1
|
Wang T, Zhou X, Chen M, Li Y, Li M, Wang R, Guo R, Gong S, Liu K. Downregulation of Dmxl2 disrupts the hearing development in mice. Neuroscience 2025; 573:322-332. [PMID: 40118164 DOI: 10.1016/j.neuroscience.2025.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025]
Abstract
Congenital hearing loss is a major type of sensorineural deafness. Recently, Dmxl2 has been identified as a new gene associated with familial deafness. However, its role in auditory development remains unclear. This study investigated the expression and localization of DmX-like protein 2 (DMXL2), encoded by Dmxl2, in the mouse cochlea at various postnatal stages. DMXL2 was predominantly expressed in inner and outer hair cells, with the highest levels at postnatal day 7, followed by a rapid decline, nearly disappearing by day 14. To elucidate Dmxl2's function, we administered short hairpin RNA (shRNA) targeting Dmxl2 to the cochlea within 24 h post-birth, effectively knocking down its expression in the mouse inner ear. This resulted in profound hearing loss in treated mice, accompanied by disruption of development of cochlear ribbon synapses and spiral ganglion cells (SGCs). In conclusion, our study demonstrates the critical role of Dmxl2 in hearing development, suggesting it as a potential molecular target for future gene therapy in hearing loss treatment.
Collapse
Affiliation(s)
- Tianying Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China.
| | - Xuan Zhou
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China.
| | - Minglin Chen
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China.
| | - Yang Li
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China.
| | - Menghua Li
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China.
| | - Rong Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China.
| | - Rui Guo
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China.
| | - Shusheng Gong
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China.
| | - Ke Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Clinical Center for Hearing Loss, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
2
|
Winkley SR, Kane PM. The ROGDI protein mutated in Kohlschutter-Tonz syndrome is a novel subunit of the Rabconnectin-3 complex implicated in V-ATPase assembly. J Biol Chem 2025; 301:108381. [PMID: 40049412 PMCID: PMC11997317 DOI: 10.1016/j.jbc.2025.108381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/11/2025] [Accepted: 03/01/2025] [Indexed: 04/01/2025] Open
Abstract
V-ATPases are highly conserved ATP-driven rotary proton pumps found widely among eukaryotes that are composed of two subcomplexes: V1 and V0. V-ATPase activity is regulated in part through reversible disassembly, during which V1 physically separates from V0 and both subcomplexes become inactive. Reassociation of V1 to V0 reactivates the complex for ATP-driven proton pumping and organelle acidification. V-ATPase reassembly in Saccharomyces cerevisiae requires the RAVE complex (Rav1, Rav2, and Skp1), and higher eukaryotes, including humans, utilize the Rabconnectin-3 complex. Mammalian Rabconnectin-3 has two subunits: Rabconnectin-3α and Rabconnectin-3β. Rabconnectin-3α isoforms are homologous to Rav1, but there is no known Rav2 homolog, and the molecular basis of the interaction between the Rabconnectin-3α and β subunits is unknown. We identified ROGDI as a Rav2 homolog and novel Rabconnectin-3 subunit. ROGDI mutations cause Kohlschutter-Tonz syndrome, an epileptic encephalopathy with amelogenesis imperfecta that has parallels to V-ATPase-related disease. ROGDI shares extensive structural homology with yeast Rav2 and can functionally replace Rav2 in yeast. ROGDI binds to the N-terminal domains of both Rabconnectin-3 α and β, similar to Rav2 binding to Rav1. Molecular modeling suggests that ROGDI may bridge the two Rabconnectin-3 subunits. ROGDI coimmunoprecipitates with Rabconnectin-3 subunits from detergent-solubilized lysates and is present with them in immunopurified lysosomes of mammalian cells. In immunofluorescence microscopy, ROGDI partially localizes with Rabconnectin-3α in acidic perinuclear lysosomes. The discovery of ROGDI as a novel Rabconnectin-3 interactor sheds new light on both Kohlschutter-Tonz syndrome and the mechanisms behind mammalian V-ATPase regulation.
Collapse
Affiliation(s)
- Samuel R Winkley
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Patricia M Kane
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.
| |
Collapse
|
3
|
Kobia FM, Castro E Almeida L, Paganoni AJ, Carminati F, Andronache A, Lavezzari F, Wade M, Vaccari T. Novel determinants of NOTCH1 trafficking and signaling in breast epithelial cells. Life Sci Alliance 2025; 8:e202403122. [PMID: 39663000 PMCID: PMC11633778 DOI: 10.26508/lsa.202403122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024] Open
Abstract
The evolutionarily conserved Notch signaling pathway controls cell-cell communication, enacting cell fate decisions during development and tissue homeostasis. Its dysregulation is associated with a wide range of diseases, including congenital disorders and cancers. Signaling outputs depend on maturation of Notch receptors and trafficking to the plasma membrane, endocytic uptake and sorting, lysosomal and proteasomal degradation, and ligand-dependent and independent proteolytic cleavages. We devised assays to follow quantitatively the trafficking and signaling of endogenous human NOTCH1 receptor in breast epithelial cells in culture. Based on such analyses, we executed a high-content screen of 2,749 human genes to identify new regulators of Notch that might be amenable to pharmacologic intervention. We uncovered 39 new NOTCH1 modulators for NOTCH1 trafficking and signaling. Among them, we find that PTPN23 and HCN2 act as positive NOTCH1 regulators by promoting endocytic trafficking and NOTCH1 maturation in the Golgi apparatus, respectively, whereas SGK3 serves as a negative regulator that can be modulated by pharmacologic inhibition. Our findings might be relevant in the search of new strategies to counteract pathologic Notch signaling.
Collapse
Affiliation(s)
- Francis M Kobia
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | | | - Alyssa Jj Paganoni
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | | | - Adrian Andronache
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | | | - Mark Wade
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Thomas Vaccari
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
4
|
Eaton AF, Danielson EC, Capen D, Merkulova M, Brown D. Dmxl1 Is an Essential Mammalian Gene that Is Required for V-ATPase Assembly and Function In Vivo. FUNCTION 2024; 5:zqae025. [PMID: 38984989 PMCID: PMC11237898 DOI: 10.1093/function/zqae025] [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: 04/16/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 07/11/2024] Open
Abstract
The proton pumping V-ATPase drives essential biological processes, such as acidification of intracellular organelles. Critically, the V-ATPase domains, V1 and VO, must assemble to produce a functional holoenzyme. V-ATPase dysfunction results in cancer, neurodegeneration, and diabetes, as well as systemic acidosis caused by reduced activity of proton-secreting kidney intercalated cells (ICs). However, little is known about the molecular regulation of V-ATPase in mammals. We identified a novel interactor of the mammalian V-ATPase, Drosophila melanogaster X chromosomal gene-like 1 (Dmxl1), aka Rabconnectin-3A. The yeast homologue of Dmxl1, Rav1p, is part of a complex that catalyzes the reversible assembly of the domains. We, therefore,hypothesized that Dmxl1 is a mammalian V-ATPase assembly factor. Here, we generated kidney IC-specific Dmxl1 knockout (KO) mice, which had high urine pH, like B1 V-ATPase KO mice, suggesting impaired V-ATPase function. Western blotting showed decreased B1 expression and B1 (V1) and a4 (VO) subunits were more intracellular and less colocalized in Dmxl1 KO ICs. In parallel, subcellular fractionation revealed less V1 associated B1 in the membrane fraction of KO cells relative to the cytosol. Furthermore, a proximity ligation assay performed using probes against B1 and a4 V-ATPase subunits also revealed decreased association. We propose that loss of Dmxl1 reduces V-ATPase holoenzyme assembly, thereby inhibiting proton pumping function. Dmxl1 may recruit the V1 domain to the membrane and facilitate assembly with the VO domain and in its absence V1 may be targeted for degradation. We conclude that Dmxl1 is a bona fide mammalian V-ATPase assembly factor.
Collapse
Affiliation(s)
- Amity F Eaton
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Elizabeth C Danielson
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Diane Capen
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Maria Merkulova
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dennis Brown
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
5
|
Akuwudike P, López-Riego M, Marczyk M, Kocibalova Z, Brückner F, Polańska J, Wojcik A, Lundholm L. Short- and long-term effects of radiation exposure at low dose and low dose rate in normal human VH10 fibroblasts. Front Public Health 2023; 11:1297942. [PMID: 38162630 PMCID: PMC10755029 DOI: 10.3389/fpubh.2023.1297942] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Experimental studies complement epidemiological data on the biological effects of low doses and dose rates of ionizing radiation and help in determining the dose and dose rate effectiveness factor. Methods Human VH10 skin fibroblasts exposed to 25, 50, and 100 mGy of 137Cs gamma radiation at 1.6, 8, 12 mGy/h, and at a high dose rate of 23.4 Gy/h, were analyzed for radiation-induced short- and long-term effects. Two sample cohorts, i.e., discovery (n = 30) and validation (n = 12), were subjected to RNA sequencing. The pool of the results from those six experiments with shared conditions (1.6 mGy/h; 24 h), together with an earlier time point (0 h), constituted a third cohort (n = 12). Results The 100 mGy-exposed cells at all abovementioned dose rates, harvested at 0/24 h and 21 days after exposure, showed no strong gene expression changes. DMXL2, involved in the regulation of the NOTCH signaling pathway, presented a consistent upregulation among both the discovery and validation cohorts, and was validated by qPCR. Gene set enrichment analysis revealed that the NOTCH pathway was upregulated in the pooled cohort (p = 0.76, normalized enrichment score (NES) = 0.86). Apart from upregulated apical junction and downregulated DNA repair, few pathways were consistently changed across exposed cohorts. Concurringly, cell viability assays, performed 1, 3, and 6 days post irradiation, and colony forming assay, seeded just after exposure, did not reveal any statistically significant early effects on cell growth or survival patterns. Tendencies of increased viability (day 6) and reduced colony size (day 21) were observed at 12 mGy/h and 23.4 Gy/min. Furthermore, no long-term changes were observed in cell growth curves generated up to 70 days after exposure. Discussion In conclusion, low doses of gamma radiation given at low dose rates had no strong cytotoxic effects on radioresistant VH10 cells.
Collapse
Affiliation(s)
- Pamela Akuwudike
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Milagrosa López-Riego
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Michal Marczyk
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, United States
| | - Zuzana Kocibalova
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Fabian Brückner
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Joanna Polańska
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Andrzej Wojcik
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | - Lovisa Lundholm
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
6
|
Hounjet J, Groot AJ, Piepers JP, Kranenburg O, Zwijnenburg DA, Rapino FA, Koster JB, Kampen KR, Vooijs MA. Iron-responsive element of Divalent metal transporter 1 (Dmt1) controls Notch-mediated cell fates. FEBS J 2023; 290:5811-5834. [PMID: 37646174 DOI: 10.1111/febs.16946] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/12/2023] [Accepted: 08/29/2023] [Indexed: 09/01/2023]
Abstract
Notch receptor activation is regulated by the intramembrane protease γ-secretase, which cleaves and liberates the Notch intracellular domain (Nicd) that regulates gene transcription. While γ-secretase cleavage is necessary, we demonstrate it is insufficient for Notch activation and requires vesicular trafficking. Here, we report Divalent metal transporter 1 (Dmt1, Slc11A2) as a novel and essential regulator of Notch signalling. Dmt1-deficient cells are defective in Notch signalling and have perturbed endolysosomal trafficking and function. Dmt1 encodes for two isoforms, with and without an iron response element (ire). We show that isoform-specific silencing of Dmt1-ire and Dmt1+ire has opposite consequences on Notch-dependent cell fates in cell lines and intestinal organoids. Loss of Dmt1-ire suppresses Notch activation and promotes differentiation, whereas loss of Dmt1+ire causes Notch activation and maintains stem-progenitor cell fates. Dmt1 isoform expression correlates with Notch and Wnt signalling in Apc-deficient intestinal organoids and human colorectal cancers. Consistently, Dmt1-ire silencing induces Notch-dependent differentiation in colorectal cancer cells. These data identify Dmt1 isoforms as binary switches controlling Notch cell fate decisions in normal and tumour cells.
Collapse
Affiliation(s)
- Judith Hounjet
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Arjan J Groot
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jolanda P Piepers
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Onno Kranenburg
- Lab Translational Oncology, Division Imaging and Cancer, University Medical Center Utrecht, The Netherlands
| | - Danny A Zwijnenburg
- Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, The Netherlands
| | - Francesca A Rapino
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Pharmacy, Giga Stem Cells, University of Liege, Belgium
| | - Jan B Koster
- Center for Experimental and Molecular Medicine, Amsterdam UMC Location University of Amsterdam, The Netherlands
| | - Kim R Kampen
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Marc A Vooijs
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
7
|
Wang H, Rubinstein JL. CryoEM of V-ATPases: Assembly, disassembly, and inhibition. Curr Opin Struct Biol 2023; 80:102592. [PMID: 37272327 DOI: 10.1016/j.sbi.2023.102592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 06/06/2023]
Abstract
Vacuolar-type ATPases (V-ATPases) are responsible for the acidification of intracellular compartments in almost all eukaryotic cells, while in some specialized cells they acidify the extracellular environment. As ubiquitous proton pumps, these large membrane-embedded enzymes are involved in many fundamental cellular processes that require tight control of pH. Consequently, V-ATPase malfunction or aberrant activity has been linked to numerous diseases. In the past ten years, electron cryomicroscopy (cryoEM) of yeast V-ATPases has revealed the architecture and rotary catalytic mechanism of these macromolecular machines. More recently, studies have revealed the structures of V-ATPases in animals and plants, uncovered aspects of how V-ATPases are assembled and regulated by reversible dissociation, and shown how V-ATPase activity can be modulated by proteins and small molecule inhibitors. In this review, we highlight these recent developments.
Collapse
Affiliation(s)
- Hanlin Wang
- Molecular Medicine Program, The Hospital for Sick Children, M5G 0A4, Toronto, Canada; Department of Biochemistry, The University of Toronto, M5G 1L7, Toronto, Canada
| | - John L Rubinstein
- Molecular Medicine Program, The Hospital for Sick Children, M5G 0A4, Toronto, Canada; Department of Biochemistry, The University of Toronto, M5G 1L7, Toronto, Canada; Department of Medical Biophysics, The University of Toronto, M5S 1A8, Toronto, Canada.
| |
Collapse
|
8
|
Su K, Collins MP, McGuire CM, Alshagawi MA, Alamoudi MK, Li Z, Forgac M. Isoform a4 of the vacuolar ATPase a subunit promotes 4T1-12B breast cancer cell-dependent tumor growth and metastasis in vivo. J Biol Chem 2022; 298:102395. [PMID: 35988642 PMCID: PMC9508560 DOI: 10.1016/j.jbc.2022.102395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 12/24/2022] Open
Abstract
The vacuolar H+-ATPase (V-ATPase) is an ATP-dependent proton pump that governs the pH of various intracellular compartments and also functions at the plasma membrane in certain cell types, including cancer cells. Membrane targeting of the V-ATPase is controlled by isoforms of subunit a, and we have previously shown that isoforms a3 and a4 are important for the migration and invasion of several breast cancer cell lines in vitro. Using CRISPR-mediated genome editing to selectively disrupt each of the four a subunit isoforms, we also recently showed that a4 is critical to plasma membrane V-ATPase localization, as well as in vitro migration and invasion of 4T1-12B murine breast cancer cells. We now report that a4 is important for the growth of 4T1-12B tumors in vivo. We found that BALB/c mice bearing a4-/- 4T1-12B allografts had significantly smaller tumors than mice in the control group. In addition, we determined that a4-/- allografts showed dramatically reduced metastases to the lung and reduced luminescence intensity of metastases to bone relative to the control group. Taken together, these results suggest that the a4 isoform of the V-ATPase represents a novel potential therapeutic target to limit breast cancer growth and metastasis.
Collapse
Affiliation(s)
- Kevin Su
- Department of Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Michael P Collins
- Department of Cellular, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Christina M McGuire
- Department of Biochemistry, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Mohammed A Alshagawi
- Department of Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Mariam K Alamoudi
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Zhen Li
- Department of Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Michael Forgac
- Department of Pharmacology and Drug Development, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA; Department of Cellular, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA; Department of Biochemistry, Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA; Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
9
|
She J, Su D, Diao R, Wang L. A Joint Model of Random Forest and Artificial Neural Network for the Diagnosis of Endometriosis. Front Genet 2022; 13:848116. [PMID: 35350240 PMCID: PMC8957986 DOI: 10.3389/fgene.2022.848116] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Endometriosis (EM), an estrogen-dependent inflammatory disease with unknown etiology, affects thousands of childbearing-age couples, and its early diagnosis is still very difficult. With the rapid development of sequencing technology in recent years, the accumulation of many sequencing data makes it possible to screen important diagnostic biomarkers from some EM-related genes. In this study, we utilized public datasets in the Gene Expression Omnibus (GEO) and Array-Express database and identified seven important differentially expressed genes (DEGs) (COMT, NAA16, CCDC22, EIF3E, AHI1, DMXL2, and CISD3) through the random forest classifier. Among these DEGs, AHI1, DMXL2, and CISD3 have never been reported to be associated with the pathogenesis of EMs. Our study indicated that these three genes might participate in the pathogenesis of EMs through oxidative stress, epithelial–mesenchymal transition (EMT) with the activation of the Notch signaling pathway, and mitochondrial homeostasis, respectively. Then, we put these seven DEGs into an artificial neural network to construct a novel diagnostic model for EMs and verified its diagnostic efficacy in two public datasets. Furthermore, these seven DEGs were included in 15 hub genes identified from the constructed protein–protein interaction (PPI) network, which confirmed the reliability of the diagnostic model. We hope the diagnostic model can provide novel sights into the understanding of the pathogenesis of EMs and contribute to the clinical diagnosis and treatment of EMs.
Collapse
Affiliation(s)
- Jiajie She
- Reproductive Medicine Centre, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Danna Su
- Reproductive Medicine Centre, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Ruiying Diao
- Reproductive Medicine Centre, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Liping Wang
- Reproductive Medicine Centre, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
10
|
Ion Channels and Pumps in Autophagy: A Reciprocal Relationship. Cells 2021; 10:cells10123537. [PMID: 34944044 PMCID: PMC8700256 DOI: 10.3390/cells10123537] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
Autophagy, the process of cellular self-degradation, is intrinsically tied to the degradative function of the lysosome. Several diseases have been linked to lysosomal degradative defects, including rare lysosomal storage disorders and neurodegenerative diseases. Ion channels and pumps play a major regulatory role in autophagy. Importantly, calcium signaling produced by TRPML1 (transient receptor potential cation channel, mucolipin subfamily) has been shown to regulate autophagic progression through biogenesis of autophagic-lysosomal organelles, activation of mTORC1 (mechanistic target of rapamycin complex 1) and degradation of autophagic cargo. ER calcium channels such as IP3Rs supply calcium for the lysosome, and lysosomal function is severely disrupted in the absence of lysosomal calcium replenishment by the ER. TRPML1 function is also regulated by LC3 (microtubule-associated protein light chain 3) and mTORC1, two critical components of the autophagic network. Here we provide an overview of the current knowledge about ion channels and pumps-including lysosomal V-ATPase (vacuolar proton-ATPase), which is required for acidification and hence proper enzymatic activity of lysosomal hydrolases-in the regulation of autophagy, and discuss how functional impairment of some of these leads to diseases.
Collapse
|
11
|
Jaskolka MC, Winkley SR, Kane PM. RAVE and Rabconnectin-3 Complexes as Signal Dependent Regulators of Organelle Acidification. Front Cell Dev Biol 2021; 9:698190. [PMID: 34249946 PMCID: PMC8264551 DOI: 10.3389/fcell.2021.698190] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
The yeast RAVE (Regulator of H+-ATPase of Vacuolar and Endosomal membranes) complex and Rabconnectin-3 complexes of higher eukaryotes regulate acidification of organelles such as lysosomes and endosomes by catalyzing V-ATPase assembly. V-ATPases are highly conserved proton pumps consisting of a peripheral V1 subcomplex that contains the sites of ATP hydrolysis, attached to an integral membrane Vo subcomplex that forms the transmembrane proton pore. Reversible disassembly of the V-ATPase is a conserved regulatory mechanism that occurs in response to multiple signals, serving to tune ATPase activity and compartment acidification to changing extracellular conditions. Signals such as glucose deprivation can induce release of V1 from Vo, which inhibits both ATPase activity and proton transport. Reassembly of V1 with Vo restores ATP-driven proton transport, but requires assistance of the RAVE or Rabconnectin-3 complexes. Glucose deprivation triggers V-ATPase disassembly in yeast and is accompanied by binding of RAVE to V1 subcomplexes. Upon glucose readdition, RAVE catalyzes both recruitment of V1 to the vacuolar membrane and its reassembly with Vo. The RAVE complex can be recruited to the vacuolar membrane by glucose in the absence of V1 subunits, indicating that the interaction between RAVE and the Vo membrane domain is glucose-sensitive. Yeast RAVE complexes also distinguish between organelle-specific isoforms of the Vo a-subunit and thus regulate distinct V-ATPase subpopulations. Rabconnectin-3 complexes in higher eukaryotes appear to be functionally equivalent to yeast RAVE. Originally isolated as a two-subunit complex from rat brain, the Rabconnectin-3 complex has regions of homology with yeast RAVE and was shown to interact with V-ATPase subunits and promote endosomal acidification. Current understanding of the structure and function of RAVE and Rabconnectin-3 complexes, their interactions with the V-ATPase, their role in signal-dependent modulation of organelle acidification, and their impact on downstream pathways will be discussed.
Collapse
Affiliation(s)
- Michael C Jaskolka
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Samuel R Winkley
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Patricia M Kane
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
12
|
Schneider L, Guo Y, Birch D, Sarkies P. Network-based visualisation reveals new insights into transposable element diversity. Mol Syst Biol 2021; 17:e9600. [PMID: 34169647 PMCID: PMC8226279 DOI: 10.15252/msb.20209600] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/23/2021] [Accepted: 05/27/2021] [Indexed: 01/12/2023] Open
Abstract
Transposable elements (TEs) are widespread across eukaryotic genomes, yet their content varies widely between different species. Factors shaping the diversity of TEs are poorly understood. Understanding the evolution of TEs is difficult because their sequences diversify rapidly and TEs are often transferred through non-conventional means such as horizontal gene transfer. We developed a method to track TE evolution using network analysis to visualise TE sequence and TE content across different genomes. We illustrate our method by first using a monopartite network to study the sequence evolution of Tc1/mariner elements across focal species. We identify a connection between two subfamilies associated with convergent acquisition of a domain from a protein-coding gene. Second, we use a bipartite network to study how TE content across species is shaped by epigenetic silencing mechanisms. We show that the presence of Piwi-interacting RNAs is associated with differences in network topology after controlling for phylogenetic effects. Together, our method demonstrates how a network-based approach can identify hitherto unknown properties of TE evolution across species.
Collapse
Affiliation(s)
- Lisa Schneider
- MRC London Institute of Medical SciencesLondonUK
- Institute of Clinical SciencesImperial CollegeLondonUK
- Data Sciences InstituteImperial CollegeLondonUK
| | - Yi‐Ke Guo
- Data Sciences InstituteImperial CollegeLondonUK
- Present address:
Hong Kong Baptist UniversityKowloon TsaiHong Kong
| | - David Birch
- Data Sciences InstituteImperial CollegeLondonUK
| | - Peter Sarkies
- MRC London Institute of Medical SciencesLondonUK
- Institute of Clinical SciencesImperial CollegeLondonUK
| |
Collapse
|
13
|
Breyer F, Härtlova A, Thurston T, Flynn HR, Chakravarty P, Janzen J, Peltier J, Heunis T, Snijders AP, Trost M, Ley SC. TPL-2 kinase induces phagosome acidification to promote macrophage killing of bacteria. EMBO J 2021; 40:e106188. [PMID: 33881780 PMCID: PMC8126920 DOI: 10.15252/embj.2020106188] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 02/05/2023] Open
Abstract
Tumour progression locus 2 (TPL‐2) kinase mediates Toll‐like receptor (TLR) activation of ERK1/2 and p38α MAP kinases in myeloid cells to modulate expression of key cytokines in innate immunity. This study identified a novel MAP kinase‐independent regulatory function for TPL‐2 in phagosome maturation, an essential process for killing of phagocytosed microbes. TPL‐2 catalytic activity was demonstrated to induce phagosome acidification and proteolysis in primary mouse and human macrophages following uptake of latex beads. Quantitative proteomics revealed that blocking TPL‐2 catalytic activity significantly altered the protein composition of phagosomes, particularly reducing the abundance of V‐ATPase proton pump subunits. Furthermore, TPL‐2 stimulated the phosphorylation of DMXL1, a regulator of V‐ATPases, to induce V‐ATPase assembly and phagosome acidification. Consistent with these results, TPL‐2 catalytic activity was required for phagosome acidification and the efficient killing of Staphylococcus aureus and Citrobacter rodentium following phagocytic uptake by macrophages. TPL‐2 therefore controls innate immune responses of macrophages to bacteria via V‐ATPase induction of phagosome maturation.
Collapse
Affiliation(s)
| | - Anetta Härtlova
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Teresa Thurston
- Department of Infectious Diseases, MRC Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK
| | | | | | | | - Julien Peltier
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Tiaan Heunis
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | | | - Matthias Trost
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Steven C Ley
- The Francis Crick Institute, London, UK.,Department of Immunology & Inflammation, Centre for Molecular Immunology & Inflammation, Imperial College London, London, UK
| |
Collapse
|
14
|
Eaton AF, Merkulova M, Brown D. The H +-ATPase (V-ATPase): from proton pump to signaling complex in health and disease. Am J Physiol Cell Physiol 2020; 320:C392-C414. [PMID: 33326313 PMCID: PMC8294626 DOI: 10.1152/ajpcell.00442.2020] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A primary function of the H+-ATPase (or V-ATPase) is to create an electrochemical proton gradient across eukaryotic cell membranes, which energizes fundamental cellular processes. Its activity allows for the acidification of intracellular vesicles and organelles, which is necessary for many essential cell biological events to occur. In addition, many specialized cell types in various organ systems such as the kidney, bone, male reproductive tract, inner ear, olfactory mucosa, and more, use plasma membrane V-ATPases to perform specific activities that depend on extracellular acidification. It is, however, increasingly apparent that V-ATPases are central players in many normal and pathophysiological processes that directly influence human health in many different and sometimes unexpected ways. These include cancer, neurodegenerative diseases, diabetes, and sensory perception, as well as energy and nutrient-sensing functions within cells. This review first covers the well-established role of the V-ATPase as a transmembrane proton pump in the plasma membrane and intracellular vesicles and outlines factors contributing to its physiological regulation in different cell types. This is followed by a discussion of the more recently emerging unconventional roles for the V-ATPase, such as its role as a protein interaction hub involved in cell signaling, and the (patho)physiological implications of these interactions. Finally, the central importance of endosomal acidification and V-ATPase activity on viral infection will be discussed in the context of the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Amity F Eaton
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Maria Merkulova
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dennis Brown
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Lysosome as a Central Hub for Rewiring PH Homeostasis in Tumors. Cancers (Basel) 2020; 12:cancers12092437. [PMID: 32867178 PMCID: PMC7565471 DOI: 10.3390/cancers12092437] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer cells generate large quantities of cytoplasmic protons as byproducts of aberrantly activated aerobic glycolysis and lactate fermentation. To avoid potentially detrimental acidification of the intracellular milieu, cancer cells activate multiple acid-removal pathways that promote cytosolic alkalization and extracellular acidification. Accumulating evidence suggests that in addition to the well-characterized ion pumps and exchangers in the plasma membrane, cancer cell lysosomes are also reprogrammed for this purpose. On the one hand, the increased expression and activity of the vacuolar-type H+-ATPase (V-ATPase) on the lysosomal limiting membrane combined with the larger volume of the lysosomal compartment increases the lysosomal proton storage capacity substantially. On the other hand, enhanced lysosome exocytosis enables the efficient release of lysosomal protons to the extracellular space. Together, these two steps dynamically drive proton flow from the cytosol to extracellular space. In this perspective, we provide mechanistic insight into how lysosomes contribute to the rewiring of pH homeostasis in cancer cells.
Collapse
|
16
|
Collins MP, Forgac M. Regulation and function of V-ATPases in physiology and disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183341. [PMID: 32422136 DOI: 10.1016/j.bbamem.2020.183341] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 02/07/2023]
Abstract
The vacuolar H+-ATPases (V-ATPases) are essential, ATP-dependent proton pumps present in a variety of eukaryotic cellular membranes. Intracellularly, V-ATPase-dependent acidification functions in such processes as membrane traffic, protein degradation, autophagy and the coupled transport of small molecules. V-ATPases at the plasma membrane of certain specialized cells function in such processes as bone resorption, sperm maturation and urinary acidification. V-ATPases also function in disease processes such as pathogen entry and cancer cell invasiveness, while defects in V-ATPase genes are associated with disorders such as osteopetrosis, renal tubular acidosis and neurodegenerative diseases. This review highlights recent advances in our understanding of V-ATPase structure, mechanism, function and regulation, with an emphasis on the signaling pathways controlling V-ATPase assembly in mammalian cells. The role of V-ATPases in cancer and other human pathologies, and the prospects for therapeutic intervention, are also discussed.
Collapse
Affiliation(s)
- Michael P Collins
- Cell, Molecular and Developmental Biology, Tufts University Graduate School of Biomedical Sciences, United States of America
| | - Michael Forgac
- Cell, Molecular and Developmental Biology, Tufts University Graduate School of Biomedical Sciences, United States of America; Dept. of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, United States of America.
| |
Collapse
|
17
|
Biallelic DMXL2 mutations impair autophagy and cause Ohtahara syndrome with progressive course. Brain 2019; 142:3876-3891. [DOI: 10.1093/brain/awz326] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/25/2019] [Accepted: 09/04/2019] [Indexed: 12/27/2022] Open
Abstract
Esposito et al. identify biallelic loss-of-function mutations in DMXL2, encoding a v-ATPase regulatory protein, in three sibling pairs exhibiting Ohtahara syndrome with a progressive course. Patient-derived fibroblasts and Dmxl2-silenced mouse hippocampal neurons show defective lysosomal function and autophagy, resulting in the latter in impaired neuronal development and synapse formation.
Collapse
|
18
|
Gillingham AK, Bertram J, Begum F, Munro S. In vivo identification of GTPase interactors by mitochondrial relocalization and proximity biotinylation. eLife 2019; 8:45916. [PMID: 31294692 PMCID: PMC6639074 DOI: 10.7554/elife.45916] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022] Open
Abstract
The GTPases of the Ras superfamily regulate cell growth, membrane traffic and the cytoskeleton, and a wide range of diseases are caused by mutations in particular members. They function as switchable landmarks with the active GTP-bound form recruiting to the membrane a specific set of effector proteins. The GTPases are precisely controlled by regulators that promote acquisition of GTP (GEFs) or its hydrolysis to GDP (GAPs). We report here MitoID, a method for identifying effectors and regulators by performing in vivo proximity biotinylation with mitochondrially-localized forms of the GTPases. Applying this to 11 human Rab GTPases identified many known effectors and GAPs, as well as putative novel effectors, with examples of the latter validated for Rab2, Rab5, Rab9 and Rab11. MitoID can also efficiently identify effectors and GAPs of Rho and Ras family GTPases such as Cdc42, RhoA, Rheb, and N-Ras, and can identify GEFs by use of GDP-bound forms.
Collapse
Affiliation(s)
| | - Jessie Bertram
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Farida Begum
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Sean Munro
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
19
|
Crummy E, Mani M, Thellman JC, Martin TFJ. The priming factor CAPS1 regulates dense-core vesicle acidification by interacting with rabconnectin3β/WDR7 in neuroendocrine cells. J Biol Chem 2019; 294:9402-9415. [PMID: 31004036 PMCID: PMC6579465 DOI: 10.1074/jbc.ra119.007504] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/21/2019] [Indexed: 12/20/2022] Open
Abstract
Vacuolar-type H+-ATPases (V-ATPases) contribute to pH regulation and play key roles in secretory and endocytic pathways. Dense-core vesicles (DCVs) in neuroendocrine cells are maintained at an acidic pH, which is part of the electrochemical driving force for neurotransmitter loading and is required for hormonal propeptide processing. Genetic loss of CAPS1 (aka calcium-dependent activator protein for secretion, CADPS), a vesicle-bound priming factor required for DCV exocytosis, dissipates the pH gradient across DCV membranes and reduces neurotransmitter loading. However, the basis for CAPS1 binding to DCVs and for its regulation of vesicle pH has not been determined. Here, MS analysis of CAPS1 immunoprecipitates from brain membrane fractions revealed that CAPS1 associates with a rabconnectin3 (Rbcn3) complex comprising Dmx-like 2 (DMXL2) and WD repeat domain 7 (WDR7) proteins. Using immunofluorescence microscopy, we found that Rbcn3α/DMXL2 and Rbcn3β/WDR7 colocalize with CAPS1 on DCVs in human neuroendocrine (BON) cells. The shRNA-mediated knockdown of Rbcn3β/WDR7 redistributed CAPS1 from DCVs to the cytosol, indicating that Rbcn3β/WDR7 is essential for optimal DCV localization of CAPS1. Moreover, cell-free experiments revealed direct binding of CAPS1 to Rbcn3β/WDR7, and cell assays indicated that Rbcn3β/WDR7 recruits soluble CAPS1 to membranes. As anticipated by the reported association of Rbcn3 with V-ATPase, we found that knocking down CAPS1, Rbcn3α, or Rbcn3β in neuroendocrine cells impaired rates of DCV reacidification. These findings reveal a basis for CAPS1 binding to DCVs and for CAPS1 regulation of V-ATPase activity via Rbcn3β/WDR7 interactions.
Collapse
Affiliation(s)
- Ellen Crummy
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| | - Muralidharan Mani
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| | - John C Thellman
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| | - Thomas F J Martin
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
| |
Collapse
|
20
|
Hounjet J, Habets R, Schaaf MB, Hendrickx TC, Barbeau LMO, Yahyanejad S, Rouschop KM, Groot AJ, Vooijs M. The anti-malarial drug chloroquine sensitizes oncogenic NOTCH1 driven human T-ALL to γ-secretase inhibition. Oncogene 2019; 38:5457-5468. [PMID: 30967635 DOI: 10.1038/s41388-019-0802-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 01/30/2019] [Accepted: 03/19/2019] [Indexed: 12/26/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive cancer arising from T-cell progenitors. Although current treatments, including chemotherapy and glucocorticoids, have significantly improved survival, T-ALL remains a fatal disease and new treatment options are needed. Since more than 60% of T-ALL cases bear oncogenic NOTCH1 mutations, small molecule inhibitors of NOTCH1 signalling; γ-secretase inhibitors (GSI), are being actively investigated for the treatment of T-ALL. Unfortunately, GSI have shown limited clinical efficacy and dose-limiting toxicities. We hypothesized that by combining known drugs, blocking NOTCH activity through another mechanism, may synergize with GSI enabling equal efficacy at a lower concentration. Here, we show that the clinically used anti-malarial drug chloroquine (CQ), an inhibitor of lysosomal function and autophagy, decreases T-ALL cell viability and proliferation. This effect of CQ was not observed in GSI-resistant T-ALL cell lines. Mechanistically, CQ impairs the redox balance, induces ds DNA breaks and activates the DNA damage response. CQ also interferes with intracellular trafficking and processing of oncogenic NOTCH1. Interestingly, we show for the first time that the addition of CQ to γ-secretase inhibition has a synergistic therapeutic effect on T-ALL and reduces the concentration of GSI required to obtain a reduction in cell viability and a block of proliferation. Overall, our results suggest that CQ may be a promising repurposed drug in the treatment of T-ALL, as a single treatment or in combination with GSI, increasing the therapeutic ratio.
Collapse
Affiliation(s)
- Judith Hounjet
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands.,MAASTRO Clinic, Maastricht, The Netherlands
| | - Roger Habets
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Marco B Schaaf
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Tessa C Hendrickx
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Lydie M O Barbeau
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Sanaz Yahyanejad
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Kasper M Rouschop
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Arjan J Groot
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands
| | - Marc Vooijs
- Department of Radiotherapy/GROW, School for Developmental Biology & Oncology and Comprehensive Cancer Centre Maastricht MUMC+, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
21
|
Hayek SR, Rane HS, Parra KJ. Reciprocal Regulation of V-ATPase and Glycolytic Pathway Elements in Health and Disease. Front Physiol 2019; 10:127. [PMID: 30828305 PMCID: PMC6384264 DOI: 10.3389/fphys.2019.00127] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/31/2019] [Indexed: 12/24/2022] Open
Abstract
The ability of cells to adapt to fluctuations in glucose availability is crucial for their survival and involves the vacuolar proton-translocating ATPase (V-ATPase), a proton pump found in all eukaryotes. V-ATPase hydrolyzes ATP via its V1 domain and uses the energy released to transport protons across membranes via its Vo domain. This activity is critical for pH homeostasis and generation of a membrane potential that drives cellular metabolism. A number of stimuli have been reported to alter V-ATPase assembly in yeast and higher eukaryotes. Glucose flux is one of the strongest and best-characterized regulators of V-ATPase; this review highlights current models explaining how glycolysis and V-ATPase are coordinated in both the Saccharomyces cerevisiae model fungus and in mammalian systems. Glucose-dependent assembly and trafficking of V-ATPase, V-ATPase-dependent modulations in glycolysis, and the recent discovery that glucose signaling through V-ATPase acts as a molecular switch to dictate anabolic versus catabolic metabolism are discussed. Notably, metabolic plasticity and altered glycolytic flux are critical drivers of numerous human pathologies, and the expression and activity of V-ATPase is often altered in disease states or can be pharmacologically manipulated as treatment. This overview will specifically discuss connections between V-ATPase and glycolysis in cancer.
Collapse
Affiliation(s)
- Summer R Hayek
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Hallie S Rane
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Karlett J Parra
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
22
|
Gobé C, Elzaiat M, Meunier N, André M, Sellem E, Congar P, Jouneau L, Allais-Bonnet A, Naciri I, Passet B, Pailhoux E, Pannetier M. Dual role of DMXL2 in olfactory information transmission and the first wave of spermatogenesis. PLoS Genet 2019; 15:e1007909. [PMID: 30735494 PMCID: PMC6383954 DOI: 10.1371/journal.pgen.1007909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 02/21/2019] [Accepted: 12/19/2018] [Indexed: 12/29/2022] Open
Abstract
Gonad differentiation is a crucial step conditioning the future fertility of individuals and most of the master genes involved in this process have been investigated in detail. However, transcriptomic analyses of developing gonads from different animal models have revealed that hundreds of genes present sexually dimorphic expression patterns. DMXL2 was one of these genes and its function in mammalian gonads was unknown. We therefore investigated the phenotypes of total and gonad-specific Dmxl2 knockout mouse lines. The total loss-of-function of Dmxl2 was lethal in neonates, with death occurring within 12 hours of birth. Dmxl2-knockout neonates were weak and did not feed. They also presented defects of olfactory information transmission and severe hypoglycemia, suggesting that their premature death might be due to global neuronal and/or metabolic deficiencies. Dmxl2 expression in the gonads increased after birth, during follicle formation in females and spermatogenesis in males. DMXL2 was detected in both the supporting and germinal cells of both sexes. As Dmxl2 loss-of-function was lethal, only limited investigations of the gonads of Dmxl2 KO pups were possible. They revealed no major defects at birth. The gonadal function of Dmxl2 was then assessed by conditional deletions of the gene in gonadal supporting cells, germinal cells, or both. Conditional Dmxl2 ablation in the gonads did not impair fertility in males or females. By contrast, male mice with Dmxl2 deletions, either throughout the testes or exclusively in germ cells, presented a subtle testicular phenotype during the first wave of spermatogenesis that was clearly detectable at puberty. Indeed, Dmxl2 loss-of-function throughout the testes or in germ cells only, led to sperm counts more than 60% lower than normal and defective seminiferous tubule architecture. Transcriptomic and immunohistochemichal analyses on these abnormal testes revealed a deregulation of Sertoli cell phagocytic activity related to germ cell apoptosis augmentation. In conclusion, we show that Dmxl2 exerts its principal function in the testes at the onset of puberty, although its absence does not compromise male fertility in mice.
Collapse
Affiliation(s)
- Clara Gobé
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy-en-Josas, France
| | - Maëva Elzaiat
- UMR 7592 Institut Jacques Monod, Université Paris Diderot/CNRS, Paris, France
| | - Nicolas Meunier
- NBO, INRA, Université Paris Saclay, Jouy en Josas, France
- Université de Versailles Saint-Quentin en Yvelines, Versailles, France
| | - Marjolaine André
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy-en-Josas, France
| | - Eli Sellem
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy-en-Josas, France
- R&D Department, ALLICE, Paris, France
| | - Patrice Congar
- NBO, INRA, Université Paris Saclay, Jouy en Josas, France
| | - Luc Jouneau
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy-en-Josas, France
| | - Aurélie Allais-Bonnet
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy-en-Josas, France
- R&D Department, ALLICE, Paris, France
| | - Ikrame Naciri
- Epigenetics and Cell Fate, Université Paris Diderot, Sorbonne Paris Cité, UMR 7216 CNRS, Paris, France
| | - Bruno Passet
- UMR-GABI 1313, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Eric Pailhoux
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy-en-Josas, France
| | - Maëlle Pannetier
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy-en-Josas, France
| |
Collapse
|
23
|
Li A, Robiou-du-Pont S, Anand SS, Morrison KM, McDonald SD, Atkinson SA, Teo KK, Meyre D. Parental and child genetic contributions to obesity traits in early life based on 83 loci validated in adults: the FAMILY study. Pediatr Obes 2018; 13:133-140. [PMID: 28008729 DOI: 10.1111/ijpo.12205] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/16/2016] [Accepted: 11/18/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND The genetic influence on child obesity has not been fully elucidated. OBJECTIVE This study investigated the parental and child contributions of 83 adult body mass index (BMI)-associated single-nucleotide polymorphisms (SNPs) to obesity-related traits in children from birth to 5 years old. METHODS A total of 1402 individuals were genotyped for 83 SNPs. An unweighted genetic risk score (GRS) was generated by the sum of BMI-increasing alleles. Repeated weight and length/height were measured at birth, 1, 2, 3 and 5 years of age, and age-specific and sex-specific weight and BMI Z-scores were computed. RESULTS The GRS was significantly associated with birthweight Z-score (P = 0.03). It was also associated with weight/BMI Z-score gain between birth and 5 years old (P = 0.02 and 6.77 × 10-3 , respectively). In longitudinal analyses, the GRS was associated with weight and BMI Z-score from birth to 5 years (P = 5.91 × 10-3 and 5.08 × 10-3 , respectively). The maternal effects of rs3736485 in DMXL2 on weight and BMI variation from birth to 5 years were significantly greater compared with the paternal effects by Z test (P = 1.53 × 10-6 and 3.75 × 10-5 , respectively). CONCLUSIONS SNPs contributing to adult BMI exert their effect at birth and in early childhood. Parent-of-origin effects may occur in a limited subset of obesity predisposing SNPs.
Collapse
Affiliation(s)
- A Li
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - S Robiou-du-Pont
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - S S Anand
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - K M Morrison
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pediatrics, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - S D McDonald
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada.,Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - S A Atkinson
- Department of Pediatrics, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - K K Teo
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - D Meyre
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
24
|
Pamarthy S, Kulshrestha A, Katara GK, Beaman KD. The curious case of vacuolar ATPase: regulation of signaling pathways. Mol Cancer 2018; 17:41. [PMID: 29448933 PMCID: PMC5815226 DOI: 10.1186/s12943-018-0811-3] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/07/2018] [Indexed: 02/06/2023] Open
Abstract
The Vacuolar ATPase (V-ATPase) is a proton pump responsible for controlling the intracellular and extracellular pH of cells. The structure of V-ATPase has been highly conserved among all eukaryotic cells and is involved in diverse functions across species. V-ATPase is best known for its acidification of endosomes and lysosomes and is also important for luminal acidification of specialized cells. Several reports have suggested the involvement of V-ATPase in maintaining an alkaline intracellular and acidic extracellular pH thereby aiding in proliferation and metastasis of cancer cells respectively. Increased expression of V-ATPase and relocation to the plasma membrane aids in cancer modulates key tumorigenic cell processes like autophagy, Warburg effect, immunomoduation, drug resistance and most importantly cancer cell signaling. In this review, we discuss the direct role of V-ATPase in acidification and indirect regulation of signaling pathways, particularly Notch Signaling.
Collapse
Affiliation(s)
- Sahithi Pamarthy
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA
| | - Arpita Kulshrestha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
25
|
Kilpatrick LE, Phinney KW. Quantification of Total Vitamin-D-Binding Protein and the Glycosylated Isoforms by Liquid Chromatography–Isotope Dilution Mass Spectrometry. J Proteome Res 2017; 16:4185-4195. [DOI: 10.1021/acs.jproteome.7b00560] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Lisa E. Kilpatrick
- National Institute of Standards and Technology, Material Measurement
Laboratory, Biomolecular Measurement Division, 100 Bureau Drive, Stop 8314, Gaithersburg, Maryland 20899, United States
| | - Karen W. Phinney
- National Institute of Standards and Technology, Material Measurement
Laboratory, Biomolecular Measurement Division, 100 Bureau Drive, Stop 8314, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
26
|
Abstract
The vacuolar ATPases (V-ATPases) are a family of proton pumps that couple ATP hydrolysis to proton transport into intracellular compartments and across the plasma membrane. They function in a wide array of normal cellular processes, including membrane traffic, protein processing and degradation, and the coupled transport of small molecules, as well as such physiological processes as urinary acidification and bone resorption. The V-ATPases have also been implicated in a number of disease processes, including viral infection, renal disease, and bone resorption defects. This review is focused on the growing evidence for the important role of V-ATPases in cancer. This includes functions in cellular signaling (particularly Wnt, Notch, and mTOR signaling), cancer cell survival in the highly acidic environment of tumors, aiding the development of drug resistance, as well as crucial roles in tumor cell invasion, migration, and metastasis. Of greatest excitement is evidence that at least some tumors express isoforms of V-ATPase subunits whose disruption is not lethal, leading to the possibility of developing anti-cancer therapeutics that selectively target V-ATPases that function in cancer cells.
Collapse
Affiliation(s)
- Laura Stransky
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Program in Cellular and Molecular Physiology, Program in Biochemistry, and Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts
| | - Kristina Cotter
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Program in Cellular and Molecular Physiology, Program in Biochemistry, and Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts
| | - Michael Forgac
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Program in Cellular and Molecular Physiology, Program in Biochemistry, and Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts
| |
Collapse
|
27
|
Rabconnectin-3α is required for the morphological maturation of GnRH neurons and kisspeptin responsiveness. Sci Rep 2017; 7:42463. [PMID: 28209974 PMCID: PMC5314327 DOI: 10.1038/srep42463] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 01/11/2017] [Indexed: 12/18/2022] Open
Abstract
A few hundred hypothalamic neurons form a complex network that controls reproduction in mammals by secreting gonadotropin-releasing hormone (GnRH). Timely postnatal changes in GnRH secretion are essential for pubertal onset. During the juvenile period, GnRH neurons undergo morphological remodeling, concomitantly achieving an increased responsiveness to kisspeptin, the main secretagogue of GnRH. However, the link between GnRH neuron activity and their morphology remains unknown. Here, we show that brain expression levels of Dmxl2, which encodes the vesicular protein rabconnectin-3α, determine the capacity of GnRH neurons to be activated by kisspeptin and estradiol. We also demonstrate that Dmxl2 expression levels control the pruning of GnRH dendrites, highlighting an unexpected role for a vesicular protein in the maturation of GnRH neuronal network. This effect is mediated by rabconnectin-3α in neurons or glial cells afferent to GnRH neurons. The widespread expression of Dmxl2 in several brain areas raises the intriguing hypothesis that rabconnectin-3α could be involved in the maturation of other neuronal populations.
Collapse
|
28
|
Colacurcio DJ, Nixon RA. Disorders of lysosomal acidification-The emerging role of v-ATPase in aging and neurodegenerative disease. Ageing Res Rev 2016; 32:75-88. [PMID: 27197071 DOI: 10.1016/j.arr.2016.05.004] [Citation(s) in RCA: 352] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/02/2016] [Accepted: 05/13/2016] [Indexed: 12/21/2022]
Abstract
Autophagy and endocytosis deliver unneeded cellular materials to lysosomes for degradation. Beyond processing cellular waste, lysosomes release metabolites and ions that serve signaling and nutrient sensing roles, linking the functions of the lysosome to various pathways for intracellular metabolism and nutrient homeostasis. Each of these lysosomal behaviors is influenced by the intraluminal pH of the lysosome, which is maintained in the low acidic range by a proton pump, the vacuolar ATPase (v-ATPase). New reports implicate altered v-ATPase activity and lysosomal pH dysregulation in cellular aging, longevity, and adult-onset neurodegenerative diseases, including forms of Parkinson disease and Alzheimer disease. Genetic defects of subunits composing the v-ATPase or v-ATPase-related proteins occur in an increasingly recognized group of familial neurodegenerative diseases. Here, we review the expanding roles of the v-ATPase complex as a platform regulating lysosomal hydrolysis and cellular homeostasis. We discuss the unique vulnerability of neurons to persistent low level lysosomal dysfunction and review recent clinical and experimental studies that link dysfunction of the v-ATPase complex to neurodegenerative diseases across the age spectrum.
Collapse
|
29
|
Zhou W, He Q, Zhang C, He X, Cui Z, Liu F, Li W. BLOS2 negatively regulates Notch signaling during neural and hematopoietic stem and progenitor cell development. eLife 2016; 5. [PMID: 27719760 PMCID: PMC5094856 DOI: 10.7554/elife.18108] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/04/2016] [Indexed: 12/28/2022] Open
Abstract
Notch signaling plays a crucial role in controling the proliferation and differentiation of stem and progenitor cells during embryogenesis or organogenesis, but its regulation is incompletely understood. BLOS2, encoded by the Bloc1s2 gene, is a shared subunit of two lysosomal trafficking complexes, biogenesis of lysosome-related organelles complex-1 (BLOC-1) and BLOC-1-related complex (BORC). Bloc1s2-/- mice were embryonic lethal and exhibited defects in cortical development and hematopoiesis. Loss of BLOS2 resulted in elevated Notch signaling, which consequently increased the proliferation of neural progenitor cells and inhibited neuronal differentiation in cortices. Likewise, ablation of bloc1s2 in zebrafish or mice led to increased hematopoietic stem and progenitor cell production in the aorta-gonad-mesonephros region. BLOS2 physically interacted with Notch1 in endo-lysosomal trafficking of Notch1. Our findings suggest that BLOS2 is a novel negative player in regulating Notch signaling through lysosomal trafficking to control multiple stem and progenitor cell homeostasis in vertebrates.
Collapse
Affiliation(s)
- Wenwen Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qiuping He
- University of Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chunxia Zhang
- University of Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xin He
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Zongbin Cui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Beijing, China
| | - Feng Liu
- University of Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,Center for Medical Genetics, Beijing Children's Hospital, Capital Medical University, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China.,MOE Key Laboratory of Major Diseases in Children, Beijing, China.,Beijing Pediatric Research Institute, Beijing, China
| |
Collapse
|
30
|
DMXL2 drives epithelial to mesenchymal transition in hormonal therapy resistant breast cancer through Notch hyper-activation. Oncotarget 2016; 6:22467-79. [PMID: 26093085 PMCID: PMC4673176 DOI: 10.18632/oncotarget.4164] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/22/2015] [Indexed: 12/21/2022] Open
Abstract
The acquisition of endocrine therapy resistance in estrogen receptor α (ERα) breast cancer patients represents a major clinical problem. Notch signalling has been extensively linked to breast cancer especially in patients who fail to respond to endocrine therapy. Following activation, Notch intracellular domain is released and enters the nucleus where activates transcription of target genes. The numerous steps that cascade after activation of the receptor complicate using Notch as biomarker. Hence, this warrants the development of reliable indicators of Notch activity. DMXL2 is a novel regulator of Notch signalling not yet investigated in breast cancer. Here, we demonstrate that DMXL2 is overexpressed in a subset of endocrine therapy resistant breast cancer cell lines where it promotes epithelial to mesenchymal transition through hyper-activation of Notch signalling via V-ATPase dependent acidification. Following DMXL2 depletion or treatment with Bafilomycin A1, both EMT targets and Notch signalling pathway significantly decrease. We show for the first time that DMXL2 protein levels are significantly increased in ERα positive breast cancer patients that progress after endocrine therapy. Finally, we demonstrate that DMXL2 is a transmembrane protein with a potential extra-cellular domain. These findings identify DMXL2 as a novel, functional biomarker for ERα positive breast cancer.
Collapse
|
31
|
Pamarthy S, Jaiswal MK, Kulshreshtha A, Katara GK, Gilman-Sachs A, Beaman KD. The Vacuolar ATPase a2-subunit regulates Notch signaling in triple-negative breast cancer cells. Oncotarget 2016; 6:34206-20. [PMID: 26418877 PMCID: PMC4741446 DOI: 10.18632/oncotarget.5275] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/07/2015] [Indexed: 12/24/2022] Open
Abstract
Triple Negative Breast Cancer (TNBC) is a subtype of breast cancer with poor prognosis for which no targeted therapies are currently available. Notch signaling has been implicated in breast cancer but the factors that control Notch in TNBC are unknown. Because the Vacuolar ATPase has been shown to be important in breast cancer invasiveness, we investigated the role of a2-subunit isoform of Vacuolar ATPase (a2V) in regulating Notch signaling in TNBC. Confocal microscopy revealed that among all the ‘a’ subunit isoforms, a2V was uniquely expressed on the plasma membrane of breast cancer cells. Both a2V and NOTCH1 were elevated in TNBC tumors tissues and cell lines. a2V knockdown by siRNA as well as V-ATPase inhibition by Bafilomycin A1 (Baf A1) in TNBC cell lines enhanced Notch signaling by increasing the expression of Notch1 intracellular Domain (N1ICD). V-ATPase inhibition blocked NICD degradation by disrupting autophagy and lysosomal acidification as demonstrated by accumulation of LC3B and diminished expression of LAMP1 respectively. Importantly, treatment with Baf A1 or anti-a2V, a novel-neutralizing antibody against a2V hindered cell migration of TNBC cells. Our findings indicate that a2V regulates Notch signaling through its role in endolysosomal acidification and emerges as a potential target for TNBC.
Collapse
Affiliation(s)
- Sahithi Pamarthy
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Mukesh K Jaiswal
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Arpita Kulshreshtha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Alice Gilman-Sachs
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| |
Collapse
|
32
|
Cotter K, Stransky L, McGuire C, Forgac M. Recent Insights into the Structure, Regulation, and Function of the V-ATPases. Trends Biochem Sci 2016; 40:611-622. [PMID: 26410601 DOI: 10.1016/j.tibs.2015.08.005] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 10/23/2022]
Abstract
The vacuolar (H(+))-ATPases (V-ATPases) are ATP-dependent proton pumps that acidify intracellular compartments and are also present at the plasma membrane. They function in such processes as membrane traffic, protein degradation, virus and toxin entry, bone resorption, pH homeostasis, and tumor cell invasion. V-ATPases are large multisubunit complexes, composed of an ATP-hydrolytic domain (V1) and a proton translocation domain (V0), and operate by a rotary mechanism. This review focuses on recent insights into their structure and mechanism, the mechanisms that regulate V-ATPase activity (particularly regulated assembly and trafficking), and the role of V-ATPases in processes such as cell signaling and cancer. These developments have highlighted the potential of V-ATPases as a therapeutic target in a variety of human diseases.
Collapse
Affiliation(s)
- Kristina Cotter
- Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Laura Stransky
- Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Christina McGuire
- Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Michael Forgac
- Program in Cellular and Molecular Physiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA; Program in Biochemistry, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA; Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA.
| |
Collapse
|
33
|
Tavares B, Pintado P, Lopes SS. The role of Rabconnectin3a in cilia length regulation. Cilia 2015. [PMCID: PMC4518627 DOI: 10.1186/2046-2530-4-s1-p70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
34
|
Merkulova M, Păunescu TG, Azroyan A, Marshansky V, Breton S, Brown D. Mapping the H(+) (V)-ATPase interactome: identification of proteins involved in trafficking, folding, assembly and phosphorylation. Sci Rep 2015; 5:14827. [PMID: 26442671 PMCID: PMC4595830 DOI: 10.1038/srep14827] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 09/02/2015] [Indexed: 12/04/2022] Open
Abstract
V-ATPases (H+ ATPases) are multisubunit, ATP-dependent proton pumps that regulate pH homeostasis in virtually all eukaryotes. They are involved in key cell biological processes including vesicle trafficking, endosomal pH sensing, membrane fusion and intracellular signaling. They also have critical systemic roles in renal acid excretion and blood pH balance, male fertility, bone remodeling, synaptic transmission, olfaction and hearing. Furthermore, V-ATPase dysfunction either results in or aggravates various other diseases, but little is known about the complex protein interactions that regulate these varied V-ATPase functions. Therefore, we performed a proteomic analysis to identify V-ATPase associated proteins and construct a V-ATPase interactome. Our analysis using kidney tissue revealed V-ATPase-associated protein clusters involved in protein quality control, complex assembly and intracellular trafficking. ARHGEF7, DMXL1, EZR, NCOA7, OXR1, RPS6KA3, SNX27 and 9 subunits of the chaperonin containing TCP1 complex (CCT) were found to interact with V-ATPase for the first time in this study. Knockdown of two interacting proteins, DMXL1 and WDR7, inhibited V-ATPase-mediated intracellular vesicle acidification in a kidney cell line, providing validation for the utility of our interactome as a screen for functionally important novel V-ATPase-regulating proteins. Our data, therefore, provide new insights and directions for the analysis of V-ATPase cell biology and (patho)physiology.
Collapse
Affiliation(s)
- Maria Merkulova
- MGH Center for Systems Biology, Program in Membrane Biology &Division of Nephrology, Richard B. Simches Research Center, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Teodor G Păunescu
- MGH Center for Systems Biology, Program in Membrane Biology &Division of Nephrology, Richard B. Simches Research Center, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Anie Azroyan
- MGH Center for Systems Biology, Program in Membrane Biology &Division of Nephrology, Richard B. Simches Research Center, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Vladimir Marshansky
- MGH Center for Systems Biology, Program in Membrane Biology &Division of Nephrology, Richard B. Simches Research Center, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Sylvie Breton
- MGH Center for Systems Biology, Program in Membrane Biology &Division of Nephrology, Richard B. Simches Research Center, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Dennis Brown
- MGH Center for Systems Biology, Program in Membrane Biology &Division of Nephrology, Richard B. Simches Research Center, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
35
|
Nikolova LS, Metzstein MM. Intracellular lumen formation in Drosophila proceeds via a novel subcellular compartment. Development 2015; 142:3964-73. [PMID: 26428009 DOI: 10.1242/dev.127902] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/17/2015] [Indexed: 12/28/2022]
Abstract
Cellular tubes have diverse morphologies, including multicellular, unicellular and subcellular architectures. Subcellular tubes are found prominently within the vertebrate vasculature, the insect breathing system and the nematode excretory apparatus, but how such tubes form is poorly understood. To characterize the cellular mechanisms of subcellular tube formation, we have refined methods of high pressure freezing/freeze substitution to prepare Drosophila larvae for transmission electron microscopic (TEM) analysis. Using our methods, we have found that subcellular tube formation may proceed through a previously undescribed multimembrane intermediate composed of vesicles bound within a novel subcellular compartment. We have also developed correlative light/TEM procedures to identify labeled cells in TEM-fixed larval samples. Using this technique, we have found that Vacuolar ATPase (V-ATPase) and the V-ATPase regulator Rabconnectin-3 are required for subcellular tube formation, probably in a step resolving the intermediate compartment into a mature lumen. In general, our ultrastructural analysis methods could be useful for a wide range of cellular investigations in Drosophila larvae.
Collapse
Affiliation(s)
- Linda S Nikolova
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Mark M Metzstein
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
36
|
Smardon AM, Nasab ND, Tarsio M, Diakov TT, Kane PM. Molecular Interactions and Cellular Itinerary of the Yeast RAVE (Regulator of the H+-ATPase of Vacuolar and Endosomal Membranes) Complex. J Biol Chem 2015; 290:27511-23. [PMID: 26405040 DOI: 10.1074/jbc.m115.667634] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Indexed: 12/11/2022] Open
Abstract
The RAVE complex (regulator of the H(+)-ATPase of vacuolar and endosomal membranes) is required for biosynthetic assembly and glucose-stimulated reassembly of the yeast vacuolar H(+)-ATPase (V-ATPase). Yeast RAVE contains three subunits: Rav1, Rav2, and Skp1. Rav1 is the largest subunit, and it binds Rav2 and Skp1 of RAVE; the E, G, and C subunits of the V-ATPase peripheral V1 sector; and Vph1 of the membrane Vo sector. We identified Rav1 regions required for interaction with its binding partners through deletion analysis, co-immunoprecipitation, two-hybrid assay, and pulldown assays with expressed proteins. We find that Skp1 binding requires sequences near the C terminus of Rav1, V1 subunits E and C bind to a conserved region in the C-terminal half of Rav1, and the cytosolic domain of Vph1 binds near the junction of the Rav1 N- and C-terminal halves. In contrast, Rav2 binds to the N-terminal domain of Rav1, which can be modeled as a double β-propeller. Only the V1 C subunit binds to both Rav1 and Rav2. Using GFP-tagged RAVE subunits in vivo, we demonstrate glucose-dependent association of RAVE with the vacuolar membrane, consistent with its role in glucose-dependent V-ATPase assembly. It is known that V1 subunit C localizes to the V1-Vo interface in assembled V-ATPase complexes and is important in regulated disassembly of V-ATPases. We propose that RAVE cycles between cytosol and vacuolar membrane in a glucose-dependent manner, positioning V1 and V0 subcomplexes and orienting the V1 C subunit to promote assembly.
Collapse
Affiliation(s)
- Anne M Smardon
- From the Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| | - Negin Dehdar Nasab
- From the Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| | - Maureen Tarsio
- From the Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| | - Theodore T Diakov
- From the Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| | - Patricia M Kane
- From the Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York 13210
| |
Collapse
|
37
|
Agrawal V, Jaiswal MK, Pamarthy S, Katara GK, Kulshrestha A, Gilman-Sachs A, Hirsch E, Beaman KD. Role of Notch signaling during lipopolysaccharide-induced preterm labor. J Leukoc Biol 2015; 100:261-74. [PMID: 26373439 DOI: 10.1189/jlb.3hi0515-200rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/15/2015] [Indexed: 12/31/2022] Open
Abstract
Notch signaling pathways exert effects throughout pregnancy and are activated in response to TLR ligands. To investigate the role of Notch signaling in preterm labor, Notch receptors (Notch1-4), its ligand Delta-like protein-1, transcriptional repressor hairy and enhancer of split-1, and Notch deregulator Numb were assessed. Preterm labor was initiated on gestation d 14.5 by 1 of 2 methods: 1) inflammation-induced preterm labor: intrauterine injection of LPS (a TLR4 agonist) and 2) hormonally induced preterm labor: subcutaneous injection of mifepristone. Delta-like protein-1, Notch1, and hairy and enhancer of split-1 were elevated significantly, and Numb was decreased in the uterus and placenta of inflammation-induced preterm labor mice but remained unchanged in hormonally induced preterm labor compared with their respective controls. F4/80(+) macrophage polarization was skewed in the uterus of inflammation-induced preterm labor toward M1-positive (CD11c(+)) and double-positive [CD11c(+) (M1) and CD206(+) (M2)] cells. This process is dependent on activation of Notch signaling, as shown by suppression of M1 and M2 macrophage-associated cytokines in decidual macrophages in response to γ-secretase inhibitor (an inhibitor of Notch receptor processing) treatment ex vivo. γ-Secretase inhibitor treatment also diminished the LPS-induced secretion of proinflammatory cytokines and chemokines in decidual and placental cells cultured ex vivo. Furthermore, treatment with recombinant Delta-like protein-1 ligand enhanced the LPS-induced proinflammatory response. Notch ligands (Jagged 1 and 2 and Delta-like protein-4) and vascular endothelial growth factor and its receptor involved in angiogenesis were reduced significantly in the uterus and placenta during inflammation-induced preterm labor. These results suggest that up-regulation of Notch-related inflammation and down-regulation of angiogenesis factors may be associated with inflammation-induced preterm labor but not with hormonally induced preterm labor.
Collapse
Affiliation(s)
- Varkha Agrawal
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA;
| | - Mukesh K Jaiswal
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA; and
| | - Sahithi Pamarthy
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA; and
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA; and
| | - Arpita Kulshrestha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA; and
| | - Alice Gilman-Sachs
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA; and
| | - Emmet Hirsch
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA; Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA; and
| |
Collapse
|
38
|
Feng Q, Gao N. Keeping Wnt signalosome in check by vesicular traffic. J Cell Physiol 2015; 230:1170-80. [PMID: 25336320 DOI: 10.1002/jcp.24853] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 10/17/2014] [Indexed: 01/01/2023]
Abstract
Wg/Wnts are paracrine and autocrine ligands that activate distinct signaling pathways while being internalized through surface receptors. Converging and contrasting views are shaping our understanding of whether, where, and how endocytosis may modulate Wnt signaling. We gather considerable amount of evidences to elaborate the point that signal-receiving cells utilize distinct, flexible, and sophisticated vesicular trafficking mechanisms to keep Wnt signaling activity in check. Same molecules in a highly context-dependent fashion serve as regulatory hub for various signaling purposes: amplification, maintenance, inhibition, and termination. Updates are provided for the regulatory mechanisms related to the three critical cell surface complexes, Wnt-Fzd-LRP6, Dkk1-Kremen-LRP6, and R-spondin-LGR5-RNF43, which potently influence Wnt signaling. We pay particular attentions to how cells achieve sustained and delicate control of Wnt signaling strength by employing comprehensive aspects of vesicular trafficking.
Collapse
Affiliation(s)
- Qiang Feng
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | | |
Collapse
|
39
|
Wang SK, Hu Y, Yang J, Smith CE, Nunez SM, Richardson AS, Pal S, Samann AC, Hu JCC, Simmer JP. Critical roles for WDR72 in calcium transport and matrix protein removal during enamel maturation. Mol Genet Genomic Med 2015; 3:302-19. [PMID: 26247047 PMCID: PMC4521966 DOI: 10.1002/mgg3.143] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/02/2015] [Accepted: 03/02/2015] [Indexed: 12/19/2022] Open
Abstract
Defects in WDR72 (WD repeat-containing protein 72) cause autosomal recessive hypomaturation amelogenesis imperfecta. We generated and characterized Wdr72-knockout/lacZ-knockin mice to investigate the role of WDR72 in enamel formation. In all analyses, enamel formed by Wdr72 heterozygous mice was indistinguishable from wild-type enamel. Without WDR72, enamel mineral density increased early during the maturation stage but soon arrested. The null enamel layer was only a tenth as hard as wild-type enamel and underwent rapid attrition following eruption. Despite the failure to further mineralize enamel deposited during the secretory stage, ectopic mineral formed on the enamel surface and penetrated into the overlying soft tissue. While the proteins in the enamel matrix were successfully degraded, the digestion products remained inside the enamel. Interactome analysis of WDR72 protein revealed potential interactions with clathrin-associated proteins and involvement in ameloblastic endocytosis. The maturation stage mandibular incisor enamel did not stain with methyl red, indicating that the enamel did not acidify beneath ruffle-ended ameloblasts. Attachment of maturation ameloblasts to the enamel layer was weakened, and SLC24A4, a critical ameloblast calcium transporter, did not localize appropriately along the ameloblast distal membrane. Fewer blood vessels were observed in the papillary layer supporting ameloblasts. Specific WDR72 expression by maturation stage ameloblasts explained the observation that enamel thickness and rod decussation (established during the secretory stage) are normal in the Wdr72 null mice. We conclude that WDR72 serves critical functions specifically during the maturation stage of amelogenesis and is required for both protein removal and enamel mineralization.
Collapse
Affiliation(s)
- Shih-Kai Wang
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry 1210 Eisenhower Pl., Ann Arbor, Michigan, 48108
| | - Yuanyuan Hu
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry 1210 Eisenhower Pl., Ann Arbor, Michigan, 48108
| | - Jie Yang
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry 1210 Eisenhower Pl., Ann Arbor, Michigan, 48108 ; Department of Pediatric Dentistry, School and Hospital of Stomatology, Peking University 22 South Avenue Zhongguancun, Haidian District, Beijing, 100081, China
| | - Charles E Smith
- Facility for Electron Microscopy Research, Department of Anatomy and Cell Biology and Faculty of Dentistry, McGill University 3640 University Street, Montreal, Quebec, Canada, H3A 2B2
| | - Stephanie M Nunez
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry 1210 Eisenhower Pl., Ann Arbor, Michigan, 48108
| | - Amelia S Richardson
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry 1210 Eisenhower Pl., Ann Arbor, Michigan, 48108
| | - Soumya Pal
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry 1210 Eisenhower Pl., Ann Arbor, Michigan, 48108
| | - Andrew C Samann
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry 1210 Eisenhower Pl., Ann Arbor, Michigan, 48108
| | - Jan C-C Hu
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry 1210 Eisenhower Pl., Ann Arbor, Michigan, 48108
| | - James P Simmer
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry 1210 Eisenhower Pl., Ann Arbor, Michigan, 48108
| |
Collapse
|
40
|
Nabatov AA. The vesicle-associated function of NOD2 as a link between Crohn's disease and mycobacterial infection. Gut Pathog 2015; 7:1. [PMID: 25653718 PMCID: PMC4316803 DOI: 10.1186/s13099-015-0049-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/03/2015] [Indexed: 12/18/2022] Open
Abstract
Although Crohn’s disease (CD) etiology remains unclear, a growing body of evidence suggests that CD may include an infectious component, with Mycobacterium avium subsp. paratuberculosis (MAP) being the most likely candidate for this role. However, the molecular mechanism of the MAP involvement in CD pathogenesis remains unclear. The polymorphism of the NOD2 gene, coding for an intracellular pattern recognition receptor, is a factor of predisposition to mycobacterial infections and CD. Recent findings on NOD2 interactions and functions provide the missing pieces in the puzzle of a NOD2-mediated mechanism common for mycobacterial infections and CD. Implications of these new findings for the development of a better understanding and treatments of CD and mycobacterial infections are discussed.
Collapse
Affiliation(s)
- Alexey A Nabatov
- Maastricht Radiation Oncology, MAASTRO/GROW Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands ; Science Center, Volga Region State Academy of Physical Culture, Sport and Tourism, 33, Universiade Village, Kazan, 420138 Russia
| |
Collapse
|
41
|
Tata B, Huijbregts L, Jacquier S, Csaba Z, Genin E, Meyer V, Leka S, Dupont J, Charles P, Chevenne D, Carel JC, Léger J, de Roux N. Haploinsufficiency of Dmxl2, encoding a synaptic protein, causes infertility associated with a loss of GnRH neurons in mouse. PLoS Biol 2014; 12:e1001952. [PMID: 25248098 PMCID: PMC4172557 DOI: 10.1371/journal.pbio.1001952] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/12/2014] [Indexed: 12/20/2022] Open
Abstract
Characterization of the genetic defects causing gonadotropic deficiency has made a major contribution to elucidation of the fundamental role of Kisspeptins and Neurokinin B in puberty onset and reproduction. The absence of puberty may also reveal neurodevelopmental disorders caused by molecular defects in various cellular pathways. Investigations of these neurodevelopmental disorders may provide information about the neuronal processes controlling puberty onset and reproductive capacity. We describe here a new syndrome observed in three brothers, which involves gonadotropic axis deficiency, central hypothyroidism, peripheral demyelinating sensorimotor polyneuropathy, mental retardation, and profound hypoglycemia, progressing to nonautoimmune insulin-dependent diabetes mellitus. High-throughput sequencing revealed a homozygous in-frame deletion of 15 nucleotides in DMXL2 in all three affected patients. This homozygous deletion was associated with lower DMXL2 mRNA levels in the blood lymphocytes of the patients. DMXL2 encodes the synaptic protein rabconnectin-3α, which has been identified as a putative scaffold protein for Rab3-GAP and Rab3-GEP, two regulators of the GTPase Rab3a. We found that rabconnectin-3α was expressed in exocytosis vesicles in gonadotropin-releasing hormone (GnRH) axonal extremities in the median eminence of the hypothalamus. It was also specifically expressed in cells expressing luteinizing hormone (LH) and follicle-stimulating hormone (FSH) within the pituitary. The conditional heterozygous deletion of Dmxl2 from mouse neurons delayed puberty and resulted in very low fertility. This reproductive phenotype was associated with a lower number of GnRH neurons in the hypothalamus of adult mice. Finally, Dmxl2 knockdown in an insulin-secreting cell line showed that rabconnectin-3α controlled the constitutive and glucose-induced secretion of insulin. In conclusion, this study shows that low levels of DMXL2 expression cause a complex neurological phenotype, with abnormal glucose metabolism and gonadotropic axis deficiency due to a loss of GnRH neurons. Our findings identify rabconectin-3α as a key controller of neuronal and endocrine homeostatic processes.
Collapse
Affiliation(s)
- Brooke Tata
- Inserm, U1141, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France
| | - Lukas Huijbregts
- Inserm, U1141, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France
| | | | | | | | | | | | - Joelle Dupont
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Perrine Charles
- Genetics Department and Inserm US975, Université Pierre et Marie Curie, Hôpital la Pitié-Salpêtrière, Paris, France
| | - Didier Chevenne
- AP-HP, Laboratoire de Biochimie, Hôpital Robert Debré, Paris, France
| | - Jean-Claude Carel
- Inserm, U1141, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France
- AP-HP, Service d'Endocrinologie Diabétologie Pédiatrique et Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Hôpital Robert Debré, Paris, France
| | - Juliane Léger
- Inserm, U1141, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France
- AP-HP, Service d'Endocrinologie Diabétologie Pédiatrique et Centre de Référence des Maladies Endocriniennes Rares de la Croissance, Hôpital Robert Debré, Paris, France
| | - Nicolas de Roux
- Inserm, U1141, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Hôpital Robert Debré, Paris, France
- AP-HP, Laboratoire de Biochimie, Hôpital Robert Debré, Paris, France
- * E-mail:
| |
Collapse
|
42
|
Katsura KA, Horst JA, Chandra D, Le TQ, Nakano Y, Zhang Y, Horst OV, Zhu L, Le MH, DenBesten PK. WDR72 models of structure and function: a stage-specific regulator of enamel mineralization. Matrix Biol 2014; 38:48-58. [PMID: 25008349 PMCID: PMC4185229 DOI: 10.1016/j.matbio.2014.06.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 06/21/2014] [Accepted: 06/26/2014] [Indexed: 12/18/2022]
Abstract
Amelogenesis Imperfecta (AI) is a clinical diagnosis that encompasses a group of genetic mutations, each affecting processes involved in tooth enamel formation and thus, result in various enamel defects. The hypomaturation enamel phenotype has been described for mutations involved in the later stage of enamel formation, including Klk4, Mmp20, C4orf26, and Wdr72. Using a candidate gene approach we discovered a novel Wdr72 human mutation in association with AI to be a 5-base pair deletion (c.806_810delGGCAG; p.G255VfsX294). To gain insight into the function of WDR72, we used computer modeling of the full-length human WDR72 protein structure and found that the predicted N-terminal sequence forms two beta-propeller folds with an alpha-solenoid tail at the C-terminus. This domain iteration is characteristic of vesicle coat proteins, such as beta'-COP, suggesting a role for WDR72 in the formation of membrane deformation complexes to regulate intracellular trafficking. Our Wdr72 knockout mouse model (Wdr72(-/-)), containing a LacZ reporter knock-in, exhibited hypomineralized enamel similar to the AI phenotype observed in humans with Wdr72 mutations. MicroCT scans of Wdr72(-/-) mandibles affirmed the hypomineralized enamel phenotype occurring at the onset of the maturation stage. H&E staining revealed a shortened height phenotype in the Wdr72(-/-) ameloblasts with retained proteins in the enamel matrix during maturation stage. H(+)/Cl(-) exchange transporter 5 (CLC5), an early endosome acidifier, was co-localized with WDR72 in maturation-stage ameloblasts and decreased in Wdr72(-/-) maturation-stage ameloblasts. There were no obvious differences in RAB4A and LAMP1 immunostaining of Wdr72(-/-) mice as compared to wildtype controls. Moreover, Wdr72(-/-) ameloblasts had reduced amelogenin immunoreactivity, suggesting defects in amelogenin fragment resorption from the matrix. These data demonstrate that WDR72 has a major role in enamel mineralization, most notably during the maturation stage, and suggest a function involving endocytic vesicle trafficking, possibly in the removal of amelogenin proteins.
Collapse
Affiliation(s)
- K A Katsura
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA 94143-0422, USA
| | - J A Horst
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA 94143-0422, USA
| | - D Chandra
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA 94143-0422, USA
| | - T Q Le
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA 94143-0422, USA
| | - Y Nakano
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA 94143-0422, USA
| | - Y Zhang
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA 94143-0422, USA
| | - O V Horst
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA 94143-0422, USA
| | - L Zhu
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA 94143-0422, USA
| | - M H Le
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA 94143-0422, USA
| | - P K DenBesten
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, 513 Parnassus Ave., San Francisco, CA 94143-0422, USA
| |
Collapse
|
43
|
Tuttle AM, Hoffman TL, Schilling TF. Rabconnectin-3a regulates vesicle endocytosis and canonical Wnt signaling in zebrafish neural crest migration. PLoS Biol 2014; 12:e1001852. [PMID: 24802872 PMCID: PMC4011682 DOI: 10.1371/journal.pbio.1001852] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 03/28/2014] [Indexed: 12/15/2022] Open
Abstract
Cell migration requires dynamic regulation of cell-cell signaling and cell adhesion. Both of these processes involve endocytosis, lysosomal degradation, and recycling of ligand-receptor complexes and cell adhesion molecules from the plasma membrane. Neural crest (NC) cells in vertebrates are highly migratory cells, which undergo an epithelial-mesenchymal transition (EMT) to leave the neural epithelium and migrate throughout the body to give rise to many different derivatives. Here we show that the v-ATPase interacting protein, Rabconnectin-3a (Rbc3a), controls intracellular trafficking events and Wnt signaling during NC migration. In zebrafish embryos deficient in Rbc3a, or its associated v-ATPase subunit Atp6v0a1, many NC cells fail to migrate and misregulate expression of cadherins. Surprisingly, endosomes in Rbc3a- and Atp6v0a1-deficient NC cells remain immature but still acidify. Rbc3a loss-of-function initially downregulates several canonical Wnt targets involved in EMT, but later Frizzled-7 accumulates at NC cell membranes, and nuclear B-catenin levels increase. Presumably due to this later Wnt signaling increase, Rbc3a-deficient NC cells that fail to migrate become pigment progenitors. We propose that Rbc3a and Atp6v0a1 promote endosomal maturation to coordinate Wnt signaling and intracellular trafficking of Wnt receptors and cadherins required for NC migration and cell fate determination. Our results suggest that different v-ATPases and associated proteins may play cell-type-specific functions in intracellular trafficking in many contexts.
Collapse
Affiliation(s)
- Adam M. Tuttle
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America
| | - Trevor L. Hoffman
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
44
|
Saccharomyces cerevisiae vacuolar H+-ATPase regulation by disassembly and reassembly: one structure and multiple signals. EUKARYOTIC CELL 2014; 13:706-14. [PMID: 24706019 DOI: 10.1128/ec.00050-14] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Vacuolar H(+)-ATPases (V-ATPases) are highly conserved ATP-driven proton pumps responsible for acidification of intracellular compartments. V-ATPase proton transport energizes secondary transport systems and is essential for lysosomal/vacuolar and endosomal functions. These dynamic molecular motors are composed of multiple subunits regulated in part by reversible disassembly, which reversibly inactivates them. Reversible disassembly is intertwined with glycolysis, the RAS/cyclic AMP (cAMP)/protein kinase A (PKA) pathway, and phosphoinositides, but the mechanisms involved are elusive. The atomic- and pseudo-atomic-resolution structures of the V-ATPases are shedding light on the molecular dynamics that regulate V-ATPase assembly. Although all eukaryotic V-ATPases may be built with an inherent capacity to reversibly disassemble, not all do so. V-ATPase subunit isoforms and their interactions with membrane lipids and a V-ATPase-exclusive chaperone influence V-ATPase assembly. This minireview reports on the mechanisms governing reversible disassembly in the yeast Saccharomyces cerevisiae, keeping in perspective our present understanding of the V-ATPase architecture and its alignment with the cellular processes and signals involved.
Collapse
|
45
|
Marshansky V, Rubinstein JL, Grüber G. Eukaryotic V-ATPase: novel structural findings and functional insights. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2014; 1837:857-79. [PMID: 24508215 DOI: 10.1016/j.bbabio.2014.01.018] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 12/25/2013] [Accepted: 01/27/2014] [Indexed: 02/06/2023]
Abstract
The eukaryotic V-type adenosine triphosphatase (V-ATPase) is a multi-subunit membrane protein complex that is evolutionarily related to F-type adenosine triphosphate (ATP) synthases and A-ATP synthases. These ATPases/ATP synthases are functionally conserved and operate as rotary proton-pumping nano-motors, invented by Nature billions of years ago. In the first part of this review we will focus on recent structural findings of eukaryotic V-ATPases and discuss the role of different subunits in the function of the V-ATPase holocomplex. Despite structural and functional similarities between rotary ATPases, the eukaryotic V-ATPases are the most complex enzymes that have acquired some unconventional cellular functions during evolution. In particular, the novel roles of V-ATPases in the regulation of cellular receptors and their trafficking via endocytotic and exocytotic pathways were recently uncovered. In the second part of this review we will discuss these unique roles of V-ATPases in modulation of function of cellular receptors, involved in the development and progression of diseases such as cancer and diabetes as well as neurodegenerative and kidney disorders. Moreover, it was recently revealed that the V-ATPase itself functions as an evolutionarily conserved pH sensor and receptor for cytohesin-2/Arf-family GTP-binding proteins. Thus, in the third part of the review we will evaluate the structural basis for and functional insights into this novel concept, followed by the analysis of the potentially essential role of V-ATPase in the regulation of this signaling pathway in health and disease. Finally, future prospects for structural and functional studies of the eukaryotic V-ATPase will be discussed.
Collapse
Affiliation(s)
- Vladimir Marshansky
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Simches Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Kadmon Pharmaceuticals Corporation, Alexandria Center for Life Science, 450 East 29th Street, New York, NY 10016, USA.
| | - John L Rubinstein
- Molecular Structure and Function Program, The Hospital for Sick Children Research Institute, University of Toronto, Toronto, ON M5G 1X8, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5G 1X8, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Gerhard Grüber
- Nanyang Technological University, Division of Structural Biology and Biochemistry, School of Biological Sciences, Singapore 637551, Republic of Singapore; Bioinformatics Institute, A(⁎)STAR, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| |
Collapse
|
46
|
Abstract
Specialized cells in the body express high levels of V-ATPase in their plasma membrane and respond to hormonal and nonhormonal cues to regulate extracellular acidification. Mutations in or loss of some V-ATPase subunits cause several disorders, including renal distal tubular acidosis and male infertility. This review focuses on the regulation of V-ATPase-dependent luminal acidification in renal intercalated cells and epididymal clear cells, which are key players in these physiological processes.
Collapse
Affiliation(s)
- Sylvie Breton
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, Massachusetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
47
|
Smardon AM, Diab HI, Tarsio M, Diakov TT, Nasab ND, West RW, Kane PM. The RAVE complex is an isoform-specific V-ATPase assembly factor in yeast. Mol Biol Cell 2013; 25:356-67. [PMID: 24307682 PMCID: PMC3907276 DOI: 10.1091/mbc.e13-05-0231] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Vacuolar H+-ATPases (V-ATPases) acidify multiple organelles, and subunit isoforms help impart organelle-specific regulation of acidification. The regulator of ATPase of vacuoles and endosomes (RAVE) complex regulates organelle acidification by promoting V-ATPase assembly. This work demonstrates that RAVE is the first identified isoform-specific V-ATPase assembly factor required for control of vacuolar acidification. The regulator of ATPase of vacuoles and endosomes (RAVE) complex is implicated in vacuolar H+-translocating ATPase (V-ATPase) assembly and activity. In yeast, rav1∆ mutants exhibit a Vma− growth phenotype characteristic of loss of V-ATPase activity only at high temperature. Synthetic genetic analysis identified mutations that exhibit a full, temperature-independent Vma− growth defect when combined with the rav1∆ mutation. These include class E vps mutations, which compromise endosomal sorting. The synthetic Vma− growth defect could not be attributed to loss of vacuolar acidification in the double mutants, as there was no vacuolar acidification in the rav1∆ mutant. The yeast V-ATPase a subunit is present as two isoforms, Stv1p in Golgi and endosomes and Vph1p in vacuoles. Rav1p interacts directly with the N-terminal domain of Vph1p. STV1 overexpression suppressed the growth defects of both rav1∆ and rav1∆vph1∆, and allowed RAVE-independent assembly of active Stv1p-containing V-ATPases in vacuoles. Mutations causing synthetic genetic defects in combination with rav1∆ perturbed the normal localization of Stv1–green fluorescent protein. We propose that RAVE is necessary for assembly of Vph1-containing V-ATPase complexes but not Stv1-containing complexes. Synthetic Vma− phenotypes arise from defects in Vph1p-containing complexes caused by rav1∆, combined with defects in Stv1p-containing V-ATPases caused by the second mutation. Thus RAVE is the first isoform-specific V-ATPase assembly factor.
Collapse
Affiliation(s)
- Anne M Smardon
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210
| | | | | | | | | | | | | |
Collapse
|
48
|
Kobia F, Duchi S, Deflorian G, Vaccari T. Pharmacologic inhibition of vacuolar H+ ATPase reduces physiologic and oncogenic Notch signaling. Mol Oncol 2013; 8:207-20. [PMID: 24309677 PMCID: PMC5528540 DOI: 10.1016/j.molonc.2013.11.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 11/06/2013] [Accepted: 11/06/2013] [Indexed: 12/29/2022] Open
Abstract
Notch signaling in prominently involved in growth regulation in metazoan tissues. Because of this, Notch is often upregulated in cancer and current efforts point to developing drugs that block its activation. Notch receptor endocytosis towards acidic compartments is a recently appreciated determinant of signaling activation. Vacuolar H+ ATPase (V‐ATPase) is responsible for acidification of endocytic organelles and mutants in V‐ATPase subunit encoding genes in model organisms have been recently shown to display loss of Notch signaling. Here, we show that administration of BafilomycinA1 (BafA1), a highly specific V‐ATPase inhibitor decreases Notch signaling during Drosophila and Zebrafish development, and in human cells in culture. In normal breast cells, we find that BafA1 treatment leads to accumulation of Notch in the endo‐lysosomal system, and reduces its processing and signaling activity. In Notch‐addicted breast cancer cells, BafA1 treatment reduces growth in cells expressing membrane tethered forms of Notch, while sparing cells expressing cytoplasmic forms. In contrast, we find that V‐ATPase inhibition reduces growth of leukemia cells, without affecting Notch activatory cleavage. However, consistent with the emerging roles of V‐ATPase in controlling multiple signaling pathways, in these cells Akt activation is reduced, as it is also the case in BafA1‐treated breast cancer cells. Our data support V‐ATPase inhibition as a novel therapeutic approach to counteract tumor growth via signaling pathways regulated at the endo‐lysosomal level. V‐ATPase inhibition decreases Notch signaling during fly and fish development. V‐ATPase inhibition reduces Notch signaling in normal and breast cancer cells. V‐ATPase inhibition blocks degradation of membrane‐bound Notch forms. V‐ATPase inhibition prevents Notch cleavage and nuclear translocation. V‐ATPase inhibition reduces Akt signaling in breast cancer and T‐ALL cells.
Collapse
Affiliation(s)
- Francis Kobia
- IFOM, Istituto FIRC di Oncologia Molecolare at IFOM-IEO Campus, Via Adamello 16, 20139 Milano, Italy
| | - Serena Duchi
- IFOM, Istituto FIRC di Oncologia Molecolare at IFOM-IEO Campus, Via Adamello 16, 20139 Milano, Italy
| | - Gianluca Deflorian
- IFOM, Istituto FIRC di Oncologia Molecolare at IFOM-IEO Campus, Via Adamello 16, 20139 Milano, Italy
| | - Thomas Vaccari
- IFOM, Istituto FIRC di Oncologia Molecolare at IFOM-IEO Campus, Via Adamello 16, 20139 Milano, Italy.
| |
Collapse
|
49
|
Kasaian K, Wiseman SM, Thiessen N, Mungall KL, Corbett RD, Qian JQ, Nip KM, He A, Tse K, Chuah E, Varhol RJ, Pandoh P, McDonald H, Zeng T, Tam A, Schein J, Birol I, Mungall AJ, Moore RA, Zhao Y, Hirst M, Marra MA, Walker BA, Jones SJM. Complete genomic landscape of a recurring sporadic parathyroid carcinoma. J Pathol 2013; 230:249-60. [PMID: 23616356 DOI: 10.1002/path.4203] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 04/14/2013] [Accepted: 04/16/2013] [Indexed: 12/17/2022]
Abstract
Parathyroid carcinoma is a rare endocrine malignancy with an estimated incidence of less than 1 per million population. Excessive secretion of parathyroid hormone, extremely high serum calcium level, and the deleterious effects of hypercalcaemia are the clinical manifestations of the disease. Up to 60% of patients develop multiple disease recurrences and although long-term survival is possible with palliative surgery, permanent remission is rarely achieved. Molecular drivers of sporadic parathyroid carcinoma have remained largely unknown. Previous studies, mostly based on familial cases of the disease, suggested potential roles for the tumour suppressor MEN1 and proto-oncogene RET in benign parathyroid tumourigenesis, while the tumour suppressor HRPT2 and proto-oncogene CCND1 may also act as drivers in parathyroid cancer. Here, we report the complete genomic analysis of a sporadic and recurring parathyroid carcinoma. Mutational landscapes of the primary and recurrent tumour specimens were analysed using high-throughput sequencing technologies. Such molecular profiling allowed for identification of somatic mutations never previously identified in this malignancy. These included single nucleotide point mutations in well-characterized cancer genes such as mTOR, MLL2, CDKN2C, and PIK3CA. Comparison of acquired mutations in patient-matched primary and recurrent tumours revealed loss of PIK3CA activating mutation during the evolution of the tumour from the primary to the recurrence. Structural variations leading to gene fusions and regions of copy loss and gain were identified at a single-base resolution. Loss of the short arm of chromosome 1, along with somatic missense and truncating mutations in CDKN2C and THRAP3, respectively, provides new evidence for the potential role of these genes as tumour suppressors in parathyroid cancer. The key somatic mutations identified in this study can serve as novel diagnostic markers as well as therapeutic targets.
Collapse
Affiliation(s)
- Katayoon Kasaian
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Rabconnectin3α promotes stable activity of the H+ pump on synaptic vesicles in hair cells. J Neurosci 2012; 32:11144-56. [PMID: 22875945 DOI: 10.1523/jneurosci.1705-12.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Acidification of synaptic vesicles relies on the vacuolar-type ATPase (V-ATPase) and provides the electrochemical driving force for neurotransmitter exchange. The regulatory mechanisms that ensure assembly of the V-ATPase holoenzyme on synaptic vesicles are unknown. Rabconnectin3α (Rbc3α) is a potential candidate for regulation of V-ATPase activity because of its association with synaptic vesicles and its requirement for acidification of intracellular compartments. Here, we provide the first evidence for a role of Rbc3α in synaptic vesicle acidification and neurotransmission. In this study, we characterized mutant alleles of rbc3α isolated from a large-scale screen for zebrafish with auditory/vestibular defects. We show that Rbc3α is localized to basal regions of hair cells in which synaptic vesicles are present. To determine whether Rbc3α regulates V-ATPase activity, we examined the acidification of synaptic vesicles and localization of the V-ATPase in hair cells. In contrast to wild-type hair cells, we observed that synaptic vesicles had elevated pH, and a cytosolic subunit of the V-ATPase was no longer enriched in synaptic regions of mutant hair cells. As a consequence of defective acidification of synaptic vesicles, afferent neurons in rbc3α mutants had reduced firing rates and reduced accuracy of phase-locked action potentials in response to mechanical stimulation of hair cells. Collectively, our data suggest that Rbc3α modulates synaptic transmission in hair cells by promoting V-ATPase activity in synaptic vesicles.
Collapse
|