1
|
Attanasio C, Palladino A, Giaquinto D, Scavizzi F, Raspa M, Peres C, Anastasio C, Scocco P, Lucini C, de Girolamo P, D'Angelo L, De Felice E. Morphological phenotyping of the aging cochlea in inbred C57BL/6N and outbred CD1 mouse strains. Aging Cell 2025; 24:e14362. [PMID: 39482905 PMCID: PMC11709085 DOI: 10.1111/acel.14362] [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: 05/30/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 11/03/2024] Open
Abstract
Morphological mouse phenotyping plays a pivotal role in the translational setting and even more in the area of auditory research, where mouse is a central model organism due to the evolutionary genetic relationship and morpho-functional analogies with the human auditory system. However, some results obtained in murine models cannot be translated to humans due to the inadequate description of experimental conditions underlying poor reproducibility. We approach the characterization of the aging process of the mouse cochlea in animals up to 18 months of age belonging to two of the most used outbred (CD1) and inbred (C57BL/6N) strains. Striving to reduce any environmental variable we performed our study compliantly to the ARRIVE guidelines. We integrated instrumental data (auditory brainstem response test), with morphological analyses to correlate functional discrepancies to morphological changes and track the differences in the evolution of sensorineural hearing loss in the two strains. We featured the localization of Gipc3, Myosin VIIa, and TMC1 in hair cells of the Corti organ as well as NF 200 and the density of type I neuron in the spiral ganglion. We outlined age-related hearing loss (ARHL) in both strains, and a clear drop in the selected marker localization. However, in CD1 we detected a different trend allowing the identification of potential strain-specific mechanisms, namely an increase in myosin VIIa in 6 months aging mice in comparison to 2 months old animals. Our findings represent an asset to investigate the strain-dependent physiological trigger of ARHL providing new insights in the translational area.
Collapse
Affiliation(s)
- Chiara Attanasio
- Department of Veterinary Medicine and Animal ProductionUniversity of Naples Federico IINaplesItaly
| | - Antonio Palladino
- Department of Agricultural SciencesUniversity of Naples Federico IINaplesItaly
| | - Daniela Giaquinto
- Department of Veterinary Medicine and Animal ProductionUniversity of Naples Federico IINaplesItaly
| | - Ferdinando Scavizzi
- National Research Council, CNR—Institute of Cellular Biology and NeurobiologyMonterotondoItaly
- National Research Council, CNR—Institute of Biochemistry and Cell Biology ‐ International Campus EMMA‐INFRAFRONTIER‐IMPCMonterotondoItaly
| | - Marcello Raspa
- National Research Council, CNR—Institute of Cellular Biology and NeurobiologyMonterotondoItaly
- National Research Council, CNR—Institute of Biochemistry and Cell Biology ‐ International Campus EMMA‐INFRAFRONTIER‐IMPCMonterotondoItaly
| | - Chiara Peres
- National Research Council, CNR—Institute of Cellular Biology and NeurobiologyMonterotondoItaly
| | - Camilla Anastasio
- Department of Precision MedicineUniversity of Campania Luigi VanvitelliNaplesItaly
| | - Paola Scocco
- School of Biosciences and Veterinary MedicineUniversity of CamerinoCamerinoItaly
| | - Carla Lucini
- Department of Veterinary Medicine and Animal ProductionUniversity of Naples Federico IINaplesItaly
| | - Paolo de Girolamo
- Department of Veterinary Medicine and Animal ProductionUniversity of Naples Federico IINaplesItaly
| | - Livia D'Angelo
- Department of Veterinary Medicine and Animal ProductionUniversity of Naples Federico IINaplesItaly
| | - Elena De Felice
- School of Biosciences and Veterinary MedicineUniversity of CamerinoCamerinoItaly
| |
Collapse
|
2
|
Zhao G, Wang Y, Fan Z, Xiong J, Ertas YN, Ashammakhi N, Wang J, Ma T. Nanomaterials in crossroad of autophagy control in human cancers: Amplification of cell death mechanisms. Cancer Lett 2024; 591:216860. [PMID: 38583650 DOI: 10.1016/j.canlet.2024.216860] [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: 01/30/2024] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Cancer is the result of genetic abnormalities that cause normal cells to grow into neoplastic cells. Cancer is characterized by several distinct features, such as uncontrolled cell growth, extensive spreading to other parts of the body, and the ability to resist treatment. The scientists have stressed the development of nanostructures as novel therapeutic options in suppressing cancer, in response to the emergence of resistance to standard medicines. One of the specific mechanisms with dysregulation during cancer is autophagy. Nanomaterials have the ability to specifically carry medications and genes, and they can also enhance the responsiveness of tumor cells to standard therapy while promoting drug sensitivity. The primary mechanism in this process relies on autophagosomes and their fusion with lysosomes to break down the components of the cytoplasm. While autophagy was initially described as a form of cellular demise, it has been demonstrated to play a crucial role in controlling metastasis, proliferation, and treatment resistance in human malignancies. The pharmacokinetic profile of autophagy modulators is poor, despite their development for use in cancer therapy. Consequently, nanoparticles have been developed for the purpose of delivering medications and autophagy modulators selectively and specifically to the cancer process. Furthermore, several categories of nanoparticles have demonstrated the ability to regulate autophagy, which plays a crucial role in defining the biological characteristics and response to therapy of tumor cells.
Collapse
Affiliation(s)
- Gang Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yutao Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, 100000, China
| | - Zhongru Fan
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Jian Xiong
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Türkiye; Department of Biomedical Engineering, Erciyes University, Kayseri, 39039, Türkiye.
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, College of Engineering and Human Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| | - Jianfeng Wang
- Department of Urology, First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Ting Ma
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
3
|
Siegel F, Schmidt H, Juneja M, Smith J, Herrmann P, Kobelt D, Sharma K, Fichtner I, Walther W, Dittmar G, Volkmer R, Rathjen FG, Schlag PM, Stein U. GIPC1 regulates MACC1-driven metastasis. Front Oncol 2023; 13:1280977. [PMID: 38144523 PMCID: PMC10748395 DOI: 10.3389/fonc.2023.1280977] [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: 08/21/2023] [Accepted: 11/14/2023] [Indexed: 12/26/2023] Open
Abstract
Background Identification of cancer metastasis-relevant molecular networks is desired to provide the basis for understanding and developing intervention strategies. Here we address the role of GIPC1 in the process of MACC1-driven metastasis. MACC1 is a prognostic indicator for patient metastasis formation and metastasis-free survival. MACC1 controls gene transcription, promotes motility, invasion and proliferation of colon cancer cells in vitro, and causes tumor growth and metastasis in mice. Methods By using yeast-two-hybrid assay, mass spectrometry, co-immunoprecipitation and peptide array we analyzed GIPC1 protein binding partners, by using the MACC1 gene promoter and chromatin immunoprecipitation and electrophoretic mobility shift assay we probed for GIPC1 as transcription factor. We employed GIPC1/MACC1-manipulated cell lines for in vitro and in vivo analyses, and we probed the GIPC1/MACC1 impact using human primary colorectal cancer (CRC) tissue. Results We identified MACC1 and its paralogue SH3BP4 as protein binding partners of the protein GIPC1, and we also demonstrated the binding of GIPC1 as transcription factor to the MACC1 promoter (TSS to -60 bp). GIPC1 knockdown reduced endogenous, but not CMV promoter-driven MACC1 expression, and diminished MACC1-induced cell migration and invasion. GIPC1 suppression reduced tumor growth and metastasis in mice intrasplenically transplanted with MACC1-overexpressing CRC cells. In human primary CRC specimens, GIPC1 correlates with MACC1 expression and is of prognostic value for metastasis formation and metastasis-free survival. Combination of MACC1 and GIPC1 expression improved patient survival prognosis, whereas SH3BP4 expression did not show any prognostic value. Conclusions We identified an important, dual function of GIPC1 - as protein interaction partner and as transcription factor of MACC1 - for tumor progression and cancer metastasis.
Collapse
Affiliation(s)
- Franziska Siegel
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Hannes Schmidt
- Department Developmental Neurobiology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Manisha Juneja
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Janice Smith
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Pia Herrmann
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Dennis Kobelt
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - Kamal Sharma
- Department Developmental Neurobiology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Iduna Fichtner
- Experimental Pharmacology and Oncology, GmbH, Berlin, Germany
| | - Wolfgang Walther
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Gunnar Dittmar
- Department Mass Spectrometry, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Rudolf Volkmer
- Institute for Medicinal Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Fritz G. Rathjen
- Department Developmental Neurobiology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | - Ulrike Stein
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
4
|
Gangadaran P, Madhyastha H, Madhyastha R, Rajendran RL, Nakajima Y, Watanabe N, Velikkakath AKG, Hong CM, Gopi RV, Muthukalianan GK, Valsala Gopalakrishnan A, Jeyaraman M, Ahn BC. The emerging role of exosomes in innate immunity, diagnosis and therapy. Front Immunol 2023; 13:1085057. [PMID: 36726968 PMCID: PMC9885214 DOI: 10.3389/fimmu.2022.1085057] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/16/2022] [Indexed: 01/17/2023] Open
Abstract
Exosomes, which are nano-sized transport bio-vehicles, play a pivotal role in maintaining homeostasis by exchanging genetic or metabolic information between different cells. Exosomes can also play a vital role in transferring virulent factors between the host and parasite, thereby regulating host gene expression and the immune interphase. The association of inflammation with disease development and the potential of exosomes to enhance or mitigate inflammatory pathways support the notion that exosomes have the potential to alter the course of a disease. Clinical trials exploring the role of exosomes in cancer, osteoporosis, and renal, neurological, and pulmonary disorders are currently underway. Notably, the information available on the signatory efficacy of exosomes in immune-related disorders remains elusive and sporadic. In this review, we discuss immune cell-derived exosomes and their application in immunotherapy, including those against autoimmune connective tissue diseases. Further, we have elucidated our views on the major issues in immune-related pathophysiological processes. Therefore, the information presented in this review highlights the role of exosomes as promising strategies and clinical tools for immune regulation.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Radha Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Yuichi Nakajima
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Nozomi Watanabe
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Anoop Kumar G. Velikkakath
- Center for System Biology and Molecular Medicine, Yenepoya Research center, Yenepoya (Deemed to be University), Mangaluru, Karnataka, India
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Rahul Velikkakath Gopi
- Department of Tissue Engineering and Regeneration Technologies, Sree Chitra Thirunal Institute of Medical Sciences and Technology, Thiruvananthapuram, India
| | | | | | - Madhan Jeyaraman
- Department of Orthopaedics, Faculty of Medicine, Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu, India
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
5
|
Li X, Shi L, Wang L. A review of the mechanisms underlying the role of the GIPC3 gene in hereditary deafness. Front Synaptic Neurosci 2023; 14:1101587. [PMID: 36704659 PMCID: PMC9872657 DOI: 10.3389/fnsyn.2022.1101587] [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: 11/18/2022] [Accepted: 12/16/2022] [Indexed: 01/09/2023] Open
Abstract
The GAIP interacting protein c terminus (GIPC) genes encode a small family of proteins characterized by centrally located PDZ domains. GIPC3 encodes a 312 amino acid protein. Variants of human GIPC3 are associated with non-syndromic hearing loss. GIPC3 is one of over a hundred different genes with variants causing human deafness. Screening for variants of GIPC3 is essential for early detection of hearing loss in children and eventually treatment of deafness. Accordingly, this paper assesses the status of research developments on the role of GIPC3 in hereditary deafness and the effects of pathogenic variants on the auditory system.
Collapse
Affiliation(s)
- Xinxin Li
- Department of Otolaryngology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lin Shi
- Department of Otolaryngology, First Affiliated Hospital of Dalian Medical University, Dalian, China,*Correspondence: Lin Shi,
| | - Liang Wang
- National Joint Engineering Laboratory, Stem Cell Clinical Research Center, Regenerative Medicine Center, First Affiliated Hospital of Dalian Medical University, Dalian, China,Liang Wang,
| |
Collapse
|
6
|
Genera M, Colcombet-Cazenave B, Croitoru A, Raynal B, Mechaly A, Caillet J, Haouz A, Wolff N, Caillet-Saguy C. Interactions of the protein tyrosine phosphatase PTPN3 with viral and cellular partners through its PDZ domain: insights into structural determinants and phosphatase activity. Front Mol Biosci 2023; 10:1192621. [PMID: 37200868 PMCID: PMC10185773 DOI: 10.3389/fmolb.2023.1192621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/18/2023] [Indexed: 05/20/2023] Open
Abstract
The human protein tyrosine phosphatase non-receptor type 3 (PTPN3) is a phosphatase containing a PDZ (PSD-95/Dlg/ZO-1) domain that has been found to play both tumor-suppressive and tumor-promoting roles in various cancers, despite limited knowledge of its cellular partners and signaling functions. Notably, the high-risk genital human papillomavirus (HPV) types 16 and 18 and the hepatitis B virus (HBV) target the PDZ domain of PTPN3 through PDZ-binding motifs (PBMs) in their E6 and HBc proteins respectively. This study focuses on the interactions between the PTPN3 PDZ domain (PTPN3-PDZ) and PBMs of viral and cellular protein partners. We solved the X-ray structures of complexes between PTPN3-PDZ and PBMs of E6 of HPV18 and the tumor necrosis factor-alpha converting enzyme (TACE). We provide new insights into key structural determinants of PBM recognition by PTPN3 by screening the selectivity of PTPN3-PDZ recognition of PBMs, and by comparing the PDZome binding profiles of PTPN3-recognized PBMs and the interactome of PTPN3-PDZ. The PDZ domain of PTPN3 was known to auto-inhibit the protein's phosphatase activity. We discovered that the linker connecting the PDZ and phosphatase domains is involved in this inhibition, and that the binding of PBMs does not impact this catalytic regulation. Overall, the study sheds light on the interactions and structural determinants of PTPN3 with its cellular and viral partners, as well as on the inhibitory role of its PDZ domain on its phosphatase activity.
Collapse
Affiliation(s)
- Mariano Genera
- Institut Pasteur, Université Paris Cité, Channel Receptors Unit, Paris, France
- Sorbonne Université, Complexité du Vivant, F-75005, Paris, France
| | - Baptiste Colcombet-Cazenave
- Institut Pasteur, Université Paris Cité, Channel Receptors Unit, Paris, France
- Sorbonne Université, Complexité du Vivant, F-75005, Paris, France
| | - Anastasia Croitoru
- Institut Pasteur, Université Paris Cité, Channel Receptors Unit, Paris, France
| | - Bertrand Raynal
- Molecular Biophysics Platform-C2RT, CNRS, Institut Pasteur, Université Paris Cité, Paris, France
| | - Ariel Mechaly
- Crystallography Platform-C2RT, Institut Pasteur, Université Paris Cité, Paris, France
| | - Joël Caillet
- CNRS, Institut de Biologie Physico-Chimique, Université Paris Cité, Paris, France
| | - Ahmed Haouz
- Crystallography Platform-C2RT, Institut Pasteur, Université Paris Cité, Paris, France
| | - Nicolas Wolff
- Institut Pasteur, Université Paris Cité, Channel Receptors Unit, Paris, France
| | - Célia Caillet-Saguy
- Institut Pasteur, Université Paris Cité, Channel Receptors Unit, Paris, France
- *Correspondence: Célia Caillet-Saguy,
| |
Collapse
|
7
|
Jamshidi V, Nobakht M Gh BF, Parvin S, Bagheri H, Ghanei M, Shahriary A, Davoudi SM, Arabfard M. Proteomics analysis of chronic skin injuries caused by mustard gas. BMC Med Genomics 2022; 15:175. [PMID: 35933451 PMCID: PMC9357330 DOI: 10.1186/s12920-022-01328-3] [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: 02/05/2022] [Accepted: 08/03/2022] [Indexed: 12/02/2022] Open
Abstract
Sulfur mustard (SM) is an alkylating and forming chemical that was widely used by Iraqi forces during the Iran–Iraq wars. One of the target organs of SM is the skin. Understanding the mechanisms involved in the pathogenesis of SM may help better identify complications and find appropriate treatments. The current study collected ten SM-exposed patients with long-term skin complications and ten healthy individuals. Proteomics experiments were performed using the high-efficiency TMT10X method to evaluate the skin protein profile, and statistical bioinformatics methods were used to identify the differentially expressed proteins. One hundred twenty-nine proteins had different expressions between the two groups. Of these 129 proteins, 94 proteins had increased expression in veterans' skins, while the remaining 35 had decreased expression. The hub genes included RPS15, ACTN1, FLNA, HP, SDHC, and RPL29, and three modules were extracted from the PPI network analysis. Skin SM exposure can lead to oxidative stress, inflammation, apoptosis, and cell proliferation.
Collapse
Affiliation(s)
- Vahid Jamshidi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - B Fatemeh Nobakht M Gh
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shahram Parvin
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Education Office, Pasteur Institute of Iran, Tehran, Iran
| | - Hasan Bagheri
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Shahriary
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyyed Masoud Davoudi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masoud Arabfard
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Hou S, Zhang J, Wu Y, Junmin C, Yuyu H, He B, Yang Y, Hong Y, Chen J, Yang J, Li S. FGF22 deletion causes hidden hearing loss by affecting the function of inner hair cell ribbon synapses. Front Mol Neurosci 2022; 15:922665. [PMID: 35966010 PMCID: PMC9366910 DOI: 10.3389/fnmol.2022.922665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
Ribbon synapses are important structures in transmitting auditory signals from the inner hair cells (IHCs) to their corresponding spiral ganglion neurons (SGNs). Over the last few decades, deafness has been primarily attributed to the deterioration of cochlear hair cells rather than ribbon synapses. Hearing dysfunction that cannot be detected by the hearing threshold is defined as hidden hearing loss (HHL). The relationship between ribbon synapses and FGF22 deletion remains unknown. In this study, we used a 6-week-old FGF22 knockout mice model (Fgf22–/–) and mainly focused on alteration in ribbon synapses by applying the auditory brainstem response (ABR) test, the immunofluorescence staining, the patch-clamp recording, and quantitative real-time PCR. In Fgf22–/– mice, we found the decreased amplitude of ABR wave I, the reduced vesicles of ribbon synapses, and the decreased efficiency of exocytosis, which was suggested by a decrease in the capacitance change. Quantitative real-time PCR revealed that Fgf22–/– led to dysfunction in ribbon synapses by downregulating SNAP-25 and Gipc3 and upregulating MEF2D expression, which was important for the maintenance of ribbon synapses’ function. Our research concluded that FGF22 deletion caused HHL by affecting the function of IHC ribbon synapses and may offer a novel therapeutic target to meet an ever-growing demand for deafness treatment.
Collapse
Affiliation(s)
- Shule Hou
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jifang Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yan Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Chen Junmin
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Huang Yuyu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Baihui He
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yan Yang
- Liaoning Medical Device Test Institute, Shenyang, China
| | - Yuren Hong
- Laboratory of Electron Microscope Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiarui Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jiarui Chen,
| | - Jun Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Jun Yang,
| | - Shuna Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Shuna Li,
| |
Collapse
|
9
|
Mayya C, Naveena AH, Sinha P, Wunder C, Johannes L, Bhatia D. The roles of dynein and myosin VI motor proteins in endocytosis. J Cell Sci 2022; 135:274777. [DOI: 10.1242/jcs.259387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
ABSTRACT
Endocytosis is indispensable for multiple cellular processes, including signalling, cell adhesion, migration, as well as the turnover of plasma membrane lipids and proteins. The dynamic interplay and regulation of different endocytic entry routes requires multiple cytoskeletal elements, especially motor proteins that bind to membranes and transport vesicles along the actin and microtubule cytoskeletons. Dynein and kinesin motor proteins transport vesicles along microtubules, whereas myosins drive vesicles along actin filaments. Here, we present a brief overview of multiple endocytic pathways and our current understanding of the involvement of these motor proteins in the regulation of the different cellular entry routes. We particularly focus on structural and mechanistic details of the retrograde motor proteins dynein and myosin VI (also known as MYO6), along with their adaptors, which have important roles in the early events of endocytosis. We conclude by highlighting the key challenges in elucidating the involvement of motor proteins in endocytosis and intracellular membrane trafficking.
Collapse
Affiliation(s)
- Chaithra Mayya
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| | - A. Hema Naveena
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| | - Pankhuri Sinha
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| | - Christian Wunder
- Institut Curie, PSL Research University, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, 26 rue d'Ulm, 75248 Paris CEDEX 05, France
| | - Ludger Johannes
- Institut Curie, PSL Research University, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, 26 rue d'Ulm, 75248 Paris CEDEX 05, France
| | - Dhiraj Bhatia
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
- Center for Biomedical Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| |
Collapse
|
10
|
Johnson GP, Jonas KC. Mechanistic insight into how gonadotropin hormone receptor complexes direct signaling†. Biol Reprod 2021; 102:773-783. [PMID: 31882999 DOI: 10.1093/biolre/ioz228] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 12/29/2022] Open
Abstract
Gonadotropin hormones and their receptors play a central role in the control of male and female reproduction. In recent years, there has been growing evidence surrounding the complexity of gonadotropin hormone/receptor signaling, with it increasingly apparent that the Gαs/cAMP/PKA pathway is not the sole signaling pathway that confers their biological actions. Here we review recent literature on the different receptor-receptor, receptor-scaffold, and receptor-signaling molecule complexes formed and how these modulate and direct gonadotropin hormone-dependent intracellular signal activation. We will touch upon the more controversial issue of extragonadal expression of FSHR and the differential signal pathways activated in these tissues, and lastly, highlight the open questions surrounding the role these gonadotropin hormone receptor complexes and how this will shape future research directions.
Collapse
Affiliation(s)
| | - Kim Carol Jonas
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| |
Collapse
|
11
|
Vivekanandhan S, Madamsetty VS, Angom RS, Dutta SK, Wang E, Caulfield T, Pletnev AA, Upstill-Goddard R, Asmann YW, Chang D, Spaller MR, Mukhopadhyay D. Role of PLEXIND1/TGFβ Signaling Axis in Pancreatic Ductal Adenocarcinoma Progression Correlates with the Mutational Status of KRAS. Cancers (Basel) 2021; 13:cancers13164048. [PMID: 34439202 PMCID: PMC8393884 DOI: 10.3390/cancers13164048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Pancreatic cancer is among the most lethal cancers. The expression of PLEXIND1, a receptor, is upregulated in many cancers (including pancreatic cancer). Traditionally, PLEXIND1 is known to be involved in neuron development and mediate semaphorin signaling. However, its role and signaling in cancer is not fully understood. In our study, we present a new mechanism through which PLEXIND1 mediates its roles in cancer. For the first time, we demonstrate that it can function as a transforming growth factor beta coreceptor and modulate SMAD3 signaling. Around 90% of pancreatic cancer patients have mutant KRAS. Our work suggests that PLEXIND1 functions differently in pancreatic cancer cell lines, and the difference correlates with KRAS mutational status. Additionally, we demonstrate a novel peptide based therapeutic approach to target PLEXIND1 in cancer cells. Our work is valuable to both neuroscience and cancer fields, as it demonstrates an association between two previously unrelated signaling pathways. Abstract PLEXIND1 is upregulated in several cancers, including pancreatic ductal adenocarcinoma (PDAC). It is an established mediator of semaphorin signaling, and neuropilins are its known coreceptors. Herein, we report data to support the proposal that PLEXIND1 acts as a transforming growth factor beta (TGFβ) coreceptor, modulating cell growth through SMAD3 signaling. Our findings demonstrate that PLEXIND1 plays a pro-tumorigenic role in PDAC cells with oncogenic KRAS (KRASmut). We show in KRASmut PDAC cell lines (PANC-1, AsPC-1,4535) PLEXIND1 downregulation results in decreased cell viability (in vitro) and reduced tumor growth (in vivo). Conversely, PLEXIND1 acts as a tumor suppressor in the PDAC cell line (BxPC-3) with wild-type KRAS (KRASwt), as its reduced expression results in higher cell viability (in-vitro) and tumor growth (in vivo). Additionally, we demonstrate that PLEXIND1-mediated interactions can be selectively disrupted using a peptide based on its C-terminal sequence (a PDZ domain-binding motif), an outcome that may possess significant therapeutic implications. To our knowledge, this is the first report showing that (1) PLEXIND1 acts as a TGFβ coreceptor and mediates SMAD3 signaling, and (2) differential roles of PLEXIND1 in PDAC cell lines correlate with KRASmut and KRASwt status.
Collapse
Affiliation(s)
- Sneha Vivekanandhan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Vijay S. Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Shamit Kumar Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Thomas Caulfield
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
| | - Alexandre A. Pletnev
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA; (A.A.P.); (M.R.S.)
| | - Rosanna Upstill-Goddard
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate Switchback Road, Glasgow G12 8QQ, UK; (R.U.-G.); (D.C.)
| | - Yan W. Asmann
- Health Sciences Research, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA;
| | - David Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate Switchback Road, Glasgow G12 8QQ, UK; (R.U.-G.); (D.C.)
| | - Mark R. Spaller
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA; (A.A.P.); (M.R.S.)
- Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, Lebanon, NH 03756, USA
- Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan 215316, China
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA; (S.V.); (V.S.M.); (R.S.A.); (S.K.D.); (E.W.); (T.C.)
- Correspondence:
| |
Collapse
|
12
|
Ahmed T, Mythreye K, Lee NY. Strength and duration of GIPC-dependent signaling networks as determinants in cancer. Neoplasia 2021; 23:181-188. [PMID: 33360508 PMCID: PMC7773760 DOI: 10.1016/j.neo.2020.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 10/25/2022]
Abstract
GIPC is a PDZ-domain containing adaptor protein that regulates the cell surface expression and endocytic trafficking of numerous transmembrane receptors and signaling complexes. Interactions with over 50 proteins have been reported to date including VEGFR, insulin-like growth factor-1 receptor (IGF-1R), GPCRs, and APPL, many of which have essential roles in neuronal and cardiovascular development. In cancer, a major subset of GIPC-binding receptors and cytoplasmic effectors have been shown to promote tumorigenesis or metastatic progression, while other subsets have demonstrated strong tumor-suppressive effects. Given that these diverse pathways are widespread in normal tissues and human malignancies, precisely how these opposing signals are integrated and regulated within the same tumor setting likely depend on the strength and duration of their interactions with GIPC. This review highlights the major pathways and divergent mechanisms of GIPC signaling in various cancers and provide a rationale for emerging GIPC-targeted cancer therapies.
Collapse
Affiliation(s)
- Tasmia Ahmed
- Deparment of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Karthikeyan Mythreye
- Division of Molecular and Cellular Pathology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Nam Y Lee
- Deparment of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA; Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA.
| |
Collapse
|
13
|
Colletti M, Ceglie D, Di Giannatale A, Nazio F. Autophagy and Exosomes Relationship in Cancer: Friends or Foes? Front Cell Dev Biol 2021; 8:614178. [PMID: 33511121 PMCID: PMC7835528 DOI: 10.3389/fcell.2020.614178] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
Autophagy is an intracellular degradation process involved in the removal of proteins and damaged organelles by the formation of a double-membrane vesicle named autophagosome and degraded through fusion with lysosomes. An intricate relationship between autophagy and the endosomal and exosomal pathways can occur at different stages with important implications for normal physiology and human diseases. Recent researches have revealed that extracellular vesicles (EVs), such as exosomes, could have a cytoprotective role by inducing intracellular autophagy; on the other hand, autophagy plays a crucial role in the biogenesis and degradation of exosomes. Although the importance of these processes in cancer is well established, their interplay in tumor is only beginning to be documented. In some tumor contexts (1) autophagy and exosome-mediated release are coordinately activated, sharing the molecular machinery and regulatory mechanisms; (2) cancer cell-released exosomes impact on autophagy in recipient cells through mechanisms yet to be determined; (3) exosome-autophagy relationship could affect drug resistance and tumor microenvironment (TME). In this review, we survey emerging discoveries relevant to the exosomes and autophagy crosstalk in the context of cancer initiation, progression and recurrence. Consequently, we discuss clinical implications by targeting autophagy-exosomal pathway interaction and how this could lay a basis for the purpose of novel cancer therapeutics.
Collapse
Affiliation(s)
- Marta Colletti
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - Donatella Ceglie
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - Angela Di Giannatale
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesca Nazio
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
14
|
Kim J, Min H, Ko S, Shim YH. Depletion of gipc-1 and gipc-2 causes infertility in Caenorhabditis elegans by reducing sperm motility. Biochem Biophys Res Commun 2020; 534:219-225. [PMID: 33280819 DOI: 10.1016/j.bbrc.2020.11.108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 11/24/2022]
Abstract
The G-protein signaling pathway plays a key role in multiple cellular processes and is well conserved in eukaryotes. Although GIPC (G-protein α subunit interacting protein (GAIP)-interacting protein, C terminus) has been studied in several model organisms, little is known about its role in Caenorhabditis elegans. In the present study, we investigated the roles of gipc-1 and gipc-2 in C. elegans. We observed that they were exclusively expressed in sperm throughout the development and that gipc-1; gipc-2 double mutants were infertile. Further examination of sperm development in gipc-1; gipc-2 mutants revealed defective sperm activation and abnormal pseudopod extension that resulted in reduced sperm motility. Moreover, major sperm protein (MSP) was abnormally segregated between spermatids and residual bodies in gipc-1; gipc-2 mutants. Our findings indicate that gipc-1 and gipc-2 are required for the proper pseudopod extension of sperm during the terminal differentiation of spermatids. During this process, the segregation of MSP into spermatids is important for ensuring normal sperm motility during fertilization.
Collapse
Affiliation(s)
- Jaehoon Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hyemin Min
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, Republic of Korea
| | - Sunhee Ko
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, Republic of Korea
| | - Yhong-Hee Shim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
15
|
Gao M, Mackley IGP, Mesbahi-Vasey S, Bamonte HA, Struyvenberg SA, Landolt L, Pederson NJ, Williams LI, Bahl CD, Brooks L, Amacher JF. Structural characterization and computational analysis of PDZ domains in Monosiga brevicollis. Protein Sci 2020; 29:2226-2244. [PMID: 32914530 DOI: 10.1002/pro.3947] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/22/2022]
Abstract
Identification of the molecular networks that facilitated the evolution of multicellular animals from their unicellular ancestors is a fundamental problem in evolutionary cellular biology. Choanoflagellates are recognized as the closest extant nonmetazoan ancestors to animals. These unicellular eukaryotes can adopt a multicellular-like "rosette" state. Therefore, they are compelling models for the study of early multicellularity. Comparative studies revealed that a number of putative human orthologs are present in choanoflagellate genomes, suggesting that a subset of these genes were necessary for the emergence of multicellularity. However, previous work is largely based on sequence alignments alone, which does not confirm structural nor functional similarity. Here, we focus on the PDZ domain, a peptide-binding domain which plays critical roles in myriad cellular signaling networks and which underwent a gene family expansion in metazoan lineages. Using a customized sequence similarity search algorithm, we identified 178 PDZ domains in the Monosiga brevicollis proteome. This includes 11 previously unidentified sequences, which we analyzed using Rosetta and homology modeling. To assess conservation of protein structure, we solved high-resolution crystal structures of representative M. brevicollis PDZ domains that are homologous to human Dlg1 PDZ2, Dlg1 PDZ3, GIPC, and SHANK1 PDZ domains. To assess functional conservation, we calculated binding affinities for mbGIPC, mbSHANK1, mbSNX27, and mbDLG-3 PDZ domains from M. brevicollis. Overall, we find that peptide selectivity is generally conserved between these two disparate organisms, with one possible exception, mbDLG-3. Overall, our results provide novel insight into signaling pathways in a choanoflagellate model of primitive multicellularity.
Collapse
Affiliation(s)
- Melody Gao
- Department of Chemistry, Western Washington University, Bellingham, Washington, USA
| | - Iain G P Mackley
- Department of Chemistry, Western Washington University, Bellingham, Washington, USA
| | - Samaneh Mesbahi-Vasey
- Institute for Protein Innovation, Boston, Massachusetts, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Haley A Bamonte
- Department of Chemistry, Western Washington University, Bellingham, Washington, USA
| | - Sarah A Struyvenberg
- Department of Chemistry, Western Washington University, Bellingham, Washington, USA
| | - Louisa Landolt
- Department of Chemistry, Western Washington University, Bellingham, Washington, USA
| | - Nick J Pederson
- Department of Chemistry, Western Washington University, Bellingham, Washington, USA
| | - Lucy I Williams
- Department of Chemistry, Western Washington University, Bellingham, Washington, USA
| | - Christopher D Bahl
- Institute for Protein Innovation, Boston, Massachusetts, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Lionel Brooks
- Department of Biology, Western Washington University, Bellingham, Washington, USA
| | - Jeanine F Amacher
- Department of Chemistry, Western Washington University, Bellingham, Washington, USA
| |
Collapse
|
16
|
Deng J, Yu J, Li P, Luan X, Cao L, Zhao J, Yu M, Zhang W, Lv H, Xie Z, Meng L, Zheng Y, Zhao Y, Gang Q, Wang Q, Liu J, Zhu M, Guo X, Su Y, Liang Y, Liang F, Hayashi T, Maeda MH, Sato T, Ura S, Oya Y, Ogasawara M, Iida A, Nishino I, Zhou C, Yan C, Yuan Y, Hong D, Wang Z. Expansion of GGC Repeat in GIPC1 Is Associated with Oculopharyngodistal Myopathy. Am J Hum Genet 2020; 106:793-804. [PMID: 32413282 DOI: 10.1016/j.ajhg.2020.04.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/15/2020] [Indexed: 11/27/2022] Open
Abstract
Oculopharyngodistal myopathy (OPDM) is an adult-onset inherited neuromuscular disorder characterized by progressive ptosis, external ophthalmoplegia, and weakness of the masseter, facial, pharyngeal, and distal limb muscles. The myopathological features are presence of rimmed vacuoles (RVs) in the muscle fibers and myopathic changes of differing severity. Inheritance is variable, with either putative autosomal-dominant or autosomal-recessive pattern. Here, using a comprehensive strategy combining whole-genome sequencing (WGS), long-read whole-genome sequencing (LRS), linkage analysis, repeat-primed polymerase chain reaction (RP-PCR), and fluorescence amplicon length analysis polymerase chain reaction (AL-PCR), we identified an abnormal GGC repeat expansion in the 5' UTR of GIPC1 in one out of four families and three sporadic case subjects from a Chinese OPDM cohort. Expanded GGC repeats were further confirmed as the cause of OPDM in an additional 2 out of 4 families and 6 out of 13 sporadic Chinese individuals with OPDM, as well as 7 out of 194 unrelated Japanese individuals with OPDM. Methylation, qRT-PCR, and western blot analysis indicated that GIPC1 mRNA levels were increased while protein levels were unaltered in OPDM-affected individuals. RNA sequencing indicated p53 signaling, vascular smooth muscle contraction, ubiquitin-mediated proteolysis, and ribosome pathways were involved in the pathogenic mechanisms of OPDM-affected individuals with GGC repeat expansion in GIPC1. This study provides further evidence that OPDM is associated with GGC repeat expansions in distinct genes and highly suggests that expanded GGC repeat units are essential in the pathogenesis of OPDM, regardless of the genes in which the expanded repeats are located.
Collapse
|
17
|
O'Loughlin T, Kendrick-Jones J, Buss F. Approaches to Identify and Characterise MYO6-Cargo Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1239:355-380. [PMID: 32451866 DOI: 10.1007/978-3-030-38062-5_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Given the prevalence and importance of the actin cytoskeleton and the host of associated myosin motors, it comes as no surprise to find that they are linked to a plethora of cellular functions and pathologies. Although our understanding of the biophysical properties of myosin motors has been aided by the high levels of conservation in their motor domains and the extensive work on myosin in skeletal muscle contraction, our understanding of how the nonmuscle myosins participate in such a wide variety of cellular processes is less clear. It is now well established that the highly variable myosin tails are responsible for targeting these myosins to distinct cellular sites for specific functions, and although a number of adaptor proteins have been identified, our current understanding of the cellular processes involved is rather limited. Furthermore, as more adaptor proteins, cargoes and complexes are identified, the importance of elucidating the regulatory mechanisms involved is essential. Ca2+, and now phosphorylation and ubiquitination, are emerging as important regulators of cargo binding, and it is likely that other post-translational modifications are also involved. In the case of myosin VI (MYO6), a number of immediate binding partners have been identified using traditional approaches such as yeast two-hybrid screens and affinity-based pull-downs. However, these methods have only been successful in identifying the cargo adaptors, but not the cargoes themselves, which may often comprise multi-protein complexes. Furthermore, motor-adaptor-cargo interactions are dynamic by nature and often weak, transient and highly regulated and therefore difficult to capture using traditional affinity-based methods. In this chapter we will discuss the various approaches including functional proteomics that have been used to uncover and characterise novel MYO6-associated proteins and complexes and how this work contributes to a fuller understanding of the targeting and function(s) of this unique myosin motor.
Collapse
Affiliation(s)
- Thomas O'Loughlin
- Cambridge Institute for Medical Research, University of Cambridge, The Keith Peters Building, Cambridge, UK
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, The Keith Peters Building, Cambridge, UK.
| |
Collapse
|
18
|
Kottler VA, Feron R, Nanda I, Klopp C, Du K, Kneitz S, Helmprobst F, Lamatsch DK, Lopez-Roques C, Lluch J, Journot L, Parrinello H, Guiguen Y, Schartl M. Independent Origin of XY and ZW Sex Determination Mechanisms in Mosquitofish Sister Species. Genetics 2020; 214:193-209. [PMID: 31704715 PMCID: PMC6944411 DOI: 10.1534/genetics.119.302698] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022] Open
Abstract
Fish are known for the outstanding variety of their sex determination mechanisms and sex chromosome systems. The western (Gambusia affinis) and eastern mosquitofish (G. holbrooki) are sister species for which different sex determination mechanisms have been described: ZZ/ZW for G. affinis and XX/XY for G. holbrooki Here, we carried out restriction-site associated DNA (RAD-) and pool sequencing (Pool-seq) to characterize the sex chromosomes of both species. We found that the ZW chromosomes of G. affinis females and the XY chromosomes of G. holbrooki males correspond to different linkage groups, and thus evolved independently from separate autosomes. In interspecific hybrids, the Y chromosome is dominant over the W chromosome, and X is dominant over Z. In G. holbrooki, we identified a candidate region for the Y-linked melanic pigmentation locus, a rare male phenotype that constitutes a potentially sexually antagonistic trait and is associated with other such characteristics, e.g., large body size and aggressive behavior. We developed a SNP-based marker in the Y-linked allele of GIPC PDZ domain containing family member 1 (gipc1), which was linked to melanism in all tested G. holbrooki populations. This locus represents an example for a color locus that is located in close proximity to a putative sex determiner, and most likely substantially contributed to the evolution of the Y.
Collapse
Affiliation(s)
- Verena A Kottler
- Physiological Chemistry, Biocenter, University of Wuerzburg, 97074, Germany
| | - Romain Feron
- INRA, UR1037 Fish Physiology and Genomics, 35000 Rennes, France
- University of Lausanne and Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Indrajit Nanda
- Institute for Human Genetics, Biocenter, University of Wuerzburg, 97074, Germany
| | - Christophe Klopp
- Sigenae, Mathématiques et Informatique Appliquées de Toulouse, INRA, 31326 Castanet Tolosan, France
| | - Kang Du
- Physiological Chemistry, Biocenter, University of Wuerzburg, 97074, Germany
| | - Susanne Kneitz
- Physiological Chemistry, Biocenter, University of Wuerzburg, 97074, Germany
| | | | - Dunja K Lamatsch
- University of Innsbruck, Research Department for Limnology, Mondsee, 5310 Mondsee, Austria
| | | | - Jerôme Lluch
- INRA, US 1426, GeT-PlaGe, Genotoul, 31326 Castanet-Tolosan, France
| | - Laurent Journot
- Montpellier GenomiX (MGX), University Montpellier, CNRS, INSERM, 34094 France
| | - Hugues Parrinello
- Montpellier GenomiX (MGX), University Montpellier, CNRS, INSERM, 34094 France
| | - Yann Guiguen
- INRA, UR1037 Fish Physiology and Genomics, 35000 Rennes, France
| | - Manfred Schartl
- Physiological Chemistry, Biocenter, University of Wuerzburg, 97074, Germany
- Developmental Biochemistry, Biocenter, University of Wuerzburg, 97074, Germany
- Hagler Institute for Advanced Study and Department of Biology, Texas A&M University, College Station, Texas 77843
| |
Collapse
|
19
|
Wakeham CM, Ren G, Morgans CW. Expression and distribution of trophoblast glycoprotein in the mouse retina. J Comp Neurol 2020; 528:1660-1671. [PMID: 31891182 DOI: 10.1002/cne.24850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 02/03/2023]
Abstract
We recently identified the leucine-rich repeat (LRR) adhesion protein, trophoblast glycoprotein (TPBG), as a novel PKCα-dependent phosphoprotein in retinal rod bipolar cells (RBCs). Since TPBG has not been thoroughly examined in the retina, this study characterizes the localization and expression patterns of TPBG in the developing and adult mouse retina using two antibodies, one against the N-terminal LRR domain and the other against the C-terminal PDZ-interacting motif. Both antibodies labeled RBC dendrites in the outer plexiform layer and axon terminals in the IPL, as well as a putative amacrine cell with their cell bodies in the inner nuclear layer (INL) and a dense layer in the middle of the inner plexiform layer (IPL). In live transfected HEK293 cells, TPBG was localized to the plasma membrane with the N-terminal LRR domain facing the extracellular space. TPBG immunofluorescence in RBCs was strongly altered by the loss of TRPM1 in the adult retina, with significantly less dendritic and axon terminal labeling in TRPM1 knockout compared to wild type, despite no change in total TPBG detected by immunoblotting. During retinal development, TPBG expression increases dramatically just prior to eye opening with a time course closely correlated with that of TRPM1 expression. In the retina, LRR proteins have been implicated in the development and maintenance of functional bipolar cell synapses, and TPBG may play a similar role in RBCs.
Collapse
Affiliation(s)
- Colin M Wakeham
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Gaoying Ren
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Catherine W Morgans
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
20
|
Stanton AE, Tong X, Yang F. Extracellular matrix type modulates mechanotransduction of stem cells. Acta Biomater 2019; 96:310-320. [PMID: 31255664 PMCID: PMC8735670 DOI: 10.1016/j.actbio.2019.06.048] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 12/11/2022]
Abstract
Extracellular matrix (ECM) is comprised of different types of proteins, which change in composition and ratios during morphogenesis and disease progression. ECM proteins provide cell adhesion and impart mechanical cues to the cells. Increasing substrate stiffness has been shown to induce Yes-associated protein (YAP) translocation from the cytoplasm to the nucleus, yet these mechanistic studies used fibronectin only as the biochemical cue. How varying the types of ECM modulates mechanotransduction of stem cells remains largely unknown. Using polyacrylamide hydrogels with tunable stiffness as substrates, here we conjugated four major ECM proteins commonly used for cell adhesion: fibronectin, collagen I, collagen IV and laminin, and assessed the effects of varying ECM type and density on YAP translocation in human mesenchymal stem cells (hMSCs). For all four ECM types, increasing ECM ligand density alone can induce YAP nuclear translocation without changing substrate stiffness. The ligand threshold for such biochemical ligand-induced YAP translocation differs across ECM types. While stiffness-dependent YAP translocation can be induced by all four ECM types, each ECM requires a different optimized ligand density for this to occur. Using antibody blocking, we further identified integrin subunits specifically involved in mechanotransduction of different ECM types. Finally, we demonstrated that altering ECM type further modulates hMSC osteogenesis without changing substrate stiffness. These findings highlight the important role of ECM type in modulating mechanotransduction and differentiation of stem cells, and call for future mechanistic studies to further elucidate the role of changes in ECM compositions in mediating mechanotransduction during morphogenesis and disease progression. STATEMENT OF SIGNIFICANCE: Our study addresses a critical gap of knowledge in mechanobiology. Increasing substrate stiffness has been shown to induce nuclear YAP translocation, yet only on fibronectin-coated substrates. However, extracellular matrix (ECM) is comprised of different protein types. How varying the type of ECM modulates stem cell mechanotransduction remains largely unknown. We here reveal that the choice of ECM type can directly modulate stem cell mechanotransduction, filling this critical gap. This work has broad impacts in mechanobiology and biomaterials, as it provides the first evidence that varying ECM type can impact YAP translocation independent of substrate stiffness, opening doors for a more rational biomaterials design tuning ECM properties to control cell fate for promoting normal development and for preventing disease progression.
Collapse
Affiliation(s)
- Alice E Stanton
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Xinming Tong
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
21
|
Carretero-Ortega J, Chhangawala Z, Hunt S, Narvaez C, Menéndez-González J, Gay CM, Zygmunt T, Li X, Torres-Vázquez J. GIPC proteins negatively modulate Plexind1 signaling during vascular development. eLife 2019; 8:e30454. [PMID: 31050647 PMCID: PMC6499541 DOI: 10.7554/elife.30454] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 04/15/2019] [Indexed: 12/18/2022] Open
Abstract
Semaphorins (SEMAs) and their Plexin (PLXN) receptors are central regulators of metazoan cellular communication. SEMA-PLXND1 signaling plays important roles in cardiovascular, nervous, and immune system development, and cancer biology. However, little is known about the molecular mechanisms that modulate SEMA-PLXND1 signaling. As PLXND1 associates with GIPC family endocytic adaptors, we evaluated the requirement for the molecular determinants of their association and PLXND1's vascular role. Zebrafish that endogenously express a Plxnd1 receptor with a predicted impairment in GIPC binding exhibit low penetrance angiogenesis deficits and antiangiogenic drug hypersensitivity. Moreover, gipc mutant fish show angiogenic impairments that are ameliorated by reducing Plxnd1 signaling. Finally, GIPC depletion potentiates SEMA-PLXND1 signaling in cultured endothelial cells. These findings expand the vascular roles of GIPCs beyond those of the Vascular Endothelial Growth Factor (VEGF)-dependent, proangiogenic GIPC1-Neuropilin 1 complex, recasting GIPCs as negative modulators of antiangiogenic PLXND1 signaling and suggest that PLXND1 trafficking shapes vascular development.
Collapse
Affiliation(s)
- Jorge Carretero-Ortega
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Zinal Chhangawala
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Shane Hunt
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Carlos Narvaez
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Javier Menéndez-González
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Carl M Gay
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Tomasz Zygmunt
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Xiaochun Li
- Department of Population HealthNew York University School of MedicineNew YorkUnited States
| | - Jesús Torres-Vázquez
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| |
Collapse
|
22
|
Grun D, Adhikary G, Eckert RL. NRP-1 interacts with GIPC1 and SYX to activate p38 MAPK signaling and cancer stem cell survival. Mol Carcinog 2019; 58:488-499. [PMID: 30456845 PMCID: PMC6417965 DOI: 10.1002/mc.22943] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 01/13/2023]
Abstract
Epidermal cancer stem cells (ECS cells) comprise a limited population of cells that form aggressive, rapidly growing, and highly vascularized tumors. VEGF-A/NRP-1 signaling is a key driver of the ECS cell phenotype and aggressive tumor formation. However, relatively less is known regarding the downstream events following VEGF-A/NRP-1 interaction. In the present study, we show that VEGF-A/NRP-1, GIPC1, and Syx interact to increase RhoA-dependent p38 MAPK activity to enhance ECS cell spheroid formation, invasion, migration, and angiogenic potential. Inhibition or knockdown of NRP-1, GIPC1 or Syx attenuates RhoA and p38 activity to reduce the ECS cell phenotype, and NRP-1 knockout, or pharmacologic inhibition of VEGF-A/NRP-1 interaction or RhoA activity, reduces p38 MAPK activity and tumor growth. Moreover, expression of wild-type or constitutively-active RhoA, or p38, in NRP1-knockout cells, restores p38 activity and the ECS cell phenotype. These findings suggest that NRP-1 forms a complex with GIPC1 and Syx to activate RhoA/ROCK-dependent p38 activity to enhance the ECS cell phenotype and tumor formation.
Collapse
Affiliation(s)
- Daniel Grun
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Gautam Adhikary
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Richard L Eckert
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Reproductive Biology, University of Maryland School of Medicine, Baltimore, Maryland.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
23
|
Huang C, Yang X, Zeng B, Zeng L, Gong X, Zhou C, Xia J, Lian B, Qin Y, Yang L, Liu L, Xie P. Proteomic analysis of olfactory bulb suggests CACNA1E as a promoter of CREB signaling in microbiota-induced depression. J Proteomics 2019; 194:132-147. [DOI: 10.1016/j.jprot.2018.11.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/24/2018] [Accepted: 11/26/2018] [Indexed: 12/18/2022]
|
24
|
VEGF/Neuropilin Signaling in Cancer Stem Cells. Int J Mol Sci 2019; 20:ijms20030490. [PMID: 30678134 PMCID: PMC6387347 DOI: 10.3390/ijms20030490] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/18/2019] [Accepted: 01/19/2019] [Indexed: 02/07/2023] Open
Abstract
The function of vascular endothelial growth factor (VEGF) in cancer extends beyond angiogenesis and vascular permeability. Specifically, VEGF-mediated signaling occurs in tumor cells and this signaling contributes to key aspects of tumorigenesis including the self-renewal and survival of cancer stem cells (CSCs). In addition to VEGF receptor tyrosine kinases, the neuropilins (NRPs) are critical for mediating the effects of VEGF on CSCs, primarily because of their ability to impact the function of growth factor receptors and integrins. VEGF/NRP signaling can regulate the expression and function of key molecules that have been implicated in CSC function including Rho family guanosine triphosphatases (GTPases) and transcription factors. The VEGF/NRP signaling axis is a prime target for therapy because it can confer resistance to standard chemotherapy, which is ineffective against most CSCs. Indeed, several studies have shown that targeting either NRP1 or NRP2 can inhibit tumor initiation and decrease resistance to other therapies.
Collapse
|
25
|
Kawase J, Aoki JY, Hamada K, Ozaki A, Araki K. Identification of Sex-associated SNPs of Greater Amberjack ( Seriola dumerili). J Genomics 2018; 6:53-62. [PMID: 29861788 PMCID: PMC5970132 DOI: 10.7150/jgen.24788] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 04/25/2018] [Indexed: 11/22/2022] Open
Abstract
The sex determination systems of fish are highly diverse compared with those of mammals. Thus, performing investigations using nonmodel fish species helps to understand the highly diverse sex determination systems of fish. Because greater amberjack (Seriola dumerili) is one of the most important edible fish globally and knowledge of its sex determination system is economically important in the field of aquaculture, we are interested in the mechanisms of sex determination of Seriola species. In this study, we identified sex-associated SNPs of greater amberjack using SNP information of 10 males and 10 females by an association test. We determined that the sex-associated SNPs were on chromosome 12 and mainly covered with two scaffolds (about 7.1 Mbp). Genotypes of sex-associated SNPs indicated that females are the heterogametic sex (ZZ/ZW). Furthermore, we compared the genomic structure of greater amberjack with those of Japanese amberjack (Seriola quinqueradiata), California yellowtail (Seriola dorsalis), and medaka (Oryzias latipes). Whole-genome alignments and synteny analysis indicated that the sex determination system of greater amberjack is markedly different from that of medaka and implied that the sex determination system is conserved in the Seriola species.
Collapse
Affiliation(s)
- Junya Kawase
- Department of Life Sciences, Graduate School of Bioresources, Mie University, 1577 Kurimamachiya-cho, Tsu City, Mie 514-8507, Japan
| | - Jun-Ya Aoki
- Research Center for Aquatic Breeding, National Research Institute of Aquaculture, Fisheries Research Agency, 224 Hiruda, Tamaki-cho, Watarai, Mie 519-0423, Japan
| | - Kazuhisa Hamada
- Marine Farm Laboratory Limited Company, 309 Takahiro, Tachibaura, Otsuki-cho, Hata-gun, Kochi 788-0352, Japan
| | - Akiyuki Ozaki
- Research Center for Aquatic Breeding, National Research Institute of Aquaculture, Fisheries Research Agency, 224 Hiruda, Tamaki-cho, Watarai, Mie 519-0423, Japan
| | - Kazuo Araki
- Research Center for Aquatic Breeding, National Research Institute of Aquaculture, Fisheries Research Agency, 224 Hiruda, Tamaki-cho, Watarai, Mie 519-0423, Japan
| |
Collapse
|
26
|
Pinskey JM, Franks NE, McMellen AN, Giger RJ, Allen BL. Neuropilin-1 promotes Hedgehog signaling through a novel cytoplasmic motif. J Biol Chem 2017; 292:15192-15204. [PMID: 28667171 DOI: 10.1074/jbc.m117.783845] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/23/2017] [Indexed: 12/30/2022] Open
Abstract
Hedgehog (HH) signaling critically regulates embryonic and postnatal development as well as adult tissue homeostasis, and its perturbation can lead to developmental disorders, birth defects, and cancers. Neuropilins (NRPs), which have well-defined roles in Semaphorin and VEGF signaling, positively regulate HH pathway function, although their mechanism of action in HH signaling remains unclear. Here, using luciferase-based reporter assays, we provide evidence that NRP1 regulates HH signaling specifically at the level of GLI transcriptional activator function. Moreover, we show that NRP1 localization to the primary cilium, a key platform for HH signal transduction, does not correlate with HH signal promotion. Rather, a structure-function analysis suggests that the NRP1 cytoplasmic and transmembrane domains are necessary and sufficient to regulate HH pathway activity. Furthermore, we identify a previously uncharacterized, 12-amino acid region within the NRP1 cytoplasmic domain that mediates HH signal promotion. Overall, our results provide mechanistic insight into NRP1 function within and potentially beyond the HH signaling pathway. These insights have implications for the development of novel modulators of HH-driven developmental disorders and diseases.
Collapse
Affiliation(s)
- Justine M Pinskey
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Nicole E Franks
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Alexandra N McMellen
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Roman J Giger
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Benjamin L Allen
- From the Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
27
|
Zhang G, Chen L, Sun K, Khan AA, Yan J, Liu H, Lu A, Gu N. Neuropilin-1 (NRP-1)/GIPC1 pathway mediates glioma progression. Tumour Biol 2016; 37:13777-13788. [PMID: 27481513 DOI: 10.1007/s13277-016-5138-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 07/11/2016] [Indexed: 12/21/2022] Open
Abstract
Glioma occurs due to multi-gene abnormalities. Neuropilin-1 (NRP-1), as a transmembrane protein, involves in glioma proliferation, invasion, and migration, as well as tumor angiogenesis. The cytoplasmic protein, GAIP/RGS19-interacting protein (GIPC1), could regulate the clathrin-vesicles trafficking and recycling. Here, we show that NRP-1 co-localizes and co-immunoprecipitates with GIPC1, and the C-terminal SEA-COOH motif of NRP-1 interacts specially with the named from three proteins: PSD-95 (a 95 kDa protein involved in signaling at the post-synaptic density), DLG (the Drosophila melanogaster Discs Large protein) and ZO-1 (the zonula occludens 1 protein involved in maintenance of epithelial polarity) (PDZ) domain of GIPC1 in glioma cells. Knockdown of GIPC1 by small interfering RNA (siRNA) significantly reduces the proliferation and invasion of glioma cells in vitro and increases its apoptosis. Furthermore, si-GIPC1 prevents the action of adaptor proteins adaptor protein, phosphotyrosine interaction, PH domain and leucine zipper containing 1 (APPL1) and p130Cas and inhibits the downstream kirsten rat sarcoma viral oncogene homolog (KRAS)-ERK signaling pathway. This study demonstrated that NRP-1/GIPC1 pathway plays a vital role in glioma progression, and it is a potential important target for multi-gene combined therapeutics.
Collapse
Affiliation(s)
- Guilong Zhang
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Lukui Chen
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| | - Kouhong Sun
- Nanjing Zoonbio Biotechnology, Nanjing, 210014, China
| | - Ahsan Ali Khan
- Department of Neurosurgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jianghua Yan
- Cancer Research Center, Xiamen University, Xiamen, 361000, China
| | - Hongyi Liu
- Department of Neurosurgery, Nanjing Brain Hospital, Nanjing, 210029, China
| | - Ailin Lu
- Department of Neurosurgery, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Ning Gu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
28
|
Hayes MP, Roman DL. Regulator of G Protein Signaling 17 as a Negative Modulator of GPCR Signaling in Multiple Human Cancers. AAPS JOURNAL 2016; 18:550-9. [PMID: 26928451 DOI: 10.1208/s12248-016-9894-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/15/2016] [Indexed: 02/08/2023]
Abstract
Regulators of G protein signaling (RGS) proteins modulate G protein-coupled receptor (GPCR) signaling networks by terminating signals produced by active Gα subunits. RGS17, a member of the RZ subfamily of RGS proteins, is typically only expressed in appreciable amounts in the human central nervous system, but previous works have shown that RGS17 expression is selectively upregulated in a number of malignancies, including lung, breast, prostate, and hepatocellular carcinoma. In addition, this upregulation of RGS17 is associated with a more aggressive cancer phenotype, as increased proliferation, migration, and invasion are observed. Conversely, decreased RGS17 expression diminishes the response of ovarian cancer cells to agents commonly used during chemotherapy. These somewhat contradictory roles of RGS17 in cancer highlight the need for selective, high-affinity inhibitors of RGS17 to use as chemical probes to further the understanding of RGS17 biology. Based on current evidence, these compounds could potentially have clinical utility as novel chemotherapeutics in the treatment of lung, prostate, breast, and liver cancers. Recent advances in screening technologies to identify potential inhibitors coupled with increasing knowledge of the structural requirements of RGS-Gα protein-protein interaction inhibitors make the future of drug discovery efforts targeting RGS17 promising. This review highlights recent findings related to RGS17 as both a canonical and atypical RGS protein, its role in various human disease states, and offers insights on small molecule inhibition of RGS17.
Collapse
Affiliation(s)
- Michael P Hayes
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa, USA
| | - David L Roman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa, USA. .,Cancer Signaling and Experimental Therapeutics Program, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA. .,, 115 S. Grand Avenue, S327 PHAR, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
29
|
A GIPC1-Palmitate Switch Modulates Dopamine Drd3 Receptor Trafficking and Signaling. Mol Cell Biol 2016; 36:1019-31. [PMID: 26787837 DOI: 10.1128/mcb.00916-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/06/2016] [Indexed: 12/27/2022] Open
Abstract
Palmitoylation is involved in several neuropsychiatric and movement disorders for which a dysfunctional signaling of the dopamine D3 receptor (Drd3) is hypothesized. Computational modeling of Drd3's homologue, Drd2, has shed some light on the putative role of palmitoylation as a reversible switch for dopaminergic receptor signaling. Drd3 is presumed to be palmitoylated, based on sequence homology with Drd2, but the functional attributes afforded by Drd3 palmitoylation have not been studied. Since these receptors are major targets of antipsychotic and anti-Parkinsonian drugs, a better characterization of Drd3 signaling and posttranslational modifications, like palmitoylation, may improve the prospects for drug development. Using molecular dynamics simulations, we evaluated in silico how Drd3 palmitoylation could elicit significant remodeling of the C-terminal cytoplasmic domain to expose docking sites for signaling proteins. We tested this model in cellulo by using the interaction of Drd3 with the G-alpha interacting protein (GAIP) C terminus 1 (GIPC1) as a template. From a series of biochemical studies, live imaging, and analyses of mutant proteins, we propose that Drd3 palmitoylation acts as a molecular switch for Drd3-biased signaling via a GIPC1-dependent route, which is likely to affect the mode of action of antipsychotic drugs.
Collapse
|
30
|
Koyama T, Ozaki A, Yoshida K, Suzuki J, Fuji K, Aoki JY, Kai W, Kawabata Y, Tsuzaki T, Araki K, Sakamoto T. Identification of Sex-Linked SNPs and Sex-Determining Regions in the Yellowtail Genome. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2015; 17:502-510. [PMID: 25975833 DOI: 10.1007/s10126-015-9636-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/15/2015] [Indexed: 06/04/2023]
Abstract
Unlike the conservation of sex-determining (SD) modes seen in most mammals and birds, teleost fishes exhibit a wide variety of SD systems and genes. Hence, the study of SD genes and sex chromosome turnover in fish is one of the most interesting topics in evolutionary biology. To increase resolution of the SD gene evolutionary trajectory in fish, identification of the SD gene in more fish species is necessary. In this study, we focused on the yellowtail, a species widely cultivated in Japan. It is a member of family Carangidae in which no heteromorphic sex chromosome has been observed, and no SD gene has been identified to date. By performing linkage analysis and BAC walking, we identified a genomic region and SNPs with complete linkage to yellowtail sex. Comparative genome analysis revealed the yellowtail SD region ancestral chromosome structure as medaka-fugu. Two inversions occurred in the yellowtail linage after it diverged from the yellowtail-medaka ancestor. An association study using wild yellowtails and the SNPs developed from BAC ends identified two SNPs that can reasonably distinguish the sexes. Therefore, these will be useful genetic markers for yellowtail breeding. Based on a comparative study, it was suggested that a PDZ domain containing the GIPC protein might be involved in yellowtail sex determination. The homomorphic sex chromosomes widely observed in the Carangidae suggest that this family could be a suitable marine fish model to investigate the early stages of sex chromosome evolution, for which our results provide a good starting point.
Collapse
Affiliation(s)
- Takashi Koyama
- Faculty of Marine Science, Tokyo University of Marine Science and Technology, 4-5-7, Konan, Minato-ku, Tokyo, 108-8477, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yoshida A, Shimizu A, Asano H, Kadonosono T, Kondoh SK, Geretti E, Mammoto A, Klagsbrun M, Seo MK. VEGF-A/NRP1 stimulates GIPC1 and Syx complex formation to promote RhoA activation and proliferation in skin cancer cells. Biol Open 2015. [PMID: 26209534 PMCID: PMC4582117 DOI: 10.1242/bio.010918] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Neuropilin-1 (NRP1) has been identified as a VEGF-A receptor. DJM-1, a human skin cancer cell line, expresses endogenous VEGF-A and NRP1. In the present study, the RNA interference of VEGF-A or NRP1 suppressed DJM-1 cell proliferation. Furthermore, the overexpression of the NRP1 wild type restored shNRP1-treated DJM-1 cell proliferation, whereas NRP1 cytoplasmic deletion mutants did not. A co-immunoprecipitation analysis revealed that VEGF-A induced interactions between NRP1 and GIPC1, a scaffold protein, and complex formation between GIPC1 and Syx, a RhoGEF. The knockdown of GIPC1 or Syx reduced active RhoA and DJM-1 cell proliferation without affecting the MAPK or Akt pathway. C3 exoenzyme or Y27632 inhibited the VEGF-A-induced proliferation of DJM-1 cells. Conversely, the overexpression of the constitutively active form of RhoA restored the proliferation of siVEGF-A-treated DJM-1 cells. Furthermore, the inhibition of VEGF-A/NRP1 signaling upregulated p27, a CDK inhibitor. A cell-penetrating oligopeptide that targeted GIPC1/Syx complex formation inhibited the VEGF-A-induced activation of RhoA and suppressed DJM-1 cell proliferation. In conclusion, this new signaling pathway of VEGF-A/NRP1 induced cancer cell proliferation by forming a GIPC1/Syx complex that activated RhoA to degrade the p27 protein.
Collapse
Affiliation(s)
- Ayumi Yoshida
- Division of Engineering (Biotechnology), Graduate School of Engineering, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Akio Shimizu
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8047, Japan
| | - Hirotsugu Asano
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8047, Japan
| | - Tetsuya Kadonosono
- Biofunctional Engineering, Graduate School of Bioscience & Biotechnology, Tokyo Institute of Technology, Tokyo 226-8503, Japan
| | - Shinae Kizaka Kondoh
- Biofunctional Engineering, Graduate School of Bioscience & Biotechnology, Tokyo Institute of Technology, Tokyo 226-8503, Japan
| | - Elena Geretti
- Vascular Biology Program, Boston Children's Hospital, Departments of Surgery and Pathology and Harvard Medical School, Boston, MA 02115, USA
| | - Akiko Mammoto
- Vascular Biology Program, Boston Children's Hospital, Departments of Surgery and Pathology and Harvard Medical School, Boston, MA 02115, USA
| | - Michael Klagsbrun
- Vascular Biology Program, Boston Children's Hospital, Departments of Surgery and Pathology and Harvard Medical School, Boston, MA 02115, USA
| | - Misuzu Kurokawa Seo
- Division of Engineering (Biotechnology), Graduate School of Engineering, Kyoto Sangyo University, Kyoto 603-8555, Japan Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8047, Japan
| |
Collapse
|
32
|
Woodard GE, Jardín I, Berna-Erro A, Salido GM, Rosado JA. Regulators of G-protein-signaling proteins: negative modulators of G-protein-coupled receptor signaling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 317:97-183. [PMID: 26008785 DOI: 10.1016/bs.ircmb.2015.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regulators of G-protein-signaling (RGS) proteins are a category of intracellular proteins that have an inhibitory effect on the intracellular signaling produced by G-protein-coupled receptors (GPCRs). RGS along with RGS-like proteins switch on through direct contact G-alpha subunits providing a variety of intracellular functions through intracellular signaling. RGS proteins have a common RGS domain that binds to G alpha. RGS proteins accelerate GTPase and thus enhance guanosine triphosphate hydrolysis through the alpha subunit of heterotrimeric G proteins. As a result, they inactivate the G protein and quickly turn off GPCR signaling thus terminating the resulting downstream signals. Activity and subcellular localization of RGS proteins can be changed through covalent molecular changes to the enzyme, differential gene splicing, and processing of the protein. Other roles of RGS proteins have shown them to not be solely committed to being inhibitors but behave more as modulators and integrators of signaling. RGS proteins modulate the duration and kinetics of slow calcium oscillations and rapid phototransduction and ion signaling events. In other cases, RGS proteins integrate G proteins with signaling pathways linked to such diverse cellular responses as cell growth and differentiation, cell motility, and intracellular trafficking. Human and animal studies have revealed that RGS proteins play a vital role in physiology and can be ideal targets for diseases such as those related to addiction where receptor signaling seems continuously switched on.
Collapse
Affiliation(s)
- Geoffrey E Woodard
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Isaac Jardín
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - A Berna-Erro
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - Gines M Salido
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - Juan A Rosado
- Department of Physiology, University of Extremadura, Caceres, Spain
| |
Collapse
|
33
|
Plein A, Fantin A, Ruhrberg C. Neuropilin regulation of angiogenesis, arteriogenesis, and vascular permeability. Microcirculation 2015; 21:315-23. [PMID: 24521511 PMCID: PMC4230468 DOI: 10.1111/micc.12124] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 02/05/2014] [Indexed: 12/11/2022]
Abstract
The formation of the cardiovasculature, consisting of both the heart and blood vessels, is a critical step in embryonic development and relies on three processes termed vasculogenesis, angiogenesis, and vascular remodeling. The transmembrane protein NRP1 is an essential modulator of embryonic angiogenesis with additional roles in vessel remodeling and arteriogenesis. NRP1 also enhances arteriogenesis in adults to alleviate pathological tissue ischemia. However, in certain circumstances, vascular NRP1 signaling can be detrimental, as it may promote cancer by enhancing tumor angiogenesis or contribute to tissue edema by increasing vascular permeability. Understanding the mechanisms of NRP1 signaling is, therefore, of profound importance for the design of therapies aiming to control vascular functions. Previous work has shown that vascular NRP1 can variably serve as a receptor for two secreted glycoproteins, the VEGF-A and SEMA3A, but it also has a poorly understood role as an adhesion receptor. Here, we review current knowledge of NRP1 function during blood vessel growth and homeostasis, with special emphasis on the vascular roles of its multiple ligands and signaling partners.
Collapse
Affiliation(s)
- Alice Plein
- UCL Institute of Ophthalmology, University College London, London, UK
| | | | | |
Collapse
|
34
|
Bhattacharya S, Pal K, Sharma AK, Dutta SK, Lau JS, Yan IK, Wang E, Elkhanany A, Alkharfy KM, Sanyal A, Patel TC, Chari ST, Spaller MR, Mukhopadhyay D. GAIP interacting protein C-terminus regulates autophagy and exosome biogenesis of pancreatic cancer through metabolic pathways. PLoS One 2014; 9:e114409. [PMID: 25469510 PMCID: PMC4255029 DOI: 10.1371/journal.pone.0114409] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 11/07/2014] [Indexed: 12/14/2022] Open
Abstract
GAIP interacting protein C terminus (GIPC) is known to play an important role in a variety of physiological and disease states. In the present study, we have identified a novel role for GIPC as a master regulator of autophagy and the exocytotic pathways in cancer. We show that depletion of GIPC-induced autophagy in pancreatic cancer cells, as evident from the upregulation of the autophagy marker LC3II. We further report that GIPC regulates cellular trafficking pathways by modulating the secretion, biogenesis, and molecular composition of exosomes. We also identified the involvement of GIPC on metabolic stress pathways regulating autophagy and microvesicular shedding, and observed that GIPC status determines the loading of cellular cargo in the exosome. Furthermore, we have shown the overexpression of the drug resistance gene ABCG2 in exosomes from GIPC-depleted pancreatic cancer cells. We also demonstrated that depletion of GIPC from cancer cells sensitized them to gemcitabine treatment, an avenue that can be explored as a potential therapeutic strategy to overcome drug resistance in cancer.
Collapse
Affiliation(s)
- Santanu Bhattacharya
- Department Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Krishnendu Pal
- Department Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Anil K. Sharma
- Department Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Shamit K. Dutta
- Department Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Julie S. Lau
- Department Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Irene K. Yan
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Enfeng Wang
- Department Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ahmed Elkhanany
- Department Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Khalid M. Alkharfy
- Department Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Arunik Sanyal
- Department Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Tushar C. Patel
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Suresh T. Chari
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Mark R. Spaller
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, and Norris Cotton Cancer Center, Lebanon, New Hampshire, United States of America
| | - Debabrata Mukhopadhyay
- Department Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
35
|
Zwarts L, Van Eijs F, Callaerts P. Glia in Drosophila behavior. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2014; 201:879-93. [PMID: 25336160 DOI: 10.1007/s00359-014-0952-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 10/02/2014] [Accepted: 10/07/2014] [Indexed: 02/06/2023]
Abstract
Glial cells constitute about 10 % of the Drosophila nervous system. The development of genetic and molecular tools has helped greatly in defining different types of glia. Furthermore, considerable progress has been made in unraveling the mechanisms that control the development and differentiation of Drosophila glia. By contrast, the role of glia in adult Drosophila behavior is not well understood. We here summarize recent work describing the role of glia in normal behavior and in Drosophila models for neurological and behavioral disorders.
Collapse
Affiliation(s)
- L Zwarts
- Laboratory of Behavioral and Developmental Genetics VIB Center for the Biology of Disease, Center for Human Genetics, KULeuven, O&N IV Herestraat 49, Box 602, 3000, Louvain, Belgium
| | | | | |
Collapse
|
36
|
Pang HB, Braun GB, Friman T, Aza-Blanc P, Ruidiaz ME, Sugahara KN, Teesalu T, Ruoslahti E. An endocytosis pathway initiated through neuropilin-1 and regulated by nutrient availability. Nat Commun 2014; 5:4904. [PMID: 25277522 PMCID: PMC4185402 DOI: 10.1038/ncomms5904] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 08/02/2014] [Indexed: 12/16/2022] Open
Abstract
Neuropilins (NRPs) are trans-membrane receptors involved in axon guidance and vascular development. Many growth factors and other signalling molecules bind to NRPs through a carboxy (C)-terminal, basic sequence motif (C-end Rule or CendR motif). Peptides with this motif (CendR peptides) are taken up into cells by endocytosis. Tumour-homing CendR peptides penetrate through tumour tissue and have shown utility in enhancing drug delivery into tumours. Here we show, using RNAi screening and subsequent validation studies, that NRP1-mediated endocytosis of CendR peptides is distinct from known endocytic pathways. Ultrastructurally, CendR endocytosis resembles macropinocytosis, but is mechanistically different. We also show that nutrient-sensing networks such as mTOR signalling regulate CendR endocytosis and subsequent intercellular transport of CendR cargo, both of which are stimulated by nutrient depletion. As CendR is a bulk transport pathway, our results suggest a role for it in nutrient transport; CendR-enhanced drug delivery then makes use of this natural pathway.
Collapse
Affiliation(s)
- Hong-Bo Pang
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, USA
| | - Gary B. Braun
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, USA
- Center for Nanomedicine, and Department of Cell, Molecular and Developmental
Biology, University of California Santa Barbara, Santa Barbara, CA 93106-9610, USA
| | - Tomas Friman
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, USA
- Center for Nanomedicine, and Department of Cell, Molecular and Developmental
Biology, University of California Santa Barbara, Santa Barbara, CA 93106-9610, USA
| | - Pedro Aza-Blanc
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, USA
| | - Manuel E. Ruidiaz
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, USA
| | - Kazuki N. Sugahara
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, USA
- Department of Surgery, Columbia University, College of Physicians and
Surgeons, New York, NY 10032, USA
| | - Tambet Teesalu
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, USA
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational
Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Tartu,
50411, Estonia
| | - Erkki Ruoslahti
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La
Jolla, California 92037, USA
- Center for Nanomedicine, and Department of Cell, Molecular and Developmental
Biology, University of California Santa Barbara, Santa Barbara, CA 93106-9610, USA
| |
Collapse
|
37
|
Pal K, Pletnev AA, Dutta SK, Wang E, Zhao R, Baral A, Yadav VK, Aggarwal S, Krishnaswamy S, Alkharfy KM, Chowdhury S, Spaller MR, Mukhopadhyay D. Inhibition of endoglin-GIPC interaction inhibits pancreatic cancer cell growth. Mol Cancer Ther 2014; 13:2264-75. [PMID: 25125675 PMCID: PMC4229952 DOI: 10.1158/1535-7163.mct-14-0291] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endoglin, a 180-kDa disulfide-linked homodimeric transmembrane receptor protein mostly expressed in tumor-associated endothelial cells, is an endogenous binding partner of GAIP-interacting protein, C terminus (GIPC). Endoglin functions as a coreceptor of TβRII that binds TGFβ and is important for vascular development, and consequently has become a compelling target for antiangiogenic therapies. A few recent studies in gastrointestinal stromal tumor (GIST), breast cancer, and ovarian cancer, however, suggest that endoglin is upregulated in tumor cells and is associated with poor prognosis. These findings indicate a broader role of endoglin in tumor biology, beyond angiogenic effects. The goal of our current study is to evaluate the effects of targeting endoglin in pancreatic cancer both in vitro and in vivo. We analyzed the antiproliferative effect of both RNAi-based and peptide ligand-based inhibition of endoglin in pancreatic cancer cell lines, the latter yielding a GIPC PDZ domain-targeting lipopeptide with notable antiproliferative activity. We further demonstrated that endoglin inhibition induced a differentiation phenotype in the pancreatic cancer cells and sensitized them against conventional chemotherapeutic drug gemcitabine. Most importantly, we have demonstrated the antitumor effect of both RNAi-based and competitive inhibitor-based blocking of endoglin in pancreatic cancer xenograft models in vivo. To our knowledge, this is the first report exploring the effect of targeting endoglin in pancreatic cancer cells.
Collapse
Affiliation(s)
- Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Alexandre A Pletnev
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, Lebanon, New Hampshire
| | - Shamit K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Ruizhi Zhao
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, Lebanon, New Hampshire
| | - Aradhita Baral
- Proteomics and Structural Biology Unit, Institute of Genomics and Integrative Biology, Council for Scientific and Industrial Research, New Delhi, India
| | - Vinod Kumar Yadav
- G.N.R. Knowledge Center for Genome Informatics, Institute of Genomics and Integrative Biology, Council for Scientific and Industrial Research, New Delhi, India
| | - Suruchi Aggarwal
- G.N.R. Knowledge Center for Genome Informatics, Institute of Genomics and Integrative Biology, Council for Scientific and Industrial Research, New Delhi, India
| | | | - Khalid M Alkharfy
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota. Department of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Shantanu Chowdhury
- Proteomics and Structural Biology Unit, Institute of Genomics and Integrative Biology, Council for Scientific and Industrial Research, New Delhi, India. G.N.R. Knowledge Center for Genome Informatics, Institute of Genomics and Integrative Biology, Council for Scientific and Industrial Research, New Delhi, India
| | - Mark R Spaller
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth and Norris Cotton Cancer Center, Lebanon, New Hampshire
| | | |
Collapse
|
38
|
Wang Q, Terauchi A, Yee CH, Umemori H, Traynor JR. 5-HT1A receptor-mediated phosphorylation of extracellular signal-regulated kinases (ERK1/2) is modulated by regulator of G protein signaling protein 19. Cell Signal 2014; 26:1846-52. [PMID: 24793302 PMCID: PMC8019269 DOI: 10.1016/j.cellsig.2014.04.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 04/27/2014] [Indexed: 12/31/2022]
Abstract
The 5-HT1A receptor is a G protein coupled receptor (GPCR) that activates G proteins of the Gαi/o family. 5-HT1A receptors expressed in the raphe, hippocampus and prefrontal cortex are implicated in the control of mood and are targets for anti-depressant drugs. Regulators of G protein signaling (RGS) proteins are members of a large family that play important roles in signal transduction downstream of G protein coupled receptors (GPCRs). The main role of RGS proteins is to act as GTPase accelerating proteins (GAPs) to dampen or negatively regulate GPCR-mediated signaling. We have shown that a mouse expressing Gαi2 that is insensitive to all RGS protein GAP activity has an anti-depressant-like phenotype due to increased signaling of postsynaptic 5-HT1A receptors, thus implicating the 5-HT1A receptor-Gαi2 complex as an important target. Here we confirm that RGS proteins act as GAPs to regulate signaling to adenylate cyclase and the mitogen-activated protein kinase (MAPK) pathway downstream of the 5-HT1A receptor, using RGS-insensitive Gαi2 protein expressed in C6 cells. We go on to use short hairpin RNA (shRNA) to show that RGS19 is responsible for the GAP activity in C6 cells and also that RGS19 acts as a GAP for 5-HT1A receptor signaling in human neuroblastoma SH-SY5Y cells and primary hippocampal neurons. In addition, in both cell types the synergy between 5-HT1A receptor and the fibroblast growth factor receptor 1 in stimulating the MAPK pathway is enhanced following shRNA reduction of RGS19 expression. Thus RGS19 may be a viable new target for anti-depressant medications.
Collapse
Affiliation(s)
- Qin Wang
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Akiko Terauchi
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Christopher H Yee
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Hisashi Umemori
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - John R Traynor
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
39
|
Zhao Y, Malinauskas T, Harlos K, Jones EY. Structural insights into the inhibition of Wnt signaling by cancer antigen 5T4/Wnt-activated inhibitory factor 1. Structure 2014; 22:612-20. [PMID: 24582434 PMCID: PMC3988984 DOI: 10.1016/j.str.2014.01.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/30/2013] [Accepted: 01/13/2014] [Indexed: 01/10/2023]
Abstract
The tumor antigen 5T4/WAIF1 (Wnt-activated inhibitory factor 1; also known as Trophoblast glycoprotein TPBG) is a cell surface protein targeted in multiple cancer immunotherapy clinical trials. Recently, it has been shown that 5T4/WAIF1 inhibits Wnt/β-catenin signaling, a signaling system central to many developmental and pathological processes. Wnt/β-catenin signaling is controlled by multiple inhibitors and activators. Here, we report crystal structures for the extracellular domain of 5T4/WAIF1 at 1.8 Å resolution. They reveal a highly glycosylated, rigid core, comprising eight leucine-rich repeats (LRRs), which serves as a platform to present evolutionarily conserved surface residues in the N-terminal LRR1. Structural and cell-based analyses, coupled with previously reported in vivo data, suggest that Tyr325 plus the LRR1 surface centered on a second exposed aromatic residue, Phe97, are essential for inhibition of Wnt/β-catenin signaling. These results provide a structural basis for the development of 5T4/WAIF1-targeted therapies that preserve or block 5T4/WAIF1-mediated inhibition of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yuguang Zhao
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Tomas Malinauskas
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Karl Harlos
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK.
| |
Collapse
|
40
|
Chak K, Kolodkin AL. Function of the Drosophila receptor guanylyl cyclase Gyc76C in PlexA-mediated motor axon guidance. Development 2014; 141:136-47. [PMID: 24284209 PMCID: PMC3865755 DOI: 10.1242/dev.095968] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 09/26/2013] [Indexed: 12/17/2022]
Abstract
The second messengers cAMP and cGMP modulate attraction and repulsion mediated by neuronal guidance cues. We find that the Drosophila receptor guanylyl cyclase Gyc76C genetically interacts with Semaphorin 1a (Sema-1a) and physically associates with the Sema-1a receptor plexin A (PlexA). PlexA regulates Gyc76C catalytic activity in vitro, and each distinct Gyc76C protein domain is crucial for regulating Gyc76C activity in vitro and motor axon guidance in vivo. The cytosolic protein dGIPC interacts with Gyc76C and facilitates Sema-1a-PlexA/Gyc76C-mediated motor axon guidance. These findings provide an in vivo link between semaphorin-mediated repulsive axon guidance and alteration of intracellular neuronal cGMP levels.
Collapse
Affiliation(s)
- Kayam Chak
- The Solomon H. Snyder Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alex L. Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
41
|
Abstract
The function of vascular endothelial growth factor (VEGF) in cancer is not limited to angiogenesis and vascular permeability. VEGF-mediated signalling occurs in tumour cells, and this signalling contributes to key aspects of tumorigenesis, including the function of cancer stem cells and tumour initiation. In addition to VEGF receptor tyrosine kinases, the neuropilins are crucial for mediating the effects of VEGF on tumour cells, primarily because of their ability to regulate the function and the trafficking of growth factor receptors and integrins. This has important implications for our understanding of tumour biology and for the development of more effective therapeutic approaches.
Collapse
Affiliation(s)
- Hira Lal Goel
- Department of Cancer Biology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | | |
Collapse
|
42
|
Kermit interacts with Gαo, Vang, and motor proteins in Drosophila planar cell polarity. PLoS One 2013; 8:e76885. [PMID: 24204696 PMCID: PMC3805608 DOI: 10.1371/journal.pone.0076885] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/28/2013] [Indexed: 11/20/2022] Open
Abstract
In addition to the ubiquitous apical-basal polarity, epithelial cells are often polarized within the plane of the tissue – the phenomenon known as planar cell polarity (PCP). In Drosophila, manifestations of PCP are visible in the eye, wing, and cuticle. Several components of the PCP signaling have been characterized in flies and vertebrates, including the heterotrimeric Go protein. However, Go signaling partners in PCP remain largely unknown. Using a genetic screen we uncover Kermit, previously implicated in G protein and PCP signaling, as a novel binding partner of Go. Through pull-down and genetic interaction studies, we find that Kermit interacts with Go and another PCP component Vang, known to undergo intracellular relocalization during PCP establishment. We further demonstrate that the activity of Kermit in PCP differentially relies on the motor proteins: the microtubule-based dynein and kinesin motors and the actin-based myosin VI. Our results place Kermit as a potential transducer of Go, linking Vang with motor proteins for its delivery to dedicated cellular compartments during PCP establishment.
Collapse
|
43
|
Lanahan A, Zhang X, Fantin A, Zhuang Z, Rivera-Molina F, Speichinger K, Prahst C, Zhang J, Wang Y, Davis G, Toomre D, Ruhrberg C, Simons M. The neuropilin 1 cytoplasmic domain is required for VEGF-A-dependent arteriogenesis. Dev Cell 2013; 25:156-68. [PMID: 23639442 PMCID: PMC3774154 DOI: 10.1016/j.devcel.2013.03.019] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 01/25/2013] [Accepted: 03/27/2013] [Indexed: 12/19/2022]
Abstract
Neuropilin 1 (NRP1) plays an important but ill-defined role in VEGF-A signaling and vascular morphogenesis. We show that mice with a knockin mutation that ablates the NRP1 cytoplasmic tail (Nrp1cyto) have normal angiogenesis but impaired developmental and adult arteriogenesis. The arteriogenic defect was traced to the absence of a PDZ-dependent interaction between NRP1 and VEGF receptor 2 (VEGFR2) complex and synectin, which delayed trafficking of endocytosed VEGFR2 from Rab5+ to EAA1+ endosomes. This led to increased PTPN1 (PTP1b)-mediated dephosphorylation of VEGFR2 at Y1175, the site involved in activating ERK signaling. The Nrp1cyto mutation also impaired endothelial tubulogenesis in vitro, which could be rescued by expressing full-length NRP1 or constitutively active ERK. These results demonstrate that the NRP1 cytoplasmic domain promotes VEGFR2 trafficking in a PDZ-dependent manner to regulate arteriogenic ERK signaling and establish a role for NRP1 in VEGF-A signaling during vascular morphogenesis. The NRP1 cytoplasmic domain promotes VEGF receptor (VEGFR) 2 endocytic trafficking In its absence, VEGR2 trafficking is delayed in sorting endosomes PTP1b binds to Rab5+ sorting endosomes and dephosphorylates the Y1175 site of VEGFR2 Loss of the NRP1 cytoplasmic domain impairs developmental and adult arteriogenesis
Collapse
Affiliation(s)
- Anthony Lanahan
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Tumbarello DA, Kendrick-Jones J, Buss F. Myosin VI and its cargo adaptors - linking endocytosis and autophagy. J Cell Sci 2013; 126:2561-70. [PMID: 23781020 DOI: 10.1242/jcs.095554] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The coordinated trafficking and tethering of membrane cargo within cells relies on the function of distinct cytoskeletal motors that are targeted to specific subcellular compartments through interactions with protein adaptors and phospholipids. The unique actin motor myosin VI functions at distinct steps during clathrin-mediated endocytosis and the early endocytic pathway - both of which are involved in cargo trafficking and sorting - through interactions with Dab2, GIPC, Tom1 and LMTK2. This multifunctional ability of myosin VI can be attributed to its cargo-binding tail region that contains two protein-protein interaction interfaces, a ubiquitin-binding motif and a phospholipid binding domain. In addition, myosin VI has been shown to be a regulator of the autophagy pathway, because of its ability to link the endocytic and autophagic pathways through interactions with the ESCRT-0 protein Tom1 and the autophagy adaptor proteins T6BP, NDP52 and optineurin. This function has been attributed to facilitating autophagosome maturation and subsequent fusion with the lysosome. Therefore, in this Commentary, we discuss the relationship between myosin VI and the different myosin VI adaptor proteins, particularly with regards to the spatial and temporal regulation that is required for the sorting of cargo at the early endosome, and their impact on autophagy.
Collapse
Affiliation(s)
- David A Tumbarello
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | | | | |
Collapse
|
45
|
Katoh M. Functional proteomics, human genetics and cancer biology of GIPC family members. Exp Mol Med 2013; 45:e26. [PMID: 23743496 PMCID: PMC3701287 DOI: 10.1038/emm.2013.49] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 04/04/2013] [Indexed: 12/24/2022] Open
Abstract
GIPC1, GIPC2 and GIPC3 consist of GIPC homology 1 (GH1) domain, PDZ domain and GH2 domain. The regions around the GH1 and GH2 domains of GIPC1 are involved in dimerization and interaction with myosin VI (MYO6), respectively. The PDZ domain of GIPC1 is involved in interactions with transmembrane proteins [IGF1R, NTRK1, ADRB1, DRD2, TGFβR3 (transforming growth factorβ receptor type III), SDC4, SEMA4C, LRP1, NRP1, GLUT1, integrin α5 and VANGL2], cytosolic signaling regulators (APPL1 and RGS19) and viral proteins (HBc and HPV-18 E6). GIPC1 is an adaptor protein with dimerizing ability that loads PDZ ligands as cargoes for MYO6-dependent endosomal trafficking. GIPC1 is required for cell-surface expression of IGF1R and TGFβR3. GIPC1 is also required for integrin recycling during cell migration, angiogenesis and cytokinesis. On early endosomes, GIPC1 assembles receptor tyrosine kinases (RTKs) and APPL1 for activation of PI3K-AKT signaling, and G protein-coupled receptors (GPCRs) and RGS19 for attenuation of inhibitory Gα signaling. GIPC1 upregulation in breast, ovarian and pancreatic cancers promotes tumor proliferation and invasion, whereas GIPC1 downregulation in cervical cancer with human papillomavirus type 18 infection leads to resistance to cytostatic transforming growth factorβ signaling. GIPC2 is downregulated in acute lymphocytic leukemia owing to epigenetic silencing, while Gipc2 is upregulated in estrogen-induced mammary tumors. Somatic mutations of GIPC2 occur in malignant melanoma, and colorectal and ovarian cancers. Germ-line mutations of the GIPC3 or MYO6 gene cause nonsyndromic hearing loss. As GIPC proteins are involved in trafficking, signaling and recycling of RTKs, GPCRs, integrins and other transmembrane proteins, dysregulation of GIPCs results in human pathologies, such as cancer and hereditary deafness.
Collapse
Affiliation(s)
- Masaru Katoh
- Division of Integrative Omics and Bioinformatics, National Cancer Centre, Tokyo, Japan.
| |
Collapse
|
46
|
Wang Q, Traynor JR. Modulation of μ-opioid receptor signaling by RGS19 in SH-SY5Y cells. Mol Pharmacol 2013; 83:512-20. [PMID: 23197645 PMCID: PMC3558815 DOI: 10.1124/mol.112.081992] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 11/29/2012] [Indexed: 02/03/2023] Open
Abstract
Regulator of G-protein signaling protein 19 (RGS19), also known as Gα-interacting protein (GAIP), acts as a GTPase accelerating protein for Gαz as well as Gαi/o subunits. Interactions with GAIP-interacting protein N-terminus and GAIP-interacting protein C-terminus (GIPC) link RGS19 to a variety of intracellular proteins. Here we show that RGS19 is abundantly expressed in human neuroblastoma SH-SY5Y cells that also express µ- and δ- opioid receptors (MORs and DORs, respectively) and nociceptin receptors (NOPRs). Lentiviral delivery of short hairpin RNA specifically targeted to RGS19 reduced RGS19 protein levels by 69%, with a similar reduction in GIPC. In RGS19-depleted cells, there was an increase in the ability of MOR (morphine) but not of DOR [(4-[(R)-[(2S,5R)-4-allyl-2,5-dimethylpiperazin-1-yl](3-methoxyphenyl)methyl]-N,N-diethylbenzamide (SNC80)] or NOPR (nociceptin) agonists to inhibit forskolin-stimulated adenylyl cyclase and increase mitogen-activated protein kinase (MAPK) activity. Overnight treatment with either MOR [D-Ala, N-Me-Phe, Gly-ol(5)-enkephalin (DAMGO) or morphine] or DOR (D-Pen(5)-enkephalin or SNC80) agonists increased RGS19 and GIPC protein levels in a time- and concentration-dependent manner. The MOR-induced increase in RGS19 protein was prevented by pretreatment with pertussis toxin or the opioid antagonist naloxone. Protein kinase C (PKC) activation alone increased the level of RGS19 and inhibitors of PKC 5,6,7,13-tetrahydro-13-methyl-5-oxo-12H-indolo[2,3-a]pyrrolo[3,4-c]carbazole-12-propanenitrile and mitogen-activated protein kinase kinase 1 2-(2-amino-3-methoxyphenyl)-4H-chromen-4-one, but not protein kinase A (H89), completely blocked DAMGO-induced RGS19 protein accumulation. The findings show that RGS19 and GIPC are jointly regulated, that RGS19 is a GTPase accelerating protein for MOR with selectivity over DOR and NOPR, and that chronic MOR or DOR agonist treatment increases RGS19 levels by a PKC and the MAPK pathway-dependent mechanism.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adenylyl Cyclases/genetics
- Adenylyl Cyclases/metabolism
- Animals
- Benzamides/pharmacology
- Colforsin/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- HEK293 Cells
- Humans
- Mitogen-Activated Protein Kinases/genetics
- Mitogen-Activated Protein Kinases/metabolism
- Morphine/pharmacology
- Opioid Peptides/pharmacology
- PC12 Cells
- Piperazines/pharmacology
- Protein Kinase C/genetics
- Protein Kinase C/metabolism
- RGS Proteins/genetics
- RGS Proteins/metabolism
- Rats
- Receptors, Opioid/genetics
- Receptors, Opioid/metabolism
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Signal Transduction/drug effects
- Nociceptin Receptor
- Nociceptin
Collapse
Affiliation(s)
- Qin Wang
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109-5632, USA
| | | |
Collapse
|
47
|
Abstract
Vascular endothelial growth factors (VEGF) and their receptors play a central role in the development of cardiovascular system and in vasculature-related processes in the adult organism. Given the critical role of this signaling cascade, intricate control systems have evolved to regulate its function. A new layer of added complexity has been the demonstration of the importance of endocytosis and intracellular trafficking of VEGF receptors in the regulation of VEGF signaling. In this review, we consider an evolving link between VEGF receptor endocytosis, trafficking, and signaling and their biological function.
Collapse
Affiliation(s)
- Michael Simons
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut, USA.
| |
Collapse
|
48
|
Giese AP, Ezan J, Wang L, Lasvaux L, Lembo F, Mazzocco C, Richard E, Reboul J, Borg JP, Kelley MW, Sans N, Brigande J, Montcouquiol M. Gipc1 has a dual role in Vangl2 trafficking and hair bundle integrity in the inner ear. Development 2012; 139:3775-85. [PMID: 22991442 DOI: 10.1242/dev.074229] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Vangl2 is one of the central proteins controlling the establishment of planar cell polarity in multiple tissues of different species. Previous studies suggest that the localization of the Vangl2 protein to specific intracellular microdomains is crucial for its function. However, the molecular mechanisms that control Vangl2 trafficking within a cell are largely unknown. Here, we identify Gipc1 (GAIP C-terminus interacting protein 1) as a new interactor for Vangl2, and we show that a myosin VI-Gipc1 protein complex can regulate Vangl2 traffic in heterologous cells. Furthermore, we show that in the cochlea of MyoVI mutant mice, Vangl2 presence at the membrane is increased, and that a disruption of Gipc1 function in hair cells leads to maturation defects, including defects in hair bundle orientation and integrity. Finally, stimulated emission depletion microscopy and overexpression of GFP-Vangl2 show an enrichment of Vangl2 on the supporting cell side, adjacent to the proximal membrane of hair cells. Altogether, these results indicate a broad role for Gipc1 in the development of both stereociliary bundles and cell polarization, and suggest that the strong asymmetry of Vangl2 observed in early postnatal cochlear epithelium is mostly a 'tissue' polarity readout.
Collapse
Affiliation(s)
- Arnaud P Giese
- Planar Polarity and Plasticity Group, Inserm U862, Neurocentre Magendie, Bordeaux, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Varsano T, Taupin V, Guo L, Baterina OY, Farquhar MG. The PDZ protein GIPC regulates trafficking of the LPA1 receptor from APPL signaling endosomes and attenuates the cell's response to LPA. PLoS One 2012; 7:e49227. [PMID: 23145131 PMCID: PMC3493537 DOI: 10.1371/journal.pone.0049227] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 10/07/2012] [Indexed: 12/21/2022] Open
Abstract
Lysophosphatidic acid (LPA) mediates diverse cellular responses through the activation of at least six LPA receptors – LPA1–6, but the interacting proteins and signaling pathways that mediate the specificity of these receptors are largely unknown. We noticed that LPA1 contains a PDZ binding motif (SVV) identical to that present in two other proteins that interact with the PDZ protein GIPC. GIPC is involved in endocytic trafficking of several receptors including TrkA, VEGFR2, lutropin and dopamine D2 receptors. Here we show that GIPC binds directly to the PDZ binding motif of LPA1 but not that of other LPA receptors. LPA1 colocalizes and coimmunoprecipitates with GIPC and its binding partner APPL, an activator of Akt signaling found on APPL signaling endosomes. GIPC depletion by siRNA disturbed trafficking of LPA1 to EEA1 early endosomes and promoted LPA1 mediated Akt signaling, cell proliferation, and cell motility. We propose that GIPC binds LPA1 and promotes its trafficking from APPL-containing signaling endosomes to EEA1 early endosomes and thus attenuates LPA-mediated Akt signaling from APPL endosomes.
Collapse
Affiliation(s)
- Tal Varsano
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Vanessa Taupin
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Lixia Guo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Oscar Y. Baterina
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Marilyn G. Farquhar
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
50
|
Mu Y, Huang H, Liu S, Cai P, Gao Y. Molecular characterization and ligand binding specificity of the PDZ domain-containing protein GIPC3 from Schistosoma japonicum. Parasit Vectors 2012; 5:227. [PMID: 23050840 PMCID: PMC3504512 DOI: 10.1186/1756-3305-5-227] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 10/05/2012] [Indexed: 01/09/2023] Open
Abstract
Background Schistosomiasis is a serious global health problem that afflicts more than 230 million people in 77 countries. Long-term mass treatments with the only available drug, praziquantel, have caused growing concerns about drug resistance. PSD-95/Dlg/ZO-1 (PDZ) domain-containing proteins are recognized as potential targets for the next generation of drug development. However, the PDZ domain-containing protein family in parasites has largely been unexplored. Methods We present the molecular characteristics of a PDZ domain-containing protein, GIPC3, from Schistosoma japonicum (SjGIPC3) according to bioinformatics analysis and experimental approaches. The ligand binding specificity of the PDZ domain of SjGIPC3 was confirmed by screening an arbitrary peptide library in yeast two-hybrid (Y2H) assays. The native ligand candidates were predicted by Tailfit software based on the C-terminal binding specificity, and further validated by Y2H assays. Results SjGIPC3 is a single PDZ domain-containing protein comprised of 328 amino acid residues. Structural prediction revealed that a conserved PDZ domain was presented in the middle region of the protein. Phylogenetic analysis revealed that SjGIPC3 and other trematode orthologues clustered into a well-defined cluster but were distinguishable from those of other phyla. Transcriptional analysis by quantitative RT-PCR revealed that the SjGIPC3 gene was relatively highly expressed in the stages within the host, especially in male adult worms. By using Y2H assays to screen an arbitrary peptide library, we confirmed the C-terminal binding specificity of the SjGIPC3-PDZ domain, which could be deduced as a consensus sequence, -[SDEC]-[STIL]-[HSNQDE]-[VIL]*. Furthermore, six proteins were predicted to be native ligand candidates of SjGIPC3 based on the C-terminal binding properties and other biological information; four of these were confirmed to be potential ligands using the Y2H system. Conclusions In this study, we first characterized a PDZ domain-containing protein GIPC3 in S. japonicum. The SjGIPC3-PDZ domain is able to bind both type I and II ligand C-terminal motifs. The identification of native ligand will help reveal the potential biological function of SjGIPC3. These data will facilitate the identification of novel drug targets against S. japonicum infections.
Collapse
Affiliation(s)
- Yi Mu
- National Key Laboratory of Medical Molecular Biology, Dept, of Physiology and Pathophysiology, School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, P,R, China
| | | | | | | | | |
Collapse
|