMC7498596 DOI: 10.1038/s41419-020-02988-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 01/04/2023]
Abstract
Temozolomide (TMZ) is widely used for treating glioblastoma multiforme (GBM), however, the treatment of such brain tumors remains a challenge due to the development of resistance. Increasing studies have found that TMZ treatment could induce autophagy that may link to therapeutic resistance in GBM, but, the precise mechanisms are not fully understood. Understanding the molecular mechanisms underlying the response of GBM to chemotherapy is paramount for developing improved cancer therapeutics. In this study, we demonstrated that the loss of DOC-2/DAB2 interacting protein (DAB2IP) is responsible for TMZ-resistance in GBM through ATG9B. DAB2IP sensitized GBM to TMZ and suppressed TMZ-induced autophagy by negatively regulating ATG9B expression. A higher level of ATG9B expression was associated with GBM compared to low-grade glioma. The knockdown of ATG9B expression in GBM cells suppressed TMZ-induced autophagy as well as TMZ-resistance. Furthermore, we showed that DAB2IP negatively regulated ATG9B expression by blocking the Wnt/β-catenin pathway. To enhance the benefit of TMZ and avoid therapeutic resistance, effective combination strategies were tested using a small molecule inhibitor blocking the Wnt/β-catenin pathway in addition to TMZ. The combination treatment synergistically enhanced the efficacy of TMZ in GBM cells. In conclusion, the present study identified the mechanisms of TMZ-resistance of GBM mediated by DAB2IP and ATG9B which provides insight into a potential strategy to overcome TMZ chemo-resistance.
Collapse
Affiliation(s)
- Eun-Jin Yun
- POSTECH Biotech Center, POSTECH, Pohang, Republic of Korea.
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics and Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Seung Tae Baek
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea.
- Department of Life Sciences, POSTECH, Pohang, Republic of Korea.
| |
Collapse
|
5
|
Harrell Stewart DR, Clark GJ. Pumping the brakes on RAS - negative regulators and death effectors of RAS. J Cell Sci 2020; 133:133/3/jcs238865. [PMID: 32041893 DOI: 10.1242/jcs.238865] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mutations that activate the RAS oncoproteins are common in cancer. However, aberrant upregulation of RAS activity often occurs in the absence of activating mutations in the RAS genes due to defects in RAS regulators. It is now clear that loss of function of Ras GTPase-activating proteins (RasGAPs) is common in tumors, and germline mutations in certain RasGAP genes are responsible for some clinical syndromes. Although regulation of RAS is central to their activity, RasGAPs exhibit great diversity in their binding partners and therefore affect signaling by multiple mechanisms that are independent of RAS. The RASSF family of tumor suppressors are essential to RAS-induced apoptosis and senescence, and constitute a barrier to RAS-mediated transformation. Suppression of RASSF protein expression can also promote the development of excessive RAS signaling by uncoupling RAS from growth inhibitory pathways. Here, we will examine how these effectors of RAS contribute to tumor suppression, through both RAS-dependent and RAS-independent mechanisms.
Collapse
Affiliation(s)
- Desmond R Harrell Stewart
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY 40222, USA
| | - Geoffrey J Clark
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY 40222, USA
| |
Collapse
|
6
|
Kuo SH, Louis ED, Faust PL, Handforth A, Chang SY, Avlar B, Lang EJ, Pan MK, Miterko LN, Brown AM, Sillitoe RV, Anderson CJ, Pulst SM, Gallagher MJ, Lyman KA, Chetkovich DM, Clark LN, Tio M, Tan EK, Elble RJ. Current Opinions and Consensus for Studying Tremor in Animal Models. CEREBELLUM (LONDON, ENGLAND) 2019; 18:1036-1063. [PMID: 31124049 PMCID: PMC6872927 DOI: 10.1007/s12311-019-01037-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tremor is the most common movement disorder; however, we are just beginning to understand the brain circuitry that generates tremor. Various neuroimaging, neuropathological, and physiological studies in human tremor disorders have been performed to further our knowledge of tremor. But, the causal relationship between these observations and tremor is usually difficult to establish and detailed mechanisms are not sufficiently studied. To overcome these obstacles, animal models can provide an important means to look into human tremor disorders. In this manuscript, we will discuss the use of different species of animals (mice, rats, fruit flies, pigs, and monkeys) to model human tremor disorders. Several ways to manipulate the brain circuitry and physiology in these animal models (pharmacology, genetics, and lesioning) will also be discussed. Finally, we will discuss how these animal models can help us to gain knowledge of the pathophysiology of human tremor disorders, which could serve as a platform towards developing novel therapies for tremor.
Collapse
Affiliation(s)
- Sheng-Han Kuo
- Department of Neurology, Columbia University, 650 West 168th Street, Room 305, New York, NY, 10032, USA.
| | - Elan D Louis
- Department of Neurology, Yale School of Medicine, Yale University, 800 Howard Avenue, Ste Lower Level, New Haven, CT, 06519, USA.
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, USA.
- Center for Neuroepidemiology and Clinical Neurological Research, Yale School of Medicine, Yale University, New Haven, CT, USA.
| | - Phyllis L Faust
- Department of Pathology and Cell Biology, Columbia University Medical Center and the New York Presbyterian Hospital, New York, NY, USA
| | - Adrian Handforth
- Neurology Service, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Su-Youne Chang
- Department of Neurologic Surgery and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Billur Avlar
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Eric J Lang
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Ming-Kai Pan
- Department of Medical Research and Neurology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Lauren N Miterko
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, TX, USA
| | - Amanda M Brown
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Roy V Sillitoe
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Collin J Anderson
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | | | - Kyle A Lyman
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Lorraine N Clark
- Department of Pathology and Cell Biology, Columbia University Medical Center and the New York Presbyterian Hospital, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Murni Tio
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
- Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Rodger J Elble
- Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
7
|
Insolia V, Priori EC, Gasperini C, Coppa F, Cocchia M, Iervasi E, Ferrari B, Besio R, Maruelli S, Bernocchi G, Forlino A, Bottone MG. Prolidase enzyme is required for extracellular matrix integrity and impacts on postnatal cerebellar cortex development. J Comp Neurol 2019; 528:61-80. [PMID: 31246278 DOI: 10.1002/cne.24735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/11/2019] [Accepted: 06/20/2019] [Indexed: 12/12/2022]
Abstract
The extracellular matrix is essential for brain development, lamination, and synaptogenesis. In particular, the basement membrane below the pial meninx (pBM) is required for correct cortical development. The last step in the catabolism of the most abundant protein in pBM, collagen Type IV, requires prolidase, an exopeptidase cleaving the imidodipeptides containing pro or hyp at the C-terminal end. Mutations impairing prolidase activity lead in humans to the rare disease prolidase deficiency characterized by severe skin ulcers and mental impairment. Thus, the dark-like (dal) mouse, in which the prolidase is knocked-out, was used to investigate whether the deficiency of prolidase affects the neuronal maturation during development of a brain cortex area. Focusing on the cerebellar cortex, thinner collagen fibers and disorganized pBM were found. Aberrant cortical granule cell proliferation and migration occurred, associated to defects in brain lamination, and in particular in maturation of Purkinje neurons and formation of synaptic contacts. This study deeply elucidates a link between prolidase activity and neuronal maturation shedding new light on the molecular basis of functional aspects in the prolidase deficiency.
Collapse
Affiliation(s)
- Violetta Insolia
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Erica C Priori
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Caterina Gasperini
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Federica Coppa
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Marco Cocchia
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Erika Iervasi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Beatrice Ferrari
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Silvia Maruelli
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | | | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy
| | - Maria G Bottone
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| |
Collapse
|
8
|
He J, Huang S, Lin Z, Zhang J, Su J, Ji W, Liu X. Disabled homolog 2 interactive protein functions as a tumor suppressor in osteosarcoma cells. Oncol Lett 2018; 16:703-712. [PMID: 29963135 PMCID: PMC6019915 DOI: 10.3892/ol.2018.8776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 06/16/2017] [Indexed: 11/29/2022] Open
Abstract
The disabled homolog 2 interactive protein (DAB2IP) gene is a member of the family of Ras GTPases and functions as a tumor suppressor in many types of carcinoma; however, its function in osteosarcoma remains unclear. The aim of the present study was to determine the function of DAB2IP in osteosarcoma and normal bone cells in vitro. The expression of DAB2IP protein was assessed in osteoblast and osteosarcoma cell lines by western blot analysis. The effects of DAB2IP expression on cell proliferation, colony formation, apoptosis, cell cycle, and cell migration and invasion were evaluated by in vitro studies. DAB2IP expression was lower in osteosarcoma cell lines than in normal osteoblast cell lines. DAB2IP expression affected cell proliferation, apoptosis and cell cycle distribution. In addition, DAB2IP inhibited the migration and invasion of osteosarcoma and normal osteoblast cells. Therefore, DAB2IP may function as a tumor suppressor in osteosarcoma cell lines by inhibiting cell proliferation and invasion.
Collapse
Affiliation(s)
- Jianan He
- Department of Orthopaedic Surgery, The Sixth Affiliated Hospital of Sun Yat-sun University, Guangzhou, Guangdong 510655, P.R. China.,Department of Interventional Radiology, The Fifth Affiliated Hospital of Sun Yat-sun University, Zhuhai, Guangdong 519000, P.R. China
| | - Shuai Huang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhenhua Lin
- Department of Orthopaedic Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Jiqin Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jialin Su
- Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Weidong Ji
- Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xingmo Liu
- Department of Orthopaedic Surgery, The Sixth Affiliated Hospital of Sun Yat-sun University, Guangzhou, Guangdong 510655, P.R. China
| |
Collapse
|
9
|
zur Nedden S, Eith R, Schwarzer C, Zanetti L, Seitter H, Fresser F, Koschak A, Cameron AJ, Parker PJ, Baier G, Baier-Bitterlich G. Protein kinase N1 critically regulates cerebellar development and long-term function. J Clin Invest 2018; 128:2076-2088. [PMID: 29494346 PMCID: PMC5919825 DOI: 10.1172/jci96165] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 02/27/2018] [Indexed: 01/16/2023] Open
Abstract
Increasing evidence suggests that synapse dysfunctions are a major determinant of several neurodevelopmental and neurodegenerative diseases. Here we identify protein kinase N1 (PKN1) as a novel key player in fine-tuning the balance between axonal outgrowth and presynaptic differentiation in the parallel fiber-forming (PF-forming) cerebellar granule cells (Cgcs). Postnatal Pkn1-/- animals showed a defective PF-Purkinje cell (PF-PC) synapse formation. In vitro, Pkn1-/- Cgcs exhibited deregulated axonal outgrowth, elevated AKT phosphorylation, and higher levels of neuronal differentiation-2 (NeuroD2), a transcription factor preventing presynaptic maturation. Concomitantly, Pkn1-/- Cgcs had a reduced density of presynaptic sites. By inhibiting AKT with MK-2206 and siRNA-mediated knockdown, we found that AKT hyperactivation is responsible for the elongated axons, higher NeuroD2 levels, and reduced density of presynaptic specifications in Pkn1-/- Cgcs. In line with our in vitro data, Pkn1-/- mice showed AKT hyperactivation, elevated NeuroD2 levels, and reduced expression of PF-PC synaptic markers during stages of PF maturation in vivo. The long-term effect of Pkn1 knockout was further seen in cerebellar atrophy and mild ataxia. In summary, our results demonstrate that PKN1 functions as a developmentally active gatekeeper of AKT activity, thereby fine-tuning axonal outgrowth and presynaptic differentiation of Cgcs and subsequently the correct PF-PC synapse formation.
Collapse
Affiliation(s)
| | | | - Christoph Schwarzer
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lucia Zanetti
- Institute of Pharmacy, Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Hartwig Seitter
- Institute of Pharmacy, Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Friedrich Fresser
- Department for Pharmacology and Genetics, Division of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexandra Koschak
- Institute of Pharmacy, Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Angus J.M. Cameron
- Kinase Biology Laboratory, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Peter J. Parker
- Francis Crick Institute, London, United Kingdom
- Division of Cancer Studies, King’s College London, London, United Kingdom
| | - Gottfried Baier
- Department for Pharmacology and Genetics, Division of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
10
|
Relatively frequent switching of transcription start sites during cerebellar development. BMC Genomics 2017; 18:461. [PMID: 28610618 PMCID: PMC5470264 DOI: 10.1186/s12864-017-3834-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/31/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alternative transcription start site (TSS) usage plays important roles in transcriptional control of mammalian gene expression. The growing interest in alternative TSSs and their role in genome diversification spawned many single-gene studies on differential usages of tissue-specific or temporal-specific alternative TSSs. However, exploration of the switching usage of alternative TSS usage on a genomic level, especially in the central nervous system, is largely lacking. RESULTS In this study, We have prepared a unique set of time-course data for the developing cerebellum, as part of the FANTOM5 consortium ( http://fantom.gsc.riken.jp/5/ ) that uses their innovative capturing of 5' ends of all transcripts followed by Helicos next generation sequencing. We analyzed the usage of all transcription start sites (TSSs) at each time point during cerebellar development that provided information on multiple RNA isoforms that emerged from the same gene. We developed a mathematical method that systematically compares the expression of different TSSs of a gene to identify temporal crossover and non-crossover switching events. We identified 48,489 novel TSS switching events in 5433 genes during cerebellar development. This includes 9767 crossover TSS switching events in 1511 genes, where the dominant TSS shifts over time. CONCLUSIONS We observed a relatively high prevalence of TSS switching in cerebellar development where the resulting temporally-specific gene transcripts and protein products can play important regulatory and functional roles.
Collapse
|
11
|
Shah B, Püschel AW. Regulation of Rap GTPases in mammalian neurons. Biol Chem 2017; 397:1055-69. [PMID: 27186679 DOI: 10.1515/hsz-2016-0165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/06/2016] [Indexed: 12/15/2022]
Abstract
Small GTPases are central regulators of many cellular processes. The highly conserved Rap GTPases perform essential functions in the mammalian nervous system during development and in mature neurons. During neocortical development, Rap1 is required to regulate cadherin- and integrin-mediated adhesion. In the adult nervous system Rap1 and Rap2 regulate the maturation and plasticity of dendritic spine and synapses. Although genetic studies have revealed important roles of Rap GTPases in neurons, their regulation by guanine nucleotide exchange factors (GEFs) that activate them and GTPase activating proteins (GAPs) that inactivate them by stimulating their intrinsic GTPase activity is just beginning to be explored in vivo. Here we review how GEFs and GAPs regulate Rap GTPases in the nervous system with a focus on their in vivo function.
Collapse
|
12
|
Wang JM, Zhou TJ, Zhang XX, Lu MJ, Liu Y. Construction of a luciferase reporter gene vector containing DAB2IP promoter. Shijie Huaren Xiaohua Zazhi 2016; 24:3985-3990. [DOI: 10.11569/wcjd.v24.i28.3985] [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: 02/07/2023] Open
Abstract
AIM To construct and identify pGL3-Basic-DAB2IP-luc vector, a luciferase reporter gene vector containing DAB2IP promoter, and explore the regulatory effect of Snail on the activity of this promoter in gastric cancer cells.
METHODS The fragment of DAB2IP gene promoter (1000 bp) was amplified by PCR and then cloned into pGL3-Basic-luc vector to obtain pGL3-Basic-DAB2IP-luc vector, which was identified by restriction enzyme digestion, sequencing and biological activity detection. The activity of pGL3-Basic-DAB2IP-luc and the expression of DAB2IP were detected after transfection with pEGFP-C1-Snail in gastric cancer cells.
RESULTS Luciferase reporter system showed that the luciferase activity was significantly lower in the Snail + pGL3-Basic-DAB2IP-luc group compared to the pGL3-Basic-DAB2IP-luc group and blank group (P < 0.01 for both). In addition, the luciferase activity was significantly lower in the pGL3-Basic-DAB2IP-luc group than in the blank group (P < 0.01). Real-time PCR and Western blot results showed that mRNA and protein expression of DAB2IP was decreased in gastric cancer cells after transfection with pEGFP-C1-Snail.
CONCLUSION The transcription factor Snail inhibits the transcription and expression of DAB2IP. The vector pGL3-Basic-DAB2IP-luc has been constructed successfully and its biological activity has been identified, which provides an important tool for further study of the role of DAB2IP in transcriptional regulation in gastric cancer.
Collapse
|
13
|
Cocito C, Merighi A, Giacobini M, Lossi L. Alterations of Cell Proliferation and Apoptosis in the Hypoplastic Reeler Cerebellum. Front Cell Neurosci 2016; 10:141. [PMID: 27252624 PMCID: PMC4879145 DOI: 10.3389/fncel.2016.00141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/10/2016] [Indexed: 12/24/2022] Open
Abstract
A mutation of the reln gene gives rise to the Reeler mouse (reln−∕−) displaying an ataxic phenotype and cerebellar hypoplasia. We have characterized the neurochemistry of postnatal (P0–P60) reln−∕− mouse cerebella with specific attention to the intervention of cell proliferation and apoptosis in the P0–P25 interval. Homozygous reln−∕− mice and age-matched controls were analyzed by immunofluorescence using primary antibodies against NeuN, calbindin, GFAP, vimentin, SMI32, and GAD67. Proliferation and apoptosis were detected after a single intraperitoneal BrdU injection and by the TUNEL assay with anti-digoxigenin rhodamine-conjugated antibodies. Quantitative analysis with descriptive and predictive statistics was used to calculate cell densities (number/mm2) after fluorescent nuclear stain (TCD, total cell density), labeling with BrdU (PrCD, proliferating cell density), or TUNEL (ApoCD, apoptotic cell density). By this approach we first have shown that the temporal pattern of expression of neuronal/glial markers in postnatal cerebellum is not affected by the Reeler mutation. Then, we have demonstrated that the hypoplasia in the Reeler mouse cerebellum is consequent to reduction of cortical size and cellularity (TCD), and that TCD is, in turn, linked to quantitative differences in the extent of cell proliferation and apoptosis, as well as derangements in their temporal trends during postnatal maturation. Finally, we have calculated that PrCD is the most important predictive factor to determine TCD in the cerebellar cortex of the mutants. These results support the notion that, beside the well-known consequences onto the migration of the cerebellar neurons, the lack of Reelin results in a measurable deficit in neural proliferation.
Collapse
Affiliation(s)
- Carolina Cocito
- Laboratory of Neuroscience, Department of Veterinary Sciences, University of Turin Grugliasco, Italy
| | - Adalberto Merighi
- Laboratory of Neuroscience, Department of Veterinary Sciences, University of Turin Grugliasco, Italy
| | - Mario Giacobini
- Laboratory of Dynamical Systems and Epidemiology, Department of Veterinary Sciences, University of Turin Grugliasco, Italy
| | - Laura Lossi
- Laboratory of Neuroscience, Department of Veterinary Sciences, University of Turin Grugliasco, Italy
| |
Collapse
|
14
|
Lee GH, D'Arcangelo G. New Insights into Reelin-Mediated Signaling Pathways. Front Cell Neurosci 2016; 10:122. [PMID: 27242434 PMCID: PMC4860420 DOI: 10.3389/fncel.2016.00122] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/27/2016] [Indexed: 11/21/2022] Open
Abstract
Reelin, a multifunctional extracellular protein that is important for mammalian brain development and function, is secreted by different cell types in the prenatal or postnatal brain. The spatiotemporal regulation of Reelin expression and distribution during development relates to its multifaceted function in the brain. Prenatally Reelin controls neuronal radial migration and proper positioning in cortical layers, whereas postnatally Reelin promotes neuronal maturation, synaptic formation and plasticity. The molecular mechanisms underlying the distinct biological functions of Reelin during and after brain development involve unique and overlapping signaling pathways that are activated following Reelin binding to its cell surface receptors. Distinct Reelin ligand isoforms, such as the full-length protein or fragments generated by proteolytic cleavage differentially affect the activity of downstream signaling pathways. In this review, we discuss recent advances in our understanding of the signaling transduction pathways activated by Reelin that regulate different aspects of brain development and function. A core signaling machinery, including ApoER2/VLDLR receptors, Src/Fyn kinases, and the adaptor protein Dab1, participates in all known aspects of Reelin biology. However, distinct downstream mechanisms, such as the Crk/Rap1 pathway and cell adhesion molecules, play crucial roles in the control of neuronal migration, whereas the PI3K/Akt/mTOR pathway appears to be more important for dendrite and spine development. Finally, the NMDA receptor (NMDAR) and an unidentified receptor contribute to the activation of the MEK/Erk1/2 pathway leading to the upregulation of genes involved in synaptic plasticity and learning. This knowledge may provide new insight into neurodevelopmental or neurodegenerative disorders that are associated with Reelin dysfunction.
Collapse
Affiliation(s)
- Gum Hwa Lee
- College of Pharmacy, Chosun University Gwangju, South Korea
| | - Gabriella D'Arcangelo
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey Piscataway, NJ, USA
| |
Collapse
|
15
|
Salami F, Qiao S, Homayouni R. Expression of mouse Dab2ip transcript variants and gene methylation during brain development. Gene 2015; 568:19-24. [PMID: 25958345 DOI: 10.1016/j.gene.2015.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/15/2015] [Accepted: 05/05/2015] [Indexed: 11/24/2022]
Abstract
Dab2ip (DOC-2/DAB2 interacting protein) is a RasGAP protein which shows a growth-inhibitory effect in human prostate cancer cell lines. Recent studies have shown that Dab2ip also plays an important role in regulating dendrite development and neuronal migration during brain development. In this study, we provide a more complete description of the mouse Dab2ip (mDab2ip) gene locus and examined DNA methylation and expression of Dab2ip during cerebellar development. Analysis of cDNA sequences in public databases revealed a total of 20 possible exons for mDab2ip gene, spanning over 172kb. Using Cap Analysis of Gene Expression (CAGE) data available through FANTOM5 project, we deduced five different transcription start sites for mDab2ip. Here, we characterized three different mDab2ip transcript variants beginning with exon 1. These transcripts varied by the presence or absence of exons 3 and 5, which encode a putative nuclear localization signal and the N-terminal region of a PH-domain, respectively. The 5' region of the mDab2ip gene contains three putative CpG islands (CpG131, CpG54, and CpG85). Interestingly, CpG54 and CpG85 are localized on exons 3 and 5. Bisulfate DNA sequencing showed that methylation level of CpG54 remained constant whereas methylation of CpG85 increased during cerebellar development. Real-time PCR analysis showed that the proportion of PH-domain containing mDab2ip transcripts increased during cerebellar development, in correlation with the increase in CpG85 methylation. These data suggest that site-specific methylation of mDab2ip gene during cerebellar development may play a role in inclusion of exon 5, resulting in a Dab2ip transcript variant that encodes a full pleckstrin homology (PH) domain.
Collapse
Affiliation(s)
- Farimah Salami
- Department of Biological Sciences, University of Memphis, Memphis, TN, United States
| | - Shuhong Qiao
- Department of Biological Sciences, University of Memphis, Memphis, TN, United States
| | - Ramin Homayouni
- Department of Biological Sciences, University of Memphis, Memphis, TN, United States.
| |
Collapse
|
16
|
Pappas A, Chaiworapongsa T, Romero R, Korzeniewski SJ, Cortez JC, Bhatti G, Gomez-Lopez N, Hassan SS, Shankaran S, Tarca AL. Transcriptomics of maternal and fetal membranes can discriminate between gestational-age matched preterm neonates with and without cognitive impairment diagnosed at 18-24 months. PLoS One 2015; 10:e0118573. [PMID: 25822971 PMCID: PMC4379164 DOI: 10.1371/journal.pone.0118573] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/20/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Neurocognitive impairment among children born preterm may arise from complex interactions between genes and the intra-uterine environment. OBJECTIVES (1) To characterize the transcriptomic profiles of chorioamniotic membranes in preterm neonates with and without neurocognitive impairment via microarrays and (2) to determine if neonates with neurocognitive impairment can be identified at birth. MATERIALS/METHODS A retrospective case-control study was conducted to examine the chorioamniotic transcriptome of gestational-age matched very preterm neonates with and without neurocognitive impairment at 18-24 months' corrected-age defined by a Bayley-III Cognitive Composite Score <80 (n = 14 each). Pathway analysis with down-weighting of overlapping genes (PADOG) was performed to identify KEGG pathways relevant to the phenotype. Select differentially expressed genes were profiled using qRT-PCR and a multi-gene disease prediction model was developed using linear discriminant analysis. The model's predictive performance was tested on a new set of cases and controls (n = 19 each). RESULTS 1) 117 genes were differentially expressed among neonates with and without subsequent neurocognitive impairment (p<0.05 and fold change >1.5); 2) Gene ontology analysis indicated enrichment of 19 biological processes and 3 molecular functions; 3)PADOG identified 4 significantly perturbed KEGG pathways: oxidative phosphorylation, Parkinson's disease, Alzheimer's disease and Huntington's disease (q-value <0.1); 4) 48 of 90 selected differentially expressed genes were confirmed by qRT-PCR, including genes implicated in energy metabolism, neuronal signaling, vascular permeability and response to injury (e.g., up-regulation of SEPP1, APOE, DAB2, CD163, CXCL12, VWF; down-regulation of HAND1, OSR1)(p<0.05); and 5) a multi-gene model predicted 18-24 month neurocognitive impairment (using the ratios of OSR1/VWF and HAND1/VWF at birth) in a larger, independent set (sensitivity = 74%, at specificity = 83%). CONCLUSIONS Gene expression patterns in the chorioamniotic membranes link neurocognitive impairment in preterm infants to neurodegenerative disease pathways and might be used to predict neurocognitive impairment. Further prospective studies are needed.
Collapse
Affiliation(s)
- Athina Pappas
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Pediatrics, Division of Neonatal and Perinatal Medicine, Wayne State University, Detroit, MI, United States of America
- * E-mail: (AP); (AT)
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States of America
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States of America
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States of America
| | - Steven J. Korzeniewski
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States of America
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States of America
| | - Josef C. Cortez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Pediatrics, Division of Neonatal and Perinatal Medicine, Wayne State University, Detroit, MI, United States of America
| | - Gaurav Bhatti
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States of America
- Department of Immunology and Microbiology, Wayne State University, Detroit, MI, United States of America
| | - Sonia S. Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States of America
| | - Seetha Shankaran
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Pediatrics, Division of Neonatal and Perinatal Medicine, Wayne State University, Detroit, MI, United States of America
| | - Adi L. Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development / NIH / DHHS, Bethesda, MD and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States of America
- * E-mail: (AP); (AT)
| |
Collapse
|
17
|
Qiao S, Homayouni R. Dab2IP Regulates Neuronal Positioning, Rap1 Activity and Integrin Signaling in the Developing Cortex. Dev Neurosci 2015; 37:131-41. [PMID: 25721469 DOI: 10.1159/000369092] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 10/15/2014] [Indexed: 01/10/2023] Open
Abstract
Dab2IP (DOC-2/DAB2 interacting protein) is a GTPase-activating protein which is involved in various aspects of brain development in addition to its roles in tumor formation and apoptosis in other systems. In this study, we carefully examined the expression profile of Dab2IP and investigated its physiological role during brain development using a Dab2IP-knockdown (KD) mouse model created by retroviral insertion of a LacZ-encoding gene-trapping cassette. LacZ staining revealed that Dab2IP is expressed in the ventricular zone as well as the cortical plate and the intermediate zone. Immunohistochemical analysis showed that Dab2IP protein is localized in the leading process and proximal cytoplasmic regions of migrating neurons in the intermediate zone. Bromodeoxyuridine birth dating experiments in combination with immunohistochemical analysis using layer-specific markers showed that Dab2IP is important for proper positioning of a subset of layer II-IV neurons in the developing cortex. Notably, neuronal migration was not completely disrupted in the cerebral cortex of Dab2IP-KD mice and disruption of migration was not strictly layer specific. Previously, we found that Dab2IP regulates multipolar transition in cortical neurons. Others have shown that Rap1 regulates the transition from multipolar to bipolar morphology in migrating postmitotic neurons through N-cadherin signaling and somal translocation in the superficial layer of the cortical plate through integrin signaling. Therefore, we examined whether Rap1 and integrin signaling were affected in Dab2IP-KD brains. We found that Dab2IP-KD resulted in higher levels of activated Rap1 and integrin in the developing cortex. Taken together, our results suggest that Dab2IP plays an important role in the migration and positioning of a subpopulation of later-born (layers II-IV) neurons, likely through the regulation of Rap1 and integrin signaling.
Collapse
Affiliation(s)
- Shuhong Qiao
- Department of Biological Sciences, University of Memphis, Memphis Tenn., USA
| | | |
Collapse
|
18
|
Lin CY, Louis ED, Faust PL, Koeppen AH, Vonsattel JPG, Kuo SH. Abnormal climbing fibre-Purkinje cell synaptic connections in the essential tremor cerebellum. ACTA ACUST UNITED AC 2014; 137:3149-59. [PMID: 25273997 DOI: 10.1093/brain/awu281] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Structural changes in Purkinje cells have been identified in the essential tremor cerebellum, although the mechanisms that underlie these changes remain poorly understood. Climbing fibres provide one of the major excitatory inputs to Purkinje cells, and climbing fibre-Purkinje cell connections are essential for normal cerebellar-mediated motor control. The distribution of climbing fibre-Purkinje cell synapses on Purkinje cell dendrites is dynamically regulated and may be altered in disease states. The aim of the present study was to examine the density and distribution of climbing fibre-Purkinje cell synapses using post-mortem cerebellar tissue of essential tremor cases and controls. Using vesicular glutamate transporter type 2 immunohistochemistry, we labelled climbing fibre-Purkinje cell synapses of 12 essential tremor cases and 13 age-matched controls from the New York Brain Bank. Normally, climbing fibres form synapses mainly on the thick, proximal Purkinje cell dendrites in the inner portion of the molecular layer, whereas parallel fibres form synapses on the thin, distal Purkinje cell spiny branchlets. We observed that, compared with controls, essential tremor cases had decreased climbing fibre-Purkinje cell synaptic density, more climbing fibres extending to the outer portion of the molecular layer, and more climbing fibre-Purkinje cell synapses on the thin Purkinje cell spiny branchlets. Interestingly, in essential tremor, the increased distribution of climbing fibre-Purkinje cell synapses on the thin Purkinje cell branchlets was inversely associated with clinical tremor severity, indicating a close relationship between the altered distribution of climbing fibre-Purkinje cell connections and tremor. These findings suggest that abnormal climbing fibre-Purkinje cell connections could be of importance in the pathogenesis of essential tremor.
Collapse
Affiliation(s)
- Chi-Ying Lin
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Elan D Louis
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA 2 GH Sergievsky Centre, Columbia University, New York, NY, USA 3 Taub Institute for Research of Alzheimer's disease and the Aging Brain, Columbia University, New York, NY, USA 4 Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Phyllis L Faust
- 5 Department of Pathology and Cell Biology, Columbia University Medical Centre and the New York Presbyterian Hospital, New York, NY, USA
| | - Arnulf H Koeppen
- 6 Neurology and Research Services, Veterans Affairs Medical Centre, Albany, NY, USA 7 Departments of Neurology and Pathology, Albany Medical College, Albany, NY, USA
| | - Jean-Paul G Vonsattel
- 3 Taub Institute for Research of Alzheimer's disease and the Aging Brain, Columbia University, New York, NY, USA 5 Department of Pathology and Cell Biology, Columbia University Medical Centre and the New York Presbyterian Hospital, New York, NY, USA
| | - Sheng-Han Kuo
- 1 Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
19
|
Castagna C, Aimar P, Alasia S, Lossi L. Post-natal development of the Reeler mouse cerebellum: An ultrastructural study. Ann Anat 2014; 196:224-35. [DOI: 10.1016/j.aanat.2013.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 11/15/2013] [Accepted: 11/25/2013] [Indexed: 01/19/2023]
|
20
|
Chang SLY, Chou RH, Zeng HJ, Lin YH, Chiu TY, Yang DM, Hung SC, Lai CH, Hsieh JT, Shyu WC, Yu YL. Downregulation of DAB2IP promotes mesenchymal-to-neuroepithelial transition and neuronal differentiation of human mesenchymal stem cells. PLoS One 2013; 8:e75884. [PMID: 24073285 PMCID: PMC3779184 DOI: 10.1371/journal.pone.0075884] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 08/16/2013] [Indexed: 12/05/2022] Open
Abstract
The DOC-2/DAB2 interactive protein (DAB2IP) is a new member of the Ras GTPase–activating protein family. Recent studies have shown that, in addition to its tumor suppressive role in various tumors, DAB2IP also plays an important role in regulating neuronal migration and positioning during brain development. In this study, we determined the roles of DAB2IP in the neuronal differentiation of human mesenchymal stem cells (hMSCs). We found that lentiviral short hairpin RNA (shRNA)-mediated knockdown of DAB2IP promoted the mesenchymal-to-neuroepithelial stem cell transition (MtNeST) and neuronal differentiation, which were accompanied by a reduction of cell proliferation but not apoptosis or cellular senescence. This suggests that DAB2IP plays an important role in the neuronal induction of hMSCs. Moreover, our finding that reduction of glycogen synthase kinase 3 beta (GSK3β) activity upon LiCl pretreatment inhibited both the MtNeST and production of MAP2-positive cells upon DAB2IP knockdown suggests that this transition is most likely mediated by regulation of the GSK3β signaling pathway. Our study demonstrates that DAB2IP participates in the first step of neuron induction of hMSCs, which implies a potentially important role for DAB2IP in the MtNeST during neurogenesis.
Collapse
Affiliation(s)
- Sunny Li-Yun Chang
- Graduate Institute of Basic Medical Science, and Graduate Institute of Molecular Systems Biomedicine, China Medical University, Taichung, Taiwan
| | - Ruey-Hwang Chou
- Graduate Institute of Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Hong-Jie Zeng
- Graduate Institute of Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Yu-Hsuan Lin
- Graduate Institute of Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Tai-Yu Chiu
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Biophotonics, School of Medical Technology and Engineering and Biophotonics and Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan
| | - De-Ming Yang
- Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Biophotonics, School of Medical Technology and Engineering and Biophotonics and Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Chieh Hung
- Stem Cell Laboratory, Department of Medical Research and Education, Orthopaedics and Traumatology, Taipei Veterans General Hospital and Institute of Clinical Medicine, Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology, School of Medicine, Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Jer-Tsong Hsieh
- University of Texas, Department of Urology, Southwestern Medical Center, Dallas, Texas, United States of America
| | - Woei-Cherng Shyu
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- Translational Medicine Research Center and Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- * E-mail: (YLY); (WCS)
| | - Yung-Luen Yu
- Graduate Institute of Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- * E-mail: (YLY); (WCS)
| |
Collapse
|